Skip to main content

REVIEW article

Front. Pain Res., 27 March 2024
Sec. Abdominal and Pelvic Pain
Volume 5 - 2024 | https://doi.org/10.3389/fpain.2024.1241015

Pain mechanisms in the transgender individual: a review

Jennifer T. Anger1* Laura K. Case2 Andrew P. Baranowski3 Ardin Berger4 Rebecca M. Craft5 Lyn Ann Damitz6 Rodney Gabriel7 Tracy Harrison8 Kirsten Kaptein9 Sanghee Lee1 Anne Z. Murphy10 Engy Said7 Stacey Abigail Smith11 David A. Thomas12 Maria del C. Valdés Hernández13 Victor Trasvina1 Ursula Wesselmann14 Tony L. Yaksh2*
  • 1Department of Urology, University of California San Diego, San Diego, CA, United States
  • 2Department of Anesthesiology, University of California San Diego, San Diego, CA, United States
  • 3Pelvic Pain Medicine and Neuromodulation, University College Hospital Foundation Trust, University College London, London, United Kingdom
  • 4Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States
  • 5Department of Psychology, Washington State University, Pullman, WA, United States
  • 6Division of Plastic and Reconstructive Surgery, University of North Carolina, Chapel Hill, NC, United States
  • 7Division of Regional Anesthesia, University of California San Diego, San Diego, CA, United States
  • 8Department of OB/GYN & Reproductive Sciences, University of California San Diego, San Diego, CA, United States
  • 9Division of Plastic Surgery, University of California San Diego, San Diego, CA, United States
  • 10Neuroscience Institute, Georgia State University, Atlanta, GA, United States
  • 11Division of Infection Disease, The Hope Clinic of Emory University, Atlanta, GA, United States
  • 12Office of Research on Women's Health, National Institutes of Health, Bethesda, MD, United States
  • 13Department of Neuroimaging Sciences, Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
  • 14Departments of Anesthesiology and Perioperative Medicine/Division of Pain Medicine, Neurology and Psychology, and Consortium for Neuroengineering and Brain-Computer Interfaces, The University of Alabama at Birmingham, Birmingham, AL, United States

Specific Aim: Provide an overview of the literature addressing major areas pertinent to pain in transgender persons and to identify areas of primary relevance for future research.

Methods: A team of scholars that have previously published on different areas of related research met periodically though zoom conferencing between April 2021 and February 2023 to discuss relevant literature with the goal of providing an overview on the incidence, phenotype, and mechanisms of pain in transgender patients. Review sections were written after gathering information from systematic literature searches of published or publicly available electronic literature to be compiled for publication as part of a topical series on gender and pain in the Frontiers in Pain Research.

Results: While transgender individuals represent a significant and increasingly visible component of the population, many researchers and clinicians are not well informed about the diversity in gender identity, physiology, hormonal status, and gender-affirming medical procedures utilized by transgender and other gender diverse patients. Transgender and cisgender people present with many of the same medical concerns, but research and treatment of these medical needs must reflect an appreciation of how differences in sex, gender, gender-affirming medical procedures, and minoritized status impact pain.

Conclusions: While significant advances have occurred in our appreciation of pain, the review indicates the need to support more targeted research on treatment and prevention of pain in transgender individuals. This is particularly relevant both for gender-affirming medical interventions and related medical care. Of particular importance is the need for large long-term follow-up studies to ascertain best practices for such procedures. A multi-disciplinary approach with personalized interventions is of particular importance to move forward.

Background

The intersection of sex and pain has become a focus of extensive research at the clinical and preclinical level. Numerous sex differences exist in pain and its processing in humans (1) However, pain research frequently conflates sex and gender. Sex refers to biological aspects of maleness or femaleness. It is a complex label used to describe the totality of distinguishable characteristics of an organism, including its reproductive structure, functions, phenotype, and genotype (2, 3). On the other hand, gender refers to the psychological, behavioral, social, and cultural aspects of being male, female, or another gender identity. Expectations of gender differ by culture. However, gender is frequently assumed on the basis of sex assigned at birth; a difference in gender identity from that assumed/assigned at birth is broadly referred to as being transgender (1). The pain experience of transgender individuals has been poorly studied.

There are several reasons why pain experience may differ in individuals who are transgender (trans). One reason is the stress that many trans individuals experience due to a frequently hostile social and political environment (4, 5). Many trans individuals also experience gender dysphoria, defined as the distress and emotional discomfort from their gender incongruence, which is created and/or exacerbated by a societal environment that does not consistently accept gender diversity (6, 7). Social marginalization is a frequent risk factor for suicidality (4). Indeed, a large study of transgender adults in the United States, 40% endorsed a lifetime suicide attempt (8). The compounding effects of distress generated by gender dysphoria is also reflected in the high incidence of suicide and disabling behavioral syndromes in transgender individuals in whom the dysphoria is not addressed (4, 9, 10). As will be reviewed below, it is widely appreciated in many human conditions that such unmitigated stressors can lead to profound exacerbations of pain phenotypes (11, 12) and this may have a strong sex/gender component (13, 14).

Other differences in biology relevant to pain perception may arise from gender-affirming clinical interventions. These interventions may involve hormonal or surgical therapies to assist in modifying a person's primary and secondary sexual characteristics. Such interventions can significantly impact the psychological and physiological components of the pain experience (15, 16). These variables suggest the complexity of pain expression in the increasingly visible and diverse population of transgender adults and youth, along with the appreciation that transgender patients represent a medically underserved, stigmatized population (6). This has led the Institute of Medicine to identify transgender health as a research priority (17). However, the landmark report of the Institute of Medicine on pain published in the same year, did not address pain in transgender patients (18), and there is an urgent medical need to consider this important and understudied topic.

Overarching aims of the review

The aim of this review is to summarize key literature, addressing variables related to pain in transgender individuals. We review the background and note issues related to categorization of gender identity. Specifically, we: (i) highlight the impact of discrimination, life experiences, and hormonal and surgical treatments on pain perception; (ii) consider aspects of the biology of neural processing of pain and potential differences in cis and trans individuals; (iii) consider the contribution of preclinical studies to understanding of the nexus of sex, gender, and biology. We also address the role of the NIH in supporting issues pertinent to pain in transgender individuals, and directions for future research. The breadth of the topic precluded an in-depth review of each of the topical areas, but it is our aim to address pivotal issues for which we feel information or answers are not currently available and as such, point to future directions of research. As a prelude to this discussion, it is important to appreciate the complex interplay between medical issues and associated pain issues that pertain to transgender patients. A comprehensive listing of relevant concepts is presented in the Appendix (Supplementary Table S1) of this paper.

Search strategies

A group of twenty-one participants formed the core of the ad hoc study group (Pain in Transgender Patients). This group met periodically though zoom conferencing between August 2021 and February 2023. This group created a series of specific topics that they considered to reflect essential issues relevant to the question of pain in transgender patients. These topical areas were assigned to individuals who have expertise in the respective areas. Each created a series of topical paragraphs that drew from the published and publicly available literature. These paragraphs were compiled and editorially revised. Compiled drafts were redistributed to the core group for revisions and corrections. The final drafts were approved and submitted.

Transgender patient populations

Population estimates

As noted, gender-minoritized persons are an often marginalized populations in the health system (19). Current electronic health records in many health organizations are not positioned or equipped to store and share gender-related information appropriately (20). Further estimates of the size of the transgender population are complicated, as gender diverse individuals do not represent a homogeneous population. Broadly speaking, the World Professional Association for Transgender Health (WPATH) Standards of Care defines gender nonconformity or incongruence as “the extent to which a person's gender identity, role, or expression differs from the cultural norms prescribed for people of a particular sex” (21). Gender incongruence was previously coded in the International Statistical Classification of Diseases and Related Health Problems of the World Health Organization (WHO) as a ‘mental disorder’. In the new International Classification of Diseases and Related Health Problems (ICD-11), gender incongruence is coded under ’sexual health’ (22, 23). This replaces previous diagnostic categories such as “transsexualism” and “gender identity disorder of children” with “gender incongruence of adolescence and adulthood” and “gender incongruence of childhood”.

A global survey in 2021 noted that 2% of respondents from 27 countries identified as transgender, non-binary/non-conforming, or gender fluid (24). The Behavioral Risk Factor Surveillance System (BRFSS) telephone survey in the US estimates that adult transgender population range from 0.3% to 0.8% of the population across several states (25, 26), whereas a review of population-based surveys (2006–2016) found that 0.39% of respondents identified as transgender when this was an option (27). Transgender self-identification appears more common among younger age groups (28). This may relate to decreases on stigma and increased availability of services for trans individuals, as well as changes in categories of gender.

Issues related to data collection in relation to gender identity

The numbers quoted above, often used to estimate population characteristics, have been challenged as underestimates for several reasons (29). First, transgender identity is not often ascertained in many federal census or national health surveys or is too narrowly defined. Lett and Everhart (30) have introduced the use of other sources to address transgender identity. These include the Youth Risk Behavior Surveillance System (31), the National Crime Victimization Survey (29), the General Social Survey (32), and the National HIV Behavioral Surveillance System (33). Second, determining gender identity through survey questions depends upon self-identification. Responders may not identify themselves as transgender on surveys, despite expressing a gender that is different than their gender assigned at birth (30). Third, a simple question and a one-step query methodology to determine transgender identity may misclassify transgender people (30).

The absence of inclusive concepts, terms, and codes for gender, sex, and sexual orientation has contributed to the inability to correctly identify sex and gender minorities in data sets and electronic health records. As a result, we can express only modest certainty about and the prevalence of transgender identity and more specifically about specific health needs and long-term health outcomes among transgender individuals (34). Practically, the prevalence of transgender identity has been defined by physical alterations as with medical or surgical transitions (3537). However, as will be reviewed below, while useful and important measures, the complexity of gender identity, the effects of hormonal therapies and the impact of the societal milieu contributing to gender dysphoria, can complicate the linkage of these physical characteristics with gender assignment, and there is often little discussion considering general implementation of such systems alongside self-identification (38, 39).

An important element in advancing our understanding of the conditions and issues faced by transgender populations is the development of gender-appropriate data collection tools and healthcare records. It must be recognized that approaches employing International Classification of Diseases (ICD) codes or simple text linked to trans-related procedures prioritizes populations of individuals who exhibit higher utilization of gender-affirming therapies/surgeries, often with better insurance coverage and lower poverty rates (2). For these reasons, it has been argued that the gold standard for gender assignment in patients is self-identification (21).

Pain in relation to gender identity

Differences in pain sensitivity/responsivity and incidence in cisgender (cis) men and women groups are well documented, wherein it is reported that women often display greater sensitivity than men (5, 40). Such differences are frequently noted in specific syndromes, such as migraine (41) musculoskeletal pain (41, 42), temporomandibular joint syndromes (43, 44), fibromyalgia (45, 46); rheumatoid, osteo- and psoriatic arthritis (4749); and Sjögren's disease (50). Some of these pain syndromes, such as migraine headaches, are more frequent in cis women, and often overlap with other pain conditions (51). Other pain syndromes, such as cluster headache, as more frequent in cis men (52). Furthermore, several studies have been conducted to highlight how gender biases in pharmacological and non-pharmacological pain management differ in cismen and ciswomen. Although significant differences were found, the data often depended on both the treatment type and pain characteristics. However, the etiology of these differences is complex given differential treatment of men and women in pain (5).

While there are differences in pain between cis men and women, potential differences in trans individuals are much less well characterized, and systematic work is scant. Survey-based studies have suggested a higher prevalence of pain symptoms among transgender people compared to the general population (53), a difference which appears to be more pronounced for trans men as compared to cis men than between trans women and cis women (5355). Aloisi and colleagues studied transgender women and men who had undergone hormonal treatment for at least one year, and retrospectively report whether pain had changed from before to after treatment. Approximately 30% of transgender women reported painful conditions, and 80% of those stated that these painful conditions started after the onset of hormone treatment (55). In contrast, approximately 60% of transgender men reported pain, but nearly half of the participants reported that pain lessened after the onset of hormone treatment (55). However, these studies included very small sample sizes, and further study with much larger samples is needed to confirm these potential differences

In a study involving 51 African-American cis and transgender individuals living with HIV and chronic pain, Strath and colleagues (56) reported that trans women and cis women demonstrated a greater temporal summation for heat pain stimuli or stimulation compared to cisgender men. Trans women showed greater mechanical summation of the sensation produced by repeated mechanical nociceptive stimulation than either cis women or men. Similarly, trans and cis women reported greater chronic pain severity as compared to cis men. Migraine prevalence in trans women taking gender affirming hormone therapy was similar to that of cis women in population estimates (57).

Many trans individuals undergo painful clinical interventions in gender affirmation protocols, including hormone replacement and surgeries (5860). However, the impact of these interventions on pain may differ by gender. A recent systematic review examining the literature on gender minority health provides little information specifically related to pain (61). Pain has been indicated as one of the monitoring parameters for feminizing hormone therapy (62, 63). A study of 47 transgender women and 26 transgender men reported that up to 25% of trans women developed chronic pain with estrogen and/or anti-androgen therapy whereas 60% of trans men experienced significant improvement in chronic headache along with testosterone treatment (55). Trans women treated with estrogen display a prevalence of migraine similar to that of cis women, including aura (57, 64, 65). In contrast, a study that recruited 115 transgender individuals from a specialized clinic in Israel reported a high prevalence of fibromyalgia symptoms among transgender individuals in general and in transgender men in particular as compared to transgender women (53). A variety of rheumatic pathologies are known to have a strong immune mediated component, and sexual dimorphism occurs in innate and adaptive immunity and contributes to differences in the prevalence of rheumatic diseases (66). A recent review commented on the limited literature on arthritis among trans individuals but noted that, though the number of cases was only 14, eleven cases of such immune mediated disorders occur in trans women (67). In parallel with the functional significance of immune cells on pain mechanisms, there is a greater predisposition to chronic pain among individuals lacking a Y chromosome (68, 69). However, gender identity in transgender patients may play as important a role in the pain experience as the genetics associated with sex (56).

Medical care for transgender pain patients

A prospective survey found that 90% of pediatric anesthesiologists (n = 374) were comfortable interacting with transgender patients but only 50% of the respondents felt equipped with the knowledge to manage the surgical needs of transgender patients (70). This and other studies (7072) demonstrate a fundamental need to understand pain mechanisms in the pediatric transgender population and to provide specific training for clinical care teams including anesthesiologists. In this regard, Standards of Care are issued and updated by WPATH (World Professional Association for Transgender Health), providing health care guidelines for clinical outcomes in target populations including transgender individuals (73). Implementation of questions about pain perception and its burden in transgender populations would provide detailed information that includes, but is not limited to, age, medical conditions, hormonal or surgical status, all of which may enhance our understanding of pain in relation to gender. Furthermore, the shifting paradigm in transgender and gender-diverse populations needs to be embedded in health care (2). Approaches to pain management should consider the significant pharmacological and surgical interventions that many transgender individuals receive. In the following section we provide an overview of these interventions and their implications.

Gender affirming surgical interventions

Transgender patients may undergo a range of gender-affirming surgical interventions (74). Procedures often thought of as feminizing may include breast augmentation, facial feminization (7578), and gender-affirming pelvic surgery (GAPS), or “bottom” surgery, which usually includes a penectomy (removing the penis), bilateral orchiectomy (removing the testicles), vaginoplasty (creating a vagina) (49, 79), clitoroplasty (creating a clitoris from the penile glans), and labiaplasty (creating labia minora and majora). Shallow depth vaginoplasty, or vulvoplasty, is an alternative to full-depth vaginoplasty for women who do not wish to engage in penetrating vaginal intercourse (80).

Procedures often thought of as masculinizing may include mastectomy (chest or “top” surgery), masculinizing facial surgery, and masculinizing “bottom” surgery which may involve a hysterectomy with or without salpingo-oophorectomy (removal of fallopian tubes and ovaries), vaginectomy (removing the vaginal canal), phalloplasty (creation of a phallus using a pedicle-based flap), metoidioplasty (creating a smaller penis from the clitoris after it is elongated with testosterone therapy), and/or scrotoplasty (creation of a scrotum from labia majora).

Many trans individuals also undergo vocal cord surgeries including thyroplasty for transgender men, or glottoplasty for transgender women (78, 8183). These interventions are rarely performed for transgender people under the age of 18 (84, 85) and are never performed under the age of 16, despite prevalent misconception.

Gender-affirming medical and surgical treatments can increase positive body image in trans individuals by aligning bodily appearance with gender and desired sex, as a study on transgender individuals in Zurich found (86). Gender affirming hormone therapies have also led to an increase in body image and self-esteem (87). Similarly, hair removal procedures (desired by nearly 90% of transgender people) were associated with significant mental health benefits (88, 89).

In a comprehensive literature review, several insights were provided regarding gender affirming surgery (GAS) and trends in its use: (i) GAS is more common in transgender men than in transgender women, and least common in gender non-binary or nonconforming populations; (ii) Genital procedures are less common than chest surgery, with prevalence rates for genital procedures being about 25%–50% for transgender men and 5%–10% for transgender women (28, 90, 91). Gender-affirming mastectomy, or “top” surgery, is a common procedure for trans men. The demand for top surgery by transmen has increased 15% since 2019, with 8,548 procedures performed in 2020 (92). The incidence of GAS utilization among transgender people is mainly influenced by age, income, and race, with the primary variable for GAS utilization being income (90).

It should be noted that, while gender affirming surgeries are commonly associated with transgender patients, many of these procedures are similarly implemented in cis patients. For instance, a cisgender woman undergoing a breast augmentation may do so for gender affirming purposes, and cis men who have had genital damage may seek cosmetic surgical changes as a form of gender affirmation (93, 94). Individuals born with genital differences may also undergo gender affirming procedures (95).

Future directions

A change in policy

Although gender-affirming medical (hormonal) therapy and surgical treatments are associated with a significant improvement in gender dysphoria (91, 9698), there remains a paucity of data measuring outcomes across the many treatment domains, including complications and physical and mental health outcomes. Section 1557 of the Affordable Care Act became effective July 18, 2016, prohibiting discrimination in healthcare on the basis of a number of protected characteristics, including sex. The new regulations codified existing informal guidance from the Department of Health and Human Services, interpreting this provision to include protections from discrimination based on gender identity and sex stereotyping. Section 1557 regulations explicitly prohibit discrimination based on gender identity—making it clear that most insurers cannot deny or limit coverage because a treatment is related to an individual's gender transition. This change in codification, which increased access to medical and surgical therapy relevant to transgender individuals, resulted in a rise in the number of gender affirming surgeries performed in the US (90), as well as the number of gender health programs in this country.

Pain phenotypes in patients undergoing gender affirmation surgeries

Despite the increasing incidence of gender affirming surgery (GAS), few studies have analyzed the prevalence and severity of post-operative pain immediately following GAS. Longer term reviews are sparse and yield conflicting results. To date, there are few reviews summarizing quality-of-life outcomes following gender-affirming surgeries over periods of greater than 1 year (99101). Of the data that does exist, two long term follow up studies, found that gender affirming surgery alleviates gender dysphoria by improving quality of life, and psychological symptoms but also that there are still high psychiatric morbidity and suicide rates after gender reassignment surgery compared to the general population (98, 102). Several informative reviews focus on specific types of surgery, including phalloplasty (103, 104), vaginoplasty (105, 106), and laryngoplasty (107, 108). Furthermore, a recent review examined outcomes of chest, facial, and vocal cord surgeries (101). The consensus across reviews is that surgery improves transgender individuals’ quality of life. These reviews, while informative, do not explicitly consider longer term (i.e., > 1 year) quality-of-life outcomes following gender affirming surgery, or compare outcomes for each type of gender affirming surgery in transgender men and women. Patients tend to report positive transitioning outcomes within the first year after surgery (101, 109, 110). In a longer-term evaluation of transgender women's health and well-being after feminizing surgery, Lindqvist (111) found that transgender women reported increased health-related quality of life after feminizing surgery, but health-related quality of life declined in the following years. Hence, early evaluations (i.e., < 1 year), while informative, need to be supplemented with information about longer term functioning.

There is limited research on persistent postsurgical pain following mastectomy for gender dysphoria, even though it is well documented and common after mastectomy for breast cancer (112). In oncologic mastectomies, the likelihood of developing chronic pain is commonly appreciated and is often associated with an enduring neuropathic phenotype (113, 114). When breast tissue is surgically removed, sensory nerves traveling through these tissues are transected, stretched, or are caught up in scar during the healing process. These nerve injuries can lead to chronic pain due to the development of neuromas and/or scar contraction. Altered sensation, including “pins and needles”, shock-like, burning, or stabbing pain provide evidence of nerve injury as a cause for the pain. Response to local anesthetic nerve blocks can confirm that the source of the chronic pain is the injured nerves. The degree and frequency of persistent pain has been linked to the intensity of acute postoperative pain (115), pre-existing chronic pain conditions (116), as well as psychological factors such as anxiety and depression (117). Using acute postoperative pain scales and opioid consumption, Verdecchia and colleagues (118) found that the transgender top surgery patients consumed fewer opioids and reported lower pain scores than cisgender breast reduction patients. This may be explained by the effects of prior ongoing gender affirming hormone therapy (GAHT) on pain scores but more research is needed to confirm this (118). Similarly, Robinson et al. (119) prospectively compared top surgery to oncologic mastectomy without reconstruction and to mammoplasty breast reduction. They found that patients who underwent oncologic mastectomy consumed similar quantities of opioids to top surgery patients. In both studies, patients received pectoralis nerve blocks. A retrospective chart review (120) evaluated complication rates following top surgery, and surveyed patients about their experience with persistent postoperative pain. More than 25% of top surgery patients reported chronic pain, but 95% (n = 77) of these patients did not require analgesics. The potential role of concurrent hormone therapies in these studies was unfortunately not indicated. Generally, all participants were overprescribed opioids in the study. This may affect patients with a higher potential for substance abuse, which may be the reason these results conflict with Verdecchia and colleagues’ findings. The limited and conflicting data describing the prevalence and severity of pain following top surgery makes it challenging for physicians to counsel their patients on their risk of developing postoperative pain, as well as to predict opioid prescription needs.

Robust outcomes data after vaginoplasty and the prevalence of long-term gynecologic issues is lacking (121, 122). A past study that examined the prevalence of patient-reported symptoms and adverse reactions in 80 patients undergoing vaginoplasty found that 54% sought care within the first year after the procedure. The most common adverse outcomes included impaired wound healing / hypergranulation (13% / 39%), urinary dysfunction (19%). Sexual dysfunction issues were noted (34%) with anorgasmia (11%) and dyspareunia (11%) being most frequent (123). In a systematic review and meta-analysis that examined the outcome and complications of 3388 transgender females after penile inversion vaginoplasty, the mean prevalence of reported urinary complications ranged from 5.0%–11.9% with the most common symptoms being splayed stream (11.7%), meatal stenosis (6.9%), and irritative symptoms (frequency, urgency, nocturia) (11.5%) (124). In trans men, phalloplasty and metoidioplasty are the two most common genital surgeries (81, 125). Metoidioplasty is reported to have a complication rate of 10%–37% with postvoid dribble and/or a spraying stream (33%) and urethrocutaneous fistulas (5%–23%) being the most common (81). Phalloplasty may employ a free flap procedure, often with the radial forearm providing the donor site and the target tissue innervation. Most post operative reports on bottom surgery focus on the recovery of sensory function in the genitalia whereas with breast surgery a neuropathic phenotype has been readily reported. Such neuropathic phenotype reports are virtually nonexistent following vaginoplasty and penile construction. procedures. Carlotta et al. concluded that recovery of bottom sensation post-surgery suggests a greater sensory reinnervation, both in magnitude and temporally of the genitalia than that observed in the regeneration of limb afferents (126).

Hormone therapy can influence pain in transgender patients, with testosterone therapy improving chronic pain in some transgender men, for example (55). Furthermore, mood disorders such as anxiety can help to predict persistent post-mastectomy pain syndrome in women after oncologic mastectomy (127), yet similar work has not been described in transgender individuals. Local nerve blocks could be beneficial for pain management and deserve further systematic study, as they have been demonstrated to improve pain outcomes following oncologic mastectomy (128, 129).

Future directions

Current insights into the characteristics of the pain phenotype occurring following gender affirmation surgical procedures suffer from: (i) limited attention to definitive descriptions and characterizations of the pain being reported by the individual, (ii) absence of long term follow up and appropriate and adequate characterization of the techniques employed, and (iii) influence of patients’ hormonal status on pain. Slow recognition of the pain state following breast intervention in cis women makes it unlikely that pain states in trans people will be evident unless they are specifically characterized.

There appears to be a relative lack of persistence of pain and ongoing pain phenotypes after gender affirming bottom surgery. This stands in contrast to the time course of postoperative pain for other surgeries of skin and bone, where significant pain may persist and develop into a neuropathic phenotype. The rapid reduction of pain that occurs in the bottom surgery patient may be intertwined with relief of gender dysphoria and merits further research (126).

Further, additional work characterizing anatomy and neuroplasticity is essential for understanding the nature of pain in transgender individuals. Such understanding is not only important for clinical management and determination of prognosis but is essential as a part of supporting the patient by giving an explanation for their symptoms, both pain and otherwise. Finally, it should be clear that predictions regarding the pain state in transgender patients that are based solely on those of the cis individual (as in mastectomy and breast augmentation, or genital reconstruction/bottom surgery) do not include the potential impact of transgender hormonal therapeutics or the mental health impact of gender dysphoria, societal marginalization, and social gender transition. Finally, as noted in the following section, hormonal therapies are an important component of the treatment regimens of the transgender individual and the effects of such treatment on the post operative pain state has been poorly considered, and future work in this would importantly consider this interaction.

Hormone replacement therapy (HRT) in adults

Hormone replacement therapy is a cornerstone of gender affirming medical care to address gender dysphoria (62, 73, 130). The National Transgender Discrimination Survey on Health and Health Care reports gender-affirming hormone therapy (GAHT) in up to 80% of transgender people. Other estimates suggest that approximately 30%–68% of transgender persons in the US utilize hormone therapy in pursual of congruent secondary sex characteristics (36, 131133). In gender-diverse people assigned female at birth, GAHT primarily involves administration of exogenous testosterone esters (130, 134) for masculinization. Many transgender men use testosterone therapy to suppress estrogen (134). In transgender women, hormone therapy includes use of a synthetic estrogen, usually with anti-androgens to suppress testosterone (135), and occasionally progesterone, though the risk/benefit ratio of progesterone use in trans women is understudied (136). It is important to note that the trans population is heterogeneous in therapeutic hormone use, and includes different formulations dosages, and routes of administration (oral, injection, patches). For example, while the clinical goal for some trans women may be to maintain serum concentrations of estradiol similar to those of cis women (100–200 pg/ml) (130), other patients may seek the phenotypic and physiologic effects of lower concentrations. Alternatively, trans people might have only sporadic access to prescribed therapy or may obtain and utilize hormone therapy outside of medical direction.

Hormone therapeutics and pain in transgender individuals

Evidence describing the effect of hormone replacement therapy on the experience of pain in transgender populations is inconsistent and is often limited by small sample size and subject to sampling bias. One exploratory study of trans men in Thailand found that those undergoing testosterone therapy reported higher bodily pain scores on the Short Form Health Survey 36 compared with those not utilizing testosterone (137). Conversely, a questionnaire-based study of 26 trans men found that 60% reported a significant improvement in chronic headache symptoms that had been present prior to the start of testosterone treatment (55). Meanwhile, a study of Israeli trans men did not find a significant difference between the prevalence of fibromyalgia symptoms before and after the initiation of hormonal therapy (53). A U.S. survey-based study of 100 trans men who had undergone phalloplasty and who were taking testosterone found that 51% (48/94) reported pain with sexual penetration. Pain was present before initiation of testosterone in 41.7% (20 of 48), and no significant correlation was found between pain scores and the duration of time since initiating testosterone (60). In another survey-based study of 183 trans men endorsing abdominopelvic pain, 69.4% (127/183) reported new-onset pain following initiation of testosterone GAHT, with a median time from testosterone initiation to pain onset being 1 year (138). In summary, insufficient data exist to definitively discern the specific effects of testosterone GAHT on pain in trans men.

In contrast to the small number and heterogeneity of findings observed in studies describing pain in trans male individuals, the data describing trends in pain among trans women are more consistent. Several studies have suggested that trans women experience more pain than cis men and cis women, a phenomenon that appears to be exacerbated by anti-androgen therapy and an estrogen GAHT (54, 56, 138140). In a study of 47 trans women, chronic pain was observed in 29.8%, with breast pain and headache onset occurring after initiation of estradiol and/or anti-androgen therapy in 11 of 14 affected individuals (55). Further, among trans women taking anti-androgens and high-dose estrogens, the prevalence of migraine has been suggested to increase, particularly among those taking high doses of oral estrogens (55). A positive correlation has also been demonstrated between trans women's time on GAHT and their experience of bodily pain (54). Overall, the prevalence and severity of pain in transgender women is associated with impaired mental health outcomes, with Female Sexual Function Index Pain scores (e.g., pain associated with sexual activity) accounting for a significant proportion of the Beck Depression Inventory depression score, after controlling for age and general mental health levels (139).

Research investigating pain in transgender children and adolescents is very limited. While injection site pain is a well-known side effect of any injectable medication, it is unclear whether gonadotropin releasing hormone (GnRH) agonists that some gender-diverse youth use for pubertal suppression are associated with altered pain experience. During initial cyclical administration of GnRH agonist therapy, the pituitary is stimulated to produce gonadotropic hormones which increase downstream gonadal steroid secretion. Infusions of GnRH agonists produce an initial transient increase in sex hormones, but with continued non-pulsatile stimulation, LH and FSH synthesis are inhibited and estrogen and testosterone levels decline (141). Of note, GnRH agonists have been used as a last line of pain suppression in adolescent cis girls suffering from dysmenorrhea, endometriosis, and chronic pelvic pain. However, their study did not include gender diverse patients (142144).

Hormone replacement and pain in adults

Here we briefly review the two primary classes of agents used in GAHT and their effects on pain processing. We first examine the current literature on the effects of the gonadal steroid hormones on pain in animals and cis gendered men and women to hypothesize their effects on pain in transgender individuals.

Testosterone & pain

Regarding androgenic influence on pain, one study collected pain scores of 127 cis gender men and 145 cis gender women. The study reported that testosterone levels were negatively correlated with pain after knee replacement at the operated knee, e.g., high testosterone was correlated with lower pain scores, in both cisgender men and women osteoarthritis patients (145). Similarly, androgen level has been reported in limited reports to be negatively correlated with pain in a study of 40 cycling women tested with noxious stimuli in the lab (146), as well as negatively correlated with days/month of pelvic pain, menstrual pain, and headache in 56 cycling women (147). In laboratory rodents, exogenous testosterone administered to adults may increase pain thresholds (i.e., decrease or delay response to noxious stimuli) in gonadectomized males and females [e.g., (148)], although testosterone administered to gonadally intact rodents may not alter pain in either sex [e.g., (149)]. Because testosterone (or estradiol) administration to gonadally intact animals triggers negative feedback to the hypothalamic-pituitary-gonadal axis (thereby decreasing endogenous gonadal hormone release) (150), exogenous hormone effects may differ in gonadally intact vs. gonadectomized individuals. When androgens do reduce pain, the reduction coincides with its attenuating effects on inflammation (151). However, it has been shown that pelvic pain in transgender men often occurs after initiation of testosterone therapy (152). Given the known androgen sensitivity of the pelvic floor musculature (153), further research into pelvic floor muscle dysfunction is warranted.

Estrogens & Pain

Menstrual cycle phase may alter pain in cisgender women with migraine, fibromyalgia, and temporomandibular pain; typically, pain worsens during periods of rapid estrogenic decline, such as during the late luteal phase (154). However, there is disagreement among studies examining the interactions between ovarian hormones and pain (155). In women without chronic pain, somatosensory thresholds are not systematically affected by cyclic changes in ovarian hormones (154, 156). In laboratory rodents, estradiol may increase pain-related behaviors in both sexes (157), but estradiol has also been observed to decrease pain-related behaviors (e.g., in gonadally intact females using a visceral pain model (149). It has been suggested that estrogenic influence on pain depends on the extent to which a given pain disorder involves immune, skeletal, cardiovascular (and other) systems, because estrogens can influence each of these systems differently (158). Estrogens have diverse effects on peripheral organs and tissues, and in the brain as mediated by estrogen receptor alpha (ERalpha, playing a critical role in reproductive neuroendocrine function and behavior) and estrogen receptor beta (ERbeta, present in populations of hypothalamic GnRH, corticotropin releasing hormone, vasopressin, oxytocin, and prolactin expressing neurons) (159).

Estrogenic modulation of pain may depend on the type of chronic pain (or pain model in animal studies), whether the estrogen-treated individual is gonadectomized, and, among other variables, whether estrogens are combined with progestins (157, 159).

Other GAHT therapeutics

To support gender transition, anti-androgenic treatments are sometimes used; these include 5α-reductase inhibitors, progestins, and the mineralocorticoid receptor/androgen receptor antagonist spironolactone which functions as a testosterone blocker (160). Little is known about the impact of these drugs alone or in combination with estrogens—which is how they would typically be taken for gender transition—on chronic pain. Spironolactone in isolation has been reported to decrease inflammatory pain-related behavior in male mice (161) and neuropathic pain-related behavior in rats (162), but it did not alter fibromyalgia pain in women (163), or pain in human osteoarthritis patients of either sex (164). Research is needed to determine whether combined estrogens + anti-androgens are likely to affect chronic pain in preoperative vs. postoperative trans individuals.

Future directions

It should be stressed that there is a relative paucity of data substantiating the relationships between hormones and pain expression in transgender and cisgender individuals. Additional work in this area promises insight into effective pain management for people, especially those receiving exogenous hormones.

Mechanisms driving gonadal hormone effects on pain

Aside from their effects on inflammatory cascades, steroid hormones appear to modulate cortical processing of pain-related stimuli (165167), and preclinical studies using rodents have shown differential excitability of CNS targets in male and female organisms by the same endogenous steroid hormones (168). Although steroid hormone action predominantly involves nuclear signaling and regulation of gene expression, gonadal hormones have also been shown to exert direct influence on the peripheral and central nervous systems (158, 168173). Mechanisms potentially underlying these phenomena include pronociceptive interactions between the G-protein coupled estrogen receptor, the ionotropic 5HT3A serotonin receptor, NMDA receptors, and glutaminergic signaling in the CNS (174176). Estrogens may also negatively regulate inhibitory neurotransmission through glycine and GABA downregulation (177179). Importantly, estradiol may also negatively regulate the antinociceptive endocannabinoid and opioid systems (180182). Together, these mechanistic data suggest a pronociceptive effect of estrogens that may contribute to the higher levels and rates of pain in cisgender and transgender women observed in cohort and population-based studies. However, it should be noted that these studies have been conducted in cisgender individuals, and their findings may not be generalizable to transgender and gender-diverse individuals.

Hormone modulation of analgesic drug action

Gonadal hormones can influence the potency/efficacy of some analgesics and anesthetics. The extent of this influence among transgender individuals who take exogenous hormones may also depend on whether they stop hormones before surgery. In fact, the practice of stopping hormones is losing popularity as the literature grows with evidence that hormone cessation is not necessary. For example, in male and female rats, testosterone can increase morphine's antinociceptive potency on some but not all tests of acute pain (183). Estradiol also modulates morphine antinociception in gonadectomized female rats, but the direction of effect depends on estradiol dose, site of administration (peripheral or central), and timing (184); these shifts are paralleled by modest fluctuations in morphine potency across the estrous cycle (185) but see (186).

Finally, the testosterone blocker spironolactone, commonly used in the U.S., has been found to increase the antinociceptive effects of morphine and oxycodone in male rats, and this interaction appears to be pharmacokinetic (187, 188). Centrally, the antinociceptive effects of morphine are mediated, in part, via binding to the mu opioid receptor (MOR) within the midbrain periaqueductal gray (PAG). Both androgen (AR) and estrogen (ERα) receptors are located within the PAG, making this a likely anatomical substrate whereby gonadal steroids influence pain and analgesia (189). Several mechanisms have been proposed to account for gonadal hormone modulation of opioid analgesia. First, estrogens have been shown to attenuate MOR signaling by uncoupling MOR from its K + channel, thereby reducing morphine-induced hyperpolarization (179, 190), and by inducing MOR internalization resulting in reduced receptor availability (191). Estradiol has also been shown to facilitate the formation of MOR/KOR heterodimers in the spinal cord, thereby augmenting morphine's effects in females (192). Neuroimmune factors have also been implicated. In rodents, low doses of estradiol, comparable to normal circulating levels, increase proinflammatory cytokine production (193), which is known to reduce morphine antinociception in a TLR4-dependent manner (194196). Opioids are metabolized via glucuronidation to produce two metabolites: morphine-6-glucuronide (M6G), which has a high affinity for MOR and is considered pro-analgesic, and morphine-3-glucuronide (M3G), which has high affinity for TLR4 and opposes morphine action. In rodents, M3G levels are 2-fold higher in both plasma and PAG following systemic morphine (197), which is likely why direct PAG administration of the MOR-selective metabolite M6G results in a greater analgesic response than morphine alone in females (195). Opioid and barbiturate pharmacokinetics may be altered by both androgens and estrogens as shown in rat models [e.g., (198201)]. Overall, a multitude of pharmacokinetic and pharmacodynamic mechanisms underlie sex chromosome and gonadal hormone modulation of analgesia and anesthesia, some of which may be relevant to transgender individuals who have chronic pain or undergo surgery.

Future directions

The preclinical model does not presume to mirror the complexity of the estradiol and testosterone modulation of pain in transgendered populations. However, the preclinical models can offer direction for future studies to be observed in transgender populations. Although there is a growing literature, at least in laboratory animals, examining estradiol modulation of pain in rodents identified as female at birth and testosterone modulation of pain in rodents assigned male, there are few preclinical studies on hormone reversal using gonadectomized rodents and almost none in gonadally intact rodents. Both types of studies, but particularly the latter, are needed to model the biological effects of hormone treatment alone, or hormone treatment with orchiectomy or oophorectomy, in transgender patients. Additionally, in trans women, the use of estrogenic treatment is often accompanied by an anti-androgenic treatment (i.e., testosterone blockers), but very little is known about the combination of estrogens plus commonly used demasculinizing treatments on chronic pain in either clinical or preclinical studies. Studies manipulating gonadal hormones are relatively straightforward to conduct in laboratory rodents. Combining these manipulations with a variety of chronic pain models that have predictive validity (35) will greatly improve our understanding of how interventions common to the human gender transition process may affect pain associated with various pathologies. Additionally, gonadal hormone manipulations in rodents that mirror those used for human gender transitions could be used to further our understanding of how gonadal hormones are likely to affect the potency and efficacy of analgesics and anesthetics commonly used to treat pain clinically, including in transgender individuals.

A second potentially important issue relates to age or developmental stage of hormone exposure (as discussed above in puberty suppressive interventions). We do not know whether age influences the impact of hormones on pain and analgesia. This question could be readily addressed via laboratory rodent studies that examine the impact of hormone manipulation (via exogenous hormone treatment with and without gonadectomy) at pre-pubertal vs. post-pubertal ages (and for the latter, from adolescent to older adult) on pain and analgesic potency/efficacy. Nearly all pain research in rodents is currently conducted in post-pubertal adolescents to young adults (37). Because individuals may seek medical interventions for gender transition at various ages, it is imperative to determine whether the impact of typical hormone treatment regimens on pain and analgesia is age-dependent. To the extent that sex differences in anesthetic effects are known to change with age (202, 203), it might be predicted that the impact of gender affirming hormone treatments on pain and analgesia will depend on an individual's age. These observations in preclinical models are clearly suggestive of the information and clinical research issues that remain to be defined in transgender humans.

Hormone therapeutics for pubertal suppression in transgender youth

An important focus of hormonal therapeutics is the use of gonadotropin-releasing hormone analogues used to suppress endogenous puberty in transgender adolescents. The incidence of such therapy was reported to be 2.5% (∼515) of transgender people (n = 20,619) in one study (204). In select cases, both cisgender and transgender youth both utilize pubertal inhibition (“puberty blocker”) therapy. It should be noted that this therapeutic intervention is not new but has been utilized since the 1950s for use in children with central precocious puberty which results from premature activation of the hypothalamic-pituitary-gonadal (HPG) axis, and for endometriosis. Because of this, use in puberty suppression is explicitly an on-label use for GnRH analogues (205207).

Gonadotropin-releasing hormone (GnRH) agonists delivered as slow-release depot formulations are prescribed to suppress puberty for transgender adolescents. GnRH agonist therapy inhibits gonadal sex steroid production by persistently activating and desensitizing the GnRH receptor. This desensitization suppresses luteinizing hormone and follicle-stimulating hormone release from the anterior pituitary gland preventing the progression of puberty for the duration of GnRH agonist use (208). GnRH agonist use has been a component of therapy to manage gender dysphoria in adolescents (209). However, because GAHT is restricted to those above age 16 (recent guidelines suggest potential for use as early as 14), pubertal inhibition therapy is provided following reaching Tanner stage 2 as an intervening therapy. This provides the individual with more time to decide what physical interventions they may prefer. Several studies and reviews have suggested favorable mental health outcomes of puberty blockade (210212). The basis for these outcomes is that pubertal suppression prevents gender-incongruent changes which are not easily reversible (e.g., breast development in trans boys, changes in voice and bone structure in trans girls) (204, 213215). Beyond mental health, liabilities of pubertal suppression are poorly studied and controversial. Preclinical data has shown GnRH to have multiple effects outside of the hypothalamic-pituitary-gonadal (HPG) axis (216). While pubertal suppression with long term use of GnRH agonist therapy has been shown to have a positive effect on issues such as gender dysphoria, a variety of long-term effects have been identified, including polycystic ovary syndrome in trans boys, changes in body composition, metabolic profiles and bone mineral density, along with short term side effects such as headaches, hot flushes, mood swings and injection site reactions (rashes, bruising and sterile abscess formation) (217). Aside from the sequelae of acute treatment, systematic assessments data of the long-term effect of puberty suppression and pain biology is needed (218).

Research investigating pain in transgender children and adolescents on pubertal blockers is limited. While injection site pain is a well-known side effect of any injectable medication, it is unclear whether GnRH agonists used for pubertal suppression are associated with altered pain experiences in gender diverse youth. Adults and post-pubertal adolescents may therefore experience breast, pelvic, or testicular pain during the initial steroid hormone administration within the first 3 months, while recurring administration to achieve gonadal suppression may be associated with a moderate incidence (<10%–20%) of arthralgias, headaches, anxiety/depression/ irritability or dyspareunia among individuals with vaginas (143, 144). In addition, as GnRH agonists are first line therapy for the treatment of endometriosis, initiation of this therapy may lessen endometriosis-related pelvic pain, dyspareunia, and dyschezia in trans men (143). Despite the complex interactions between GnRH agonists and other forms of gender affirming hormone therapy, a retrospective case series of 8 youth (6 AFAB, 2 AMAB) diagnosed with gender dysphoria during chronic pain treatment demonstrated improved scores on the Pain Burden Interview, Functional Disability Inventory, and Pain Catastrophizing Scales which coincided with synergistic therapies focused on gender affirmation and pain (72). However, a study that compared 4,778 transgender teens to 630,200 cisgender students in California middle and high schools found on a self-report survey that transgender students were about 2−1/2 times more likely than non-transgender students to have used cocaine/methamphetamine and about 2.8 times as likely to have experienced use of inhalant, twice as likely to have used prescription pain medication and more than 3 times as likely to use cigarettes (219). Additional research that examines possible correlations among endogenous hormone suppression, exogenous hormone administration, mental health, gender dysphoria, and pain in transgender populations is needed. Further, is this important to consider in relationship to other variables, such as surgical status, more complex regimens (including progesterone, for instance), previous use of GnRH therapy, etc.? What might these studies look like? What kind of patient profiles should be detected? What about in cis women with low estrogen or cis men with high estrogen?

Future directions

While puberty suppression is effective in managing the gender dysphoria for many young transgender patients, there is little work characterizing the long-term impact of this treatment on pain or analgesia. The preclinical data reviewed here makes the case that this issue should be studied in more depth due to this treatment being used as a main suppressor of puberty. Further, will remain important this important to consider. Pain expression and phenotype in relationship to other variables, such as surgical status, more complex regimens, previous use of GnRH therapy, etc.? What might these studies look like? What kind of patient profiles should be detected? What about in cis women with low estrogen or cis men with high estrogen?

Sex differences in immune/inflammatory processes

Considering the changes in patient biology that result from gender affirming medical procedures in many transgender individuals, it is worthwhile to briefly consider sexually dimorphic responses to pathogens, immunizations, and inflammation (66, 220). As examples, dendritic cells isolated from cis women are generally associated with increased Type 1 interferon responses upon activation, as compared to dendritic cells from men, while stimulated macrophages/monocytes in cis women generally release significantly more pro-inflammatory cytokines (220). These divergent inflammatory responses contribute, in part, to the sex-specific differences observed in HIV-1 (221) and SARS-CoV-2 (222, 223) pathologies, and they have been attributed to (i) differences in gonadal hormones and ii) genes located on sex chromosomes. Social factors also contribute to one's risk of infection, and gender has been shown to play a role in behaviors associated with contracting COVID-19, such as wearing masks.

Indeed, in addition to peripheral immune cells, estrogens can influence inflammatory responses of CNS microglial cells and astrocytes. It is hypothesized that estrogens can exert a neuroprotective, anti-inflammatory effect in Parkinson's disease, by attenuating microglial and astrocyte responses to inflammatory stimuli (224). In addition to observational studies linking estrogens with reduced Parkinson's risk, estrogens have been used as a treatment for Parkinson's disease symptoms (224, 225). A similar mechanism is hypothesized for the apparent effect of estrogen treatment to reduce neuropathic pain (226, 227). That is, estrogen reduces microglial and astrocyte inflammatory responses via activation of estrogen receptors within these cells (226229).

Hormone replacement and immunity

Membrane-associated estrogen receptors (ESRs) act as G protein-coupled receptors to rapidly alter gene regulation and protein function (132, 230). Intracellular ESRs act as hormone-dependent transcription factors. Thus, the presence or absence of systemic estrogens directly influences immune cell transcriptome and functional activity. Not surprisingly, medically guided increases or decreases in circulating estrogens are highly relevant to immunity and inflammation, as nearly every cellular subset of the adaptive and the innate immune system express estrogen receptors (ESRalpha /ESRbeta (231). The influence of estrogens on immune cell function has been widely observed (220), with estrogen associated with reduced cytotoxic functional capacity of innate Natural Killer (NK) cells in animal models (232), e.g., lymphocytes that bind and kill tumor and virus-infected cells without antigen stimulation. In humans, not only do circulating NK cells isolated from cisgender women demonstrate reduced killing compared to those isolated from cisgender men (233), but cytotoxic capacity of NK cells isolated from those who take oral contraceptives is lower than those who do not (233). Furthermore, NK activity increases during menopause (234), when estrogen levels are reduced, and decreases during pregnancy (235), when estrogen levels increase. This ability of gonadal hormones to directly modulate NK function could be highly relevant within the context of neuropathic pain in transgender persons utilizing GAHT (236, 237).

The gonadal hormone milieu in which immune cells mature also influences their subsequent responsivity to estrogen. Latency reversal agents can be used in vitro to induce CD4 + memory T cells silently infected with HIV to produce viral RNA (238). With cells isolated from HIV + cis men (cells that presumably matured in a relatively “low estrogen” environment), the potency of latency reversal agents is reduced by simultaneous treatment with an estrogen. In contrast, in cells that matured in cis women, a relatively “high estrogen” environment, this reversal was completely abrogated by co-treatment with an estrogen. The duration and consistency of GAHT in trans persons could therefore result in a mosaic of gonadal hormone-responsive immune cells. Relatively short-lived innate immune cells might take on the associated transcriptome and phenotype of maturing in their therapeutic environment relatively quickly, similar to what is observed in post-menopausal cis women who utilize hormone replacement therapy (234). Conversely, long-lived adaptive and tissue-resident immune cells might retain the phenotypic characteristics (e.g., hormone responsivity and receptor expression) of the sex assigned at birth for more extended periods. Central nervous system microglial cells, for example, can persist for decades (239).

Future directions

Given the influence of gonadal hormones on immunity and inflammation, ensuring that transgender populations are included in basic, translational, and clinical research trials is important. Transgender persons who choose gender affirming therapies could have immune and inflammatory profiles distinct from those of cisgender individuals. Duration and consistency of GAHT should be considered, and study entry criteria that excludes transgender individuals from participation should be explicitly acknowledged. Thus, while the influence of gonadal hormones on peripheral and CNS inflammation and immunity has been investigated in a sex-binary manner, similar studies examining gonadal hormones in transgender persons are necessary (240).

Sex chromosomes

Consideration of the issues of pain patients (241, 242) requires consideration of chromosomal status. Sexual differentiation of pain sensitivity may begin very early in life in humans (243) and in preclinical models (244). As discussed below, preclinical studies show that chromosomal and gonadal hormonal (organizational and activational) mechanisms robustly contribute to sexual differentiation in the expression of the pain phenotype and correspondingly interventions that down regulate the transmission of the afferent signal and its integration, [e.g., analgesia (183, 245, 246)].

Healthy adult cis women typically show lower pain thresholds and pain tolerance than men when tested with various noxious stimuli (5, 242, 247), and when such evoked thresholds were assessed in patients with mononeuropathies (248). As discussed below binary differences have been typically reported in preclinical models. In migraine, inflammatory phenotypes such as rheumatoid arthritis, temporomandibular disorder, and conditions of widespread pain such as fibromyalgia are likely due to the role played by circulating immune complexes acting though adaptive immune signaling. The incidence of these pain conditions may be up to 2–3 times more common in women than men (40, 241, 249251). As a caveat, one must consider that as discussed above, there is a confound in the sex dependent differences given the influences of sexism and other social factors where providers have been shown to dismiss, under-medicate, and underreport women's pain.

These observations are consistent with the appreciation that the variation in human immune and inflammatory responses is robustly influenced by sex chromosomes (252, 253). Understanding the contribution of sex chromosomes to the pain phenotype complicated by their organization. The sex chromosomes are routinely considered in terms of their common pairing as “XX” or “XY”. In fact, a multitude of potential combinations exist, and often the number of X chromosomes directly influences inflammation and immunity (254, 255). While the X chromosome is not necessarily enriched for immune genes as compared to autosomal chromosomes, it encodes for a number of highly relevant genes, including genes associated with the inflammatory transcription regulator, nuclear factor B (NF-kB) (255, 256). In addition, mutations within these essential genes can result in severe immune defects in individuals with only one X chromosome, as for example in X-linked severe combined immunodeficiency (XSCID) (257). Regardless of the number of X chromosomes present, additional X chromosomes are typically silenced epigenetically (253, 258260). In the absence of mutations, the presence of more than one X chromosome is broadly relevant to the immune and inflammatory phenotype (258). Defining the impact of these different X pairings has been a subject of interest in preclinical literature and will be discussed further below.

Future directions

The interactions of chromosomal constituency and GAHT must be defined scientifically in transgender populations. Consideration of both gonadal hormones and sex chromosomes, e.g., “estrogen rich XY” or “estrogen low XX”, will be of scientific and practical importance in further elucidating sex- and gender-specific pathways of inflammation and pain. It is important to note that a variety of complexities serve to complicate the role of chromosomes and pain. Thus, for example, what might be the contributions of non-46,XX/46,XY chromosome combinations? What is the role of the Y chromosome in activating a cascade or does “silencing” the second X chromosome?

Environmental exposure: impacts of chronic social stress

Transgender persons frequently navigate a “pro-inflammatory” social environment which as reviewed at the outset have demonstrably significant effect upon morbidity and quality of life (261). Both acute and chronic exposure to social stressors, such as rejection, isolation, and actual or anticipated threats of physical assault have been shown to generate physiological inflammatory responses (262, 263). Social stress induces expression of NF-kB, leading to increased concentrations of circulating soluble markers of inflammation, such as IL-6 and TNF-alpha (264267). While this is true for all individuals regardless of sex or gender, trans persons disproportionately experience the burden of (i) negative external stressors, (ii) the stress of hypervigilance in anticipation of negative external stressors, and (iii) socially conditioned and internalized stress (268).

Furthermore, the Gender Minority Stress and Resilience (GMSR) model revealed that these chronic social stressors encourage trans individuals to adopt negative coping behaviors (e.g., substance abuse) which in turn also negatively influence immunity, inflammation, and health outcomes (269).

It has therefore been hypothesized that chronic exposure to social threats is a primary driver of systemic inflammation and health disparities in sex- and gender-minority populations (263). A study of trans men utilizing GAHT, typically testosterone therapy to suppress estrogen, reported that serum C Reactive Protein (CRP) concentrations, a measure of inflammation, were associated strongly with the emotional stress related to being misgendered and misnamed during daily activities. Examination of diurnal cortisol functioning in transgender men undergoing testosterone therapy revealed that elevated diurnal cortisol levels at awakening were associated with transition-specific social stressors, such as those experienced during transitioning, coming out and common life events such as utilization of gender-specific public bathrooms (270). This is highly relevant to pain experiences in transgender persons, as elevated concentrations of circulating CRP have been associated with pain sensitivity (271); however, this topic is largely underexplored within the literature.

Reviews of available literature have attempted to extrapolate data associating stress in transgender men and women to migraine incidence. These reviews have concluded that social stress, particularly stress induced by stigma in an unsupportive healthcare environment, in combination with GAHT, may contributes to the higher rates of migraine in transgender persons (272). This represents a substantial gap in the primary research literature which should be addressed directly with research addressing migraine in cis and trans populations (273). For context, many classes of environmental and societal stressors have been shown to have a direct impact on a number of measured variables, including: psychological processing, concern over financial well-being such as access to affordable housing (274), general concern over health (275), access to health care (276), and food insecurity (277).

While a survey of over 2,000 trans and gender non-conforming participants found no differences in chronic health conditions or health behaviors (278), these data are of limited utility as the survey was phone-based, and thus excluded potential participants who have experienced more severe social stress (poverty, homelessness or incarceration). Furthermore, physiological effects of social stress can be mitigated by familial and social support (279). An exploratory study found no associations between gender minority stress and resilience scales and circulating concentrations of CRP in young trans persons receiving GAHT (279, 280). Notably, 84% of these older teenagers and young adult participants lived with their parents, indicative of stable familial support, perhaps suggesting that a supportive familial structure is “anti-inflammatory” in an otherwise “pro-inflammatory” society (281, 282). These observational studies highlight the importance of family support for psychological wellbeing as well as for material survival (adequate health insurance might only be possible through parents or spouses, financial security through social security income relies on traditional family structures). This is a challenge for many transgender persons, many of whom lose their family support structure when they transition (283).

Future directions

When examining inflammation and pain outcomes in transgender populations, recruitment strategies, data analysis, and conclusions should consider the pro-inflammatory influence of social stress. Studies inclusive of transgender populations in the United States could be dominated by recruitment within urban areas, potentially overlooking the diversity of rural America. The Movement Advancement Project (MAP) released a report in 2019, that included an original analysis of the unique challenges and opportunities for transgender people in rural America. Although discrimination occurs as frequently in rural and urban areas, the structural differences in rural America amplified the effects of discrimination due to less employment opportunities and healthcare options (284). For future studies addressing the impact of social stress on transgender health, it will be important to consider the diversity of different geographic areas globally and to include both urban as well as rural populations. Recruitment strategies (e.g., telephones, engagement with advertisements on social media) could inadvertently exclude transgender persons who have experienced or are currently experiencing severe social stress. Recruitment, research, and clinical staff who interact with transgender participants should receive appropriate training to eliminate introduction of known pro-inflammatory social stressors into the research process (e.g., mis-gendering, mis-naming, and judgmental or exclusionary comments) and should preferably contain transgender team members. Data analysis and study conclusions must thoughtfully incorporate quantitative measures of participant stress. Transgender identity is not pathological, nor is it inherently a pro-inflammatory condition. However, stressors that trans persons disproportionally experience are typically considered to be pro-inflammatory, as indicated by endpoints such as CRP, as noted above (270, 280).

Impact of emotion and body image on pain in transgender people

The report of a pain state results from both peripheral nociceptive signals and sensitization (e.g., tissue injury, joint inflammation) and effects of attentional and emotional elaboration in the brain (285). Attention heightens the perceived intensity of pain (286), while negative emotions increase its unpleasantness (287289). Trauma, depression, and mood (290) alter the brain's emotional learning circuitry (291) and contribute to pain chronification (292294).

As reviewed in the preceding section, trans people face disproportionate rates of violence (295, 296) and discrimination, including in healthcare (297). Violence and discrimination have significant biological consequences which are manifested in heightened negative affect and psychopathology. The gender minority stress model posits both external and internal stressors related to gender minority status (272), leading to higher prevalence of depression, anxiety, and suicidal ideation among transgender individuals relative to the general population (298300). Prevalence of syndromes such as fibromyalgia, reflecting a likely role for circulating immune complexes resulting in amplification of pain processing, appears to be greatly heightened in transgender men and women (53). Violence is a certain risk factor for post-traumatic stress disorder (PTSD) (301), which in turn increases the risk for opioid use disorder (302). Anti-transgender bias and non-affirmation, which increase severity of PTSD symptoms (303) and risk of opioid misuse, highlight the need for systemic improvements in transgender inclusivity in medical and behavioral healthcare (304) and in peer support (298).

Gender dysphoria and body dysphoria constitute additional internal risk factors for heightened experiences of pain. Many transgender individuals experience significant gender-related body dysphoria and avoidance, particularly for body parts associated with sex differences (73, 292, 305, 306). For trans men, body dysphoria is often focused on breasts, genitals, and lack of facial hair (307309), while in trans women, body dysphoria frequently focuses on the genitals, face, and presence of facial hair (310). The effect of body dysphoria and reduced bodily self-identification on the experience of pain in transgender individuals is not known. However, there is evidence that body satisfaction and body representation in general modulate experiences of pain. Thematic analyses of transgender persons’ descriptions of their body part-specific dysphoria have highlighted independent themes of disconnection from the body (feeling that a body part is viscerally wrong, deformed, or alien), and emotional distress (negative emotional reactions and hyper fixation) (311). Body avoidance and dissociation, which may be elevated in transgender patients (312), may reduce pain perception due to reduced attention to the body part, similar to patients with spatial (hemi-) neglect (192). There have been reports that modulation of pain perception can be induced by observing a rubber hand that was perceived to be the observers own hand (313, 314). In contrast, heightened negative affect surrounding a dysphoric body part may increase pain perception. In patients with arthritis, negative body image has been correlated with greater pain severity (315). Conversely, laboratory studies using rubber limb coverings mimicking an injured or hairy limb/hand appearance showed that negative body image increases acute pain (316).

Conversely, positive emotions such as gender euphoria may reduce experiences of pain. Transgender people appear to have lower rates of phantom breasts and penises after gender-affirming surgical procedures than cisgender individuals do after these body parts are removed for medical reasons (317, 318). In addition, transgender men exhibit lower pain and opioid consumption after breast removal than do women after mastectomy for cancer or risk of cancer; although hormone therapies may play a role, this difference may reflect congruence/gender euphoria of breast removal for trans men versus the distress and incongruence for cisgender women (as well as negative emotions associated with cancer) (118). An important caveat, however, is that these procedures are not identical for transgender and cisgender patients. Euphoria can also arise from phantom body parts aligned with gender and from the use of prostheses, increasing positive identification with the body (319, 320).

Concerns about bias or body dysphoria during clinical encounters may lead patients to delay seeking care for acute pain, elevating the risk of transition to chronic pain. Provider misgendering and microaggressions are likely to amplify gender dysphoria, leading to stress, and consequently, worsening or prolonging the pain.

Future directions

We pose the following questions regarding the effects of body image on pain in transgender individuals: (1) What are the direct and indirect effects of gender and body satisfaction on pain in transgender individuals, and what variables affect this relationship? (2) Are certain pain conditions disproportionately experienced by transgender patients, and are they linked to dysphoric body parts, and/or to strategies of body avoidance or hyper fixation? (3) What is the impact of patient satisfaction with gender-affirming procedures on existing pain conditions or severity of postsurgical pain? (4) How can healthcare providers reduce body-related bias and microaggressions, and what is the effect of such improvements on body dysphoria and pain? (5) Are there novel approaches to improving body image in transgender individuals that might reduce acute or chronic pain?

Neuroimaging in the study of gender identity and pain

A topic of enduring interest across many fields of neuroscience relates to the influence of sex and gender on human brain anatomy (321, 322), function, connectivity, and metabolism. Albeit the additive impact that gene expression, own-body metabolism, environment, and society can surely have on the behavioral differences documented between cisgender men and women (323), the question remains as to how putative anatomical, functional, and metabolic brain regional characteristics might also contribute—or be shaped by—gender identity and expression. It is of note that the traditional illustration of the body map in the brain, the homunculus, by Wilder Penfield refers to males with testicles, penis, and no breasts. Only 10 years ago there was a call for the need to a full mapping of the female brain with the production of a hermunculus as a first step toward fully understanding neurological and physiological sex differences related to pain conditions and pain management (324).

Gender and body dysphoria are associated with differences in neural body representation. Prior to any surgical procedures, some trans men have reported the sensation of having a penis (317, 319, 320, 325)—similar to the phantom limbs that frequently occur after amputation (326). This phenomenon may suggest a strong representation of gender-congruent body sex in the brain. Indeed, breasts that feel incongruent on trans men elicit a lower somatosensory response in brain areas associated with salience and body ownership, but increased activation in a brain region associated with alarm, in comparison to the activation imaging in cisgender people with congruent-feeling breasts (327). Correlated differences in brain connectivity suggest differences—whether innate or acquired—in body representation (327). Similarly, neuroimaging studies have reported that trans men show weaker connections between body perception and body ownership networks (328), and decreased connectivity within the default mode network (329, 330) and anterior cingulate cortex, the latter related to lower ratings of “self” for gendered body images (331). Body ownership refers to the perceptual status of the observer's own body, which makes bodily sensations seem unique to oneself. A positron emission tomography (PET) imaging study has indicated that this was related to activation of right posterior insula and the right frontal operculum (329). Individual differences in the report of body ownership have been associated with the cortical thickness in the somatosensory regions, the temporo-parietal junction, the intraparietal areas, and the occipitotemporal cortex (330). These studies cumulatively suggest the possibility of decreased connection between sensory body representation and brain areas involved in self-identification as the potential neural correlate of the gender-related body dysphoria experienced by many trans people.

Advances in neuroimaging techniques have enabled the possibility of quantifying and studying in a great level of detail neurobiological characteristics relating to brain structure, function, connectivity, metabolite and neurotransmitter concentrations, and nerve fiber tract integrity and activity-dependent changes in blood flow (332), all which have been explored in relation to sex and gender expression. Several studies reviewing investigations on the topic (333338) refer to structural, functional, and metabolic brain features “exhibiting signs of masculinization or feminization”, as the result of analyzing results from research comparing transgender to cisgender groups of individuals. However, the very basis of assuming that male and female brains are categorically different is hugely controversial (339).

The existent literature reports sex differences in brain anatomy on a global scale, regarding absolute volumes (340342). Indeed, a consistent observation in both pediatric (343, 344) and adult (341, 345348) in vivo magnetic resonance imaging (MRI)-based morphometric analyses is an approximate 9%–12% greater brain size in males compared to females. Higher percentages of white matter in males in comparison to females (341, 349) are also frequently documented, as well as larger ratios of grey matter to white matter in females, even after correcting for the effect of total intracranial volume (345, 350). Studies also report sex differences in the relative sizes and shapes of regional brain structures, with the direction of the sex effect varying between regions, including the Broca's region (340), corpus callosum (351), amygdala, insula, and hippocampus (342, 352). This notion has been, however, challenged by meta-analyses that conclude the opposite (353, 354) after accounting for differences in head size. Research efforts by Joel et al. (339) into sex differences in the human brain have resurrected the idea of the “human brain mosaic”, proposed by McCarthy and Arnold four years earlier (355). Their findings, following analysis of MRI data of 1,400 individuals from four different datasets, revealed substantial overlap in the distribution of anatomical traits between men and women in all brain regions and connections examined. A wider study using data from 5,216 UK Biobank participants cautiously highlights limited sex differences in functional connectivity traits, while also referring to “considerable distributional overlap between sexes” in regional anatomical volumes, surface areas, and white matter microstructure (341). This reinforces the idea that human brains cannot, in fact, be distinctly categorized into two distinct classes, but rather that male and female brains are composed of “unique mosaics” of features, some of which are more common in one sex than the other and some that are common in both. It has been suggested that computational meta-analyses of MRI, personality, and behavioral studies have found that human brains do not exist in a “male” or “female” state (339, 356, 357). Yet, it is plausible that gendered constructions may alter gene expression through epigenetic means to produce, reinforce, or counteract endogenous sex differences (356) Given the plethora of conflicting and ambiguous results, the consistency and etiology of sex/gender differences in human brain structure and connectivity remain elusive (350).

Neuroimaging representation of pain

Pain depends on the integration of many sensory signals, and is also influenced by biological, psychological, and social factors (358, 359). While molecular mechanisms of peripheral pain transduction have been well developed over the last few decades (360), the organization of central pain circuits is incomplete and far from being well understood. A review on pain modulatory mechanisms proposed a network involving predominantly medial and frontal cortical brain areas, combined with specific subcortical and brain stem nuclei to confirm a “key system” in the modulation of pain (361), with the prefrontal cortex likely intervening in the generation, maintenance, and integration of pain relief (361). It has also been proposed that the central processing of pain relates to the thalamus, despite evidence that pain is also controlled by cortical mechanisms in areas such as the cingulate gyrus and insular cortex (362).

Within the neuroimaging techniques, functional MRI (fMRI) including resting-state (rs-fMRI), and positron emission tomography (PET), have emerged as important tools that provide a window into pain mechanisms, highlighting the essential role of cortical and mesolimbic brain regions in modulating pain responses (363). Neuroimaging studies have shown altered brain structure and function in chronic pain states (364). Some researchers support the hypothesis that opioids and the orbitofrontal cortex modulate pain by hedonic experience, as the ventral striatum and dopamine mediate motivation drive by a painful stimulus (365). Others identify the posterior insular cortex as an important pain center, targeted in deep brain stimulation treatments to increase pain thresholds (366). Structural and diffusion tensor neuroimaging studies of patients with chronic musculoskeletal pain provide moderate and yet inconclusive evidence that microstructural white matter and regional grey matter volume in brain regions encompassing the cingulate cortex, insula, and superior frontal and temporal gyrus relate to pain intensity and sensitivity in these patients (367). Given the varying, yet complementary, information that can be obtained from different neuroimaging techniques, the complexities in assessing and managing pain, and the lack of sensitivity and specificity of individual pain biomarkers (neuroimaging included), a framework that combines different neuroimaging modalities with non-imaging data sources (i.e., actively and passively recorded bio- and psychometrics) seems to be the way forward for personalized pain diagnosis and management (368).

Brain neuroimaging in relation to transgender identities

Neuroimaging studies suggest that, before GAHT, most transgender individuals exhibit brain features that more closely resemble their assigned gender at birth (335338). However, some neuroimaging studies in transgender individuals have found that cortical thickness (335, 338) and subcortical volumes (337) may be more similar to those of their gender identity. This is also the case with white matter microstructure (336338), especially in the inferior fronto-occipital white matter tract, connecting the parietal and frontal brain areas that mediate own-body perception (421), a finding confirmed by MRI studies (369). One recent MRI study comparing 24 cis men, 24 cis women, and 24 trans women (who had not undergone any GAHT) suggested a shift away from the brain anatomy typical of the cisgender men group towards the one typical of the cisgender women group in the transgender women (370).

Conflicting results exist in regard to volumes of subcortical regions like the nucleus accumbens and thalami in transgender populations compared to cisgender individuals, with some studies finding differences between both groups (371). A study that enrolled 66 transgender individuals (33 trans men and 33 trans women) reported larger putaminal volumes in the transgender group (especially trans women), compared to all cisgender participants (372).

It is noteworthy that hormone-dependent decline in brain volume has been reported to occur in postmenopausal females, or in females receiving long term anti-estrogens (373, 374). Conversely, post-menopausal females undergoing hormone replacement therapies display significantly larger volumes as compared to menopausal women without therapy, of several regions including superior/middle/Inferior frontal gyri, hypothalamus, inferior temporal gyrus, parahippocampal gyrus, hippocampus, cerebellar cortex, postcentral gyrus, precuneus, angular gyrus, supplementary motor area, superior occipital gyrus, and precentral gyrus (374).

Recent neuroimaging studies have shown functional, metabolic, and structural differences in transgender populations precisely in cortical areas that modulate pain and in sensori-motor regions (Supplementary Table S2), some of which are particularly relevant after hormonal treatment. Another study found an increase in resting-state functional connectivity between the left thalamus and the left sensori-motor cortex/putamen following GAHT in individuals that had a gender-affirming surgery, with an especially higher activation in a cluster within the subcallosal cortex (375). These results suggest that the hormonal therapy applied after gender affirming surgery may modulate functional connectivity in regions engaged in emotional and sensori-motor processes, which are part of the pain modulatory network. This suggestion is consistent with the research in preclinical models as discussed further below.

Limitations in the analysis of the neuroimaging literature

It is difficult to establish causality between brain structural, metabolic, and functional gender differences and psychological stress factors. In other words, it is not known whether brain appearance and function are determined by psychological stress factors due to gender differences, or vice versa. Evidence from neuroimaging studies suggests that mental processes (e.g., thoughts, feelings, beliefs, volition) significantly influence various levels of brain functioning (e.g., molecular, cellular, neural circuit) and brain plasticity (376). Specifically, there is evidence that sustained stress-related factors play a role in brain structure and function (377), and that gonadal hormones influence immunity and inflammation, both of which affect the brain, directly and indirectly (378380). A systematic review of neuroimaging studies in relation to gender (333) excludes studies on neurological diseases or aspects related with neurological outcomes, while other studies on these populations limit their analyses to sex differences assuming gender binarism and congruence between the sex assigned at birth and gender identity (381383). It is also worth noting the complexity in the analysis of factors influencing the representation of sexes in clinical trials, even in diseases that are not supposed to be sex-related (381), and the possibility of bias in the literature given the way results are reported. Also, few original studies are truly exploratory in the sense that they explore the whole brain, while most base their analyses on hypotheses that involve only a surprisingly few brain regions. This may contribute to unconscious selection bias in any literature analysis that includes them.

Future directions

Varying sample sizes, recruitment of convenience samples, duplicate reporting of findings in more than one publication, alternative protocols, and processing in neuroimaging are among the various potential factors responsible for inconsistencies in existing literature surrounding sex and gender-related differences in regional brain volumes (384). Considering that the surfeit of studies examining differences across brain substructures are typically underpowered and inconsistent (385), it is imperative that larger sample sizes are used when testing hypotheses regarding putative sex/gender differences in the human brain (341). Given the differential effect that age has on human brains, large-scale follow-up studies are crucial to delineate how sex/gender differences develop and change throughout the lifespan. In doing so, greater strides can be made in elucidating the neuroanatomic substrates and sex/gender-related differences in human behavior (349). Given individual differences in expectancies and conditioning effects of brain networks that modulate pain experiences, and the known effect of the psychosocial context that accompanies any therapy, further research is needed to estimate and document the degree of expectancy of nocebo responses in relation to gender identity and social factors (359, 376, 386). Neuromodulation, a technique used in functional neurosurgery to restore the brain functions by modulating the neuroanatomical and neurochemical circuits that regulate them, albeit compensating functional disorders (e.g., pain), could also have an effect in neuronal circuits underlying gender expression. Therefore, the impact of this technique on identifying neural circuitry that covaries with gender expression would also benefit from further investigation. Longitudinal neuroimaging studies on gender expression are currently limited to short-term investigations of the effects of gender affirming procedures (387390) and cover a geographically limited sample. Long-term neuroimaging studies on gender expression across the lifespan, specifically in relation to pain, are needed.

Can variables reflecting the role of sex be modeled in rodents?

In this final section, we raise the issue of future directions in preclinical research relevant to the understanding of pain in the context of biological variables underlying the development of sex and gender. Here, we note that much of our predictive understanding of the role of neural substrates in defining human pain biology has arisen from preclinical studies. However, the preclinical model does not presume to mirror the complexity of the biological and psychological aspects of sex and gender in humans.

As reviewed in this manuscript, there is evidence for the influence of hormones (391, 392), sex chromosome complement (393), and early life experience (394397) in the pain phenotype expressed in human variables that are evident and controllable in the preclinical models (Supplementary Figure S1). Given the limitations posed by the study of experimental variables in humans vs. animals, we cannot discern how the impact of these variables on the human experience is impacted by each individual's sex characteristics or gender expectations. Even when sex chromosome complement and hormonal profile are discordant, such as in complete androgen insensitivity syndrome in which XY genotype is accompanied by external female phenotype (398), we do not know how much gendered expectations (e.g., induced by being raised as a girl) impact brain organization and connectivity as they are established during development. Modeling aspects of sex differences in laboratory animals such as rats and mice may with caution enable us to disentangle the impact of not only hormones and sex chromosome complement (which can be distinguished in rodents much more readily than in humans), but also to begin to understand how environmental variables and gendered expectations influence biology and behavior, including the pain phenotype and the regulation of its expression. We have empirical evidence regarding the role of hormones, chromosomes, and experience (e.g., maternal care) in rodents, but this has not been achievable in humans, leaving essential questions unanswered.

Sexual differentiation of the rodent and human brain

The classic view of brain sexual differentiation begins with the onset of androgen production by the fetal testis during the 2nd trimester in humans and the last quarter of pregnancy in rats and mice (399). In rodent models, decades of evidence confirm the masculinizing impact of perinatal androgen exposure on the size of entire brain regions, nuclei and subnuclei, as well as the strength of projections, patterns of synaptic connectivity, and neurochemical identity of select neural populations (400, 401). These differences are established during a critical period that ends prenatally in humans and in the first few days of life in rodents. The critical period is not the same as the sensitive period, which is defined as the developmental window during which the brains of genetic females remain responsive to exogenous hormones (402). This sensitive period for hormonal modulation is believed to end in utero in humans but in rodents ends by the first week of life (403). By the time of birth in humans and the first week of life in rodents, hormonally induced, enduring sex differences in the brain, are firmly established, even though the brain is still highly immature, having decades left to refine in humans and almost a month in rodents. The transition from puberty to reproductive maturity is increasingly appreciated as a distinct critical/sensitive period (404, 405), but the period prior to that—toddler-to-childhood in humans and post-weaning in rodents—has been largely ignored in regard to biological origins of sex differences. In humans this period is characterized by exposure to strongly gendered expectations. It provides a speculative hypothesis, that such regional changes during this time may provide an opportunity to address the role of evolving gendered expectations in driving biological parameters such as circuit refinement, synaptic strength, and connectivity. It is clear that even in this brief overview, brain and behavioral development viz sexuality reflect multiple variable that are difficult to deconvolute (399, 406).

Efforts to parse the relative influence of hormones, sex chromosome complement, and sex role expectations

In rodents, by comparing genetically modified mice to XY males with testes and XX females with ovaries, gonadal hormone effects can now be distinguished from sex chromosome effects. This “4 core genotype” model has revealed specific sex differences that are entirely determined by hormones, others that are entirely determined by sex chromosomes, and others that appear to be a blend of both. For example, sex differences in heat pain thresholds in neonatal mice were found to depend on sex chromosome complement and be independent of gonadal status, whereas sensitivity to mu opioid agonists depended on the presence of testes (245). The 4 core genotype model could be used much more widely to untangle sex chromosome complement vs. hormonal influences on chronic pain states, as well as on analgesic sensitivity. Further research into the influence of sex chromosomes and hormones on pain and analgesia could help us to develop insights into transgender individuals who elect to transition hormonally and/or surgically at various points in development, vs. those who do not.

Impact of environmental context

Environmental context reflects conditions such as an enriched or deprived stimulus, milieu, and social and physical components of the environment in which the organism functions, but which induce stress to the degree that the environment challenges the repertoire of the organism's adaptive response. Aside from the acute effect of such environmental cues, the rearing environment has been noted to have prominent and enduring effects upon brain function and behavioral responsivity and engagement in humans and animals. In rodents, early environmental and psychosocial stressors can robustly influence cortical morphology and microglia resulting in adaptive responses at the level of synaptic, circuit, and neuroimmune signaling (407, 408).

An interesting example of preclinical research that might arise in this area relates to the question of being reared with gender expectations. Preclinically, the question would be phrased as assessment of the impact of sex role expectations on brain function. Although rodents develop to reproductive maturity much more rapidly than humans, they progress through comparable stages with corresponding epochs of brain development (Supplementary Figure S2). Human social systems and gendered expectations are extremely complex and clearly dependent on cultural differences. However, in preclinical models, the strongest gendered expectations are most readily modeled in animals and focus on nurturing. Male and female rats as young as 24 days old will both retrieve and nest pups (409). Juveniles sensitize to pups faster than adult virgin females, but adult males rarely do so, instead shunning pups or attempting to kill them (410, 411). Moreover, maternal behavior can be readily induced in adult virgin females by mimicking the hormonal milieu of pregnancy (412), but the same treatment in adult males is without effect. Thus, divergence from a common nurturing response in juvenile males and females to an opposite response by adults provides an opportunity to model how a sex role alters brain and behavior. This could be achieved by rewarding juvenile animals while they engage in nurturing and determining how that impacts adult brain and behavior. Thus, will a naturally occurring nurturing response in prepubertal males and females that diverges when they are adults shift when a sex role is manipulated? If so, such a manipulation could be utilized to begin to understand how gendered expectations early in life (in addition to sex chromosome complement and gonadal hormones) can influence brain and behavior and its impact upon pain processing using a rodent model.

The influence of development environment on brain connectivity and function extends beyond issues of sex and gender. In humans, early life stressors can markedly influence psychosocial reactivity, pain sensitivity and pain experience later in life (394397). Higher socioeconomic status shows covariance of efficient cortical networks in adulthood with effects varying with early age of exposure (413). While these early experiences can have a learning component, these developmental experiences can translate into lifelong changes in cerebral function and behavior though the actions of circulating hormones. This leads to, for example, microglial activation and changes in synaptic pruning, an altering connectivity (408), or epigenetic activation and silencing of DNA. Such changes lead to long term changes affecting how our genes are read and transcribed, producing enduring effects of early experience and the environment in humans on brain function and on psychosocial function and pain behaviors much later in life (414416).

Future directions

It should be stressed that the preclinical model does not presume to mirror the overall complexity of the biological and psychological aspects of sex and gender in humans. However, these models allow one to systematically assess issues relevant to system function. In the context of the present review, these issues include the impact of hormone therapy on neural functioning or the response of the system to surgical interventions and to environmental stressors. The use of genetically and chromosomally modified animals in conjunction with studies in humans may provide exciting insights which can illuminate the contributions of biology, sex, gender, and the environment in which the organism is born, nurtured, and matured assuming ethical standards in these studies can be met. As noted above, behavioral investigations of the environmental context in which an animal is raised would have particular significance in studies examining brain function and biology, particularly as with the role of epigenetics in developmental expression of sex related behavioral phenotypes. Continued research into models such as the gonadectomized animal and assessing depression-like symptoms displaying a sex-difference in relationship to maternal care when young.

NIH focus on transgender health

The National Institutes of Health makes it abundantly clear in its 2021–2025 Strategic Plan that it supports and promotes research that considers “the effects of sex and gender in study design, data collection and analysis, and dissemination of findings,” which “will help to inform the development of prevention strategies and interventions for everyone.” It goes on to elaborate that “Underserved groups—including Black, Latin origin, and Indigenous and Native American persons, Asian Americans and Pacific Islanders, and other persons of color; members of religious minorities; lesbian, gay, bisexual, transgender, and queer (LGBTQ+) persons; persons with disabilities; persons who live in rural areas; or persons otherwise adversely affected by persistent poverty or inequality—have distinct health needs and often experience disparities in health outcomes. NIH maintains that racial and ethnic minorities, rural residents, people with low incomes, SGM, and other populations experiencing health disparities should be included in all relevant research, such that there is sufficient representation of each population to conduct relevant analyses. Inclusivity in research generates more broadly applicable information and improves scientific understanding of the health and well-being of specific population groups,” (417). Despite these efforts, the enrollment reports for NIH-funded studies currently only allow reporting of 3 categories: male, female, and unknown/not reported.

As there is often no category for transgender identity in NIH enrollment reports, data on this population are often omitted or ‘hidden’ under the categories male, female and unknown/not reported. It will be critical to uniformly add categories for transgender identities (and other underserved groups such as sexual minorities) into NIH enrollment tables and to specify if “male” and “female” are meant to refer to cisgender men and women, respectively, or if “female” includes cisgender and transgender women and “male” includes cisgender and transgender men. This is essential so that the health and the health needs of this population can be studied and compared effectively throughout all NIH-funded studies.

The NIH, with the Office of Research on Women's Health (ORWH), developed the policy notice on Sex as a Biological Variable (SABV) which states that the “NIH expects that sex as a biological variable will be factored into research designs, analyses, and reporting in vertebrate animal and human studies. Strong justification from the scientific literature, preliminary data, or other relevant considerations must be provided for applications proposing to study only one sex…” (418). While the SABV policy does not pertain to gender at this point, including gender analysis in NIH research is being encouraged on many fronts. The ORWH considers gender a critical variable in its strategic plan which states: “…research focused on understanding scientifically important sex and gender differences as well as investigations of the many factors underlying the health of women are integral to NIH's mission to seek fundamental knowledge about the nature and behavior of living systems and the application of that knowledge to enhance health, lengthen life, and reduce illness and disability” (419).

In terms of pain research, the NIH Pain Consortium was established in 1996. It informed the pain research field via its flagship program announcement (PA), titled “Mechanisms, Models, Measurement, & Management in Pain Research” (420). This program announcement was first developed in 2006 and has historically expressed and currently expresses the NIH interest in pain, which considers sex and gender. For example, this PA asks for “Mechanisms that underlie gender and cultural differences.” This clearly provides a funding platform to address many of the key variables that this review has sought to illuminate in understanding pain and it mechanims and management in the transgender individual

Summary commentary

We have sought to review the diverse literature on the intersection between sex and pain with a focus on transgender individuals. This literature has broadly focused on the diversity of the transgender population, societal factors that impact pain in trans men and women, medical interventions as with the use of gender affirmation hormone therapies, pain phenotypes in patients undergoing gender affirmation surgeries, outcome from neuroimaging assessments, and the contributions of preclinical models. Many transgender individuals seek medical interventions to change sexual features of their bodies, to better align with their internal sense of self; the interaction of these procedures with pain is clearly under studied. The emotional consequences of gender dysphoria and societal stigma present a very heavy emotional burden that has been associated with high rates of depression in trans individuals, with significant implications for pain. Our review highlights the need to further explore the disproportionate effect of pro-inflammatory social stressors together with the impact that physiological and epigenetic factors may have (if any) to adequately address their impact on mental health and influence on pain responses in transgender patients.

Differences in gender identity, surgical interventions, exogenous hormone administration, and minority stress may result in significant differences in pain risk and pain experience in transgender patients. There is limited but increasing evidence that trans individuals may acquire pain phenotypes most similar to cisgender individuals of the same gender. Accordingly, treating and managing pain in transgender patients as in cisgender patients requires appreciation of the roles played by all facets of sex, gender, medical interventions, clinical conditions, and social factors. It should be noted that we still lack data about pain phenotypes in nonbinary people and how they may perceive pain.

While current clinical reports have largely revealed favorable outcomes in patient quality of life, this review has also emphasized the need for further long-term, large-scale, research in pain mechanisms and clinical interventions in transgender individuals, and, with it, the need for a concerted effort to advance our understanding of the influence of the long term outcomes of such interventions in the physiological and emotional aspects underpinning pain responses. Similarly, more research is required to better address pain phenotypes in transgender patients undergoing hormonal and surgical interventions. Advances in epigenetics will provide exciting insights into the impact of life and environmental influences (stressors) and their interaction with sex assigned at birth and gender as it addresses the mechanism of pain, analgesia, and other psychological parameters.

Due to the complexity of the factors that may affect pain perception and considering the limitations of current research data, every individual presenting with pain, acute or chronic, requires a personalized approach to pain management that takes into account the factors considered in this review. Future research targeted at these issues is of paramount importance to human welfare in general and to trans individuals in particular.

Author contributions

All authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication.

Conflicts of interest

ETS and UCSD has received funding and/or product for research from Epimed International (Dallas, TX) SPR Therapeutics (Cleveland, OH), Infutronix (Natick, MA), and Avanos (Irvine, CA). LC. Associate Editor for Frontiers in Pain Research. LC did not participate in the journal review process leading to the acceptance of this manuscript. MV. Chief Specialty Editor in Frontiers in Medical Technology. UW serves on the External Consultant Board for the “NIH Preclinical Screening Platform for Pain” (NIH/NINDS). In her capacity as a special government employee of the US Food and Drug Administration (FDA), she has served as a voting member of the FDA Anesthetic and Analgesic Drug Products Advisory Committee. In the past 3 years she has received compensation for serving on advisory boards or for consulting activities for Aphrodite Health Inc., Wilmington, DE, Avenue Therapeutics Inc., New York, NY, Bayer Aktiengesellschaft, Leverkusen, Germany, Biohaven Pharmaceuticals, New Haven, CT, and Syneos Health, Morrisville, NC, all unrelated to the submitted work. Reports research grants from the US National Institutes of Health. TY reports being a founder of Raft Pharmaceutical, SAB Navega Pharma all unrelated to the submitted work and is the Editor in Chief for Frontiers in Pain Research. TY did not participate in the journal review process leading to the acceptance of this manuscript.

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fpain.2024.1241015/full#supplementary-material

Supplementary Figure S1

Relative contributions of variables to sex differences in brain and behavior. We have empirical evidence regarding the role of hormones, chromosomes, and experience (e.g., maternal care) in rodents, but this has not been achievable in humans, leaving an essential question unanswered.

Supplementary Figure S2

Epochs of development in humans and rodents. Hormonal effects on the developing brain are largely complete prior to birth and re-emerge at puberty, but the intervening years are characterized by dynamic brain development sensitive to modulation by environment and experience.

Supplementary Table S1

Concepts and terminology. It is appropriate that we here consider an annotated nomenclature and associated concepts relevant to commentary related to gender and sex characteristics.

Supplementary Table S2

Summary of the relevant findings of papers showing whether or not and in which way the specific brain cortical areas that intervene in pain modulation differ in relation to transgender identity. The papers showing primary analyses were identified by searching Web of Science with the terms “neuroimaging”, “transgender, *” and “cort*” on 26.01.2022 (i.e., reviews and opinion papers were excluded). MRI, magnetic resonance imaging; fMRI, functional magnetic resonance imaging; rs-fMRI, resting state fMRI; PET, positron emission tomography.

References

1. Matsuno E, Budge SL. Non-binary/genderqueer identities: a critical review of the literature. Curr Sex Health Rep. (2017) 9:116–20. doi: 10.1007/s11930-017-0111-8

Crossref Full Text | Google Scholar

2. Kronk CA, Everhart AR, Ashley F, Thompson HM, Schall TE, Goetz TG, et al. Transgender data collection in the electronic health record: current concepts and issues. J Am Med Inform Assoc. (2022) 29(2):271–84. doi: 10.1093/jamia/ocab136

PubMed Abstract | Crossref Full Text | Google Scholar

3. Miyagi M, Guthman EM, Sun SED. Transgender rights rely on inclusive language. Science. (2021) 374(6575):1568–9. doi: 10.1126/science.abn3759

PubMed Abstract | Crossref Full Text | Google Scholar

4. Price-Feeney M, Green AE, Dorison SH. Suicidality among youth who are questioning, unsure of, or exploring their sexual identity. J Sex Res. (2021) 58(5):581–8. doi: 10.1080/00224499.2020.1832184

PubMed Abstract | Crossref Full Text | Google Scholar

5. Bartley EJ, Fillingim RB. Sex differences in pain: a brief review of clinical and experimental findings. Br J Anaesth. (2013) 111(1):52–8. doi: 10.1093/bja/aet127

PubMed Abstract | Crossref Full Text | Google Scholar

6. Winter S, Diamond M, Green J, Karasic D, Reed T, Whittle S, et al. Transgender people: health at the margins of society. Lancet. (2016) 388(10042):390–400. doi: 10.1016/S0140-6736(16)00683-8

PubMed Abstract | Crossref Full Text | Google Scholar

7. Price-Feeney M, Green AE, Dorison S. Understanding the mental health of transgender and nonbinary youth. J Adolesc Health. (2020) 66(6):684–90. doi: 10.1016/j.jadohealth.2019.11.314

PubMed Abstract | Crossref Full Text | Google Scholar

8. James SEHJ, Rankin S, Keisling M, Mottet L, Anafi M. The Report of the 2015 U.S. Transgender Survey. Washington, D.C.: National Center for Transgender Equality (2016).

Google Scholar

9. DeChants JP, Price MN, Green AE, Davis CK, Pick CJ. Association of updating identification documents with suicidal ideation and attempts among transgender and nonbinary youth. Int J Environ Res Public Health. (2022) 19(9):5016. doi: 10.3390/ijerph19095016

PubMed Abstract | Crossref Full Text | Google Scholar

10. Turban JL, Ehrensaft D. Research review: gender identity in youth: treatment paradigms and controversies. J Child Psychol Psychiatry. (2018) 59(12):1228–43. doi: 10.1111/jcpp.12833

PubMed Abstract | Crossref Full Text | Google Scholar

11. Mollet GA, Harrison DW. Emotion and pain: a functional cerebral systems integration. Neuropsychol Rev. (2006) 16(3):99–121. doi: 10.1007/s11065-006-9009-3

PubMed Abstract | Crossref Full Text | Google Scholar

12. Panisch LS, Rogers RG, Breen MT, Nutt S, Dahud S, Salazar CA. Childhood betrayal trauma, dissociation, and shame impact health-related quality of life among individuals with chronic pelvic pain. Child Abuse Negl. (2022) 131:105744. doi: 10.1016/j.chiabu.2022.105744

PubMed Abstract | Crossref Full Text | Google Scholar

13. Hu H, Liu W, Liu Y, Pan J, Zheng X. Association of depressive symptoms and risk of knee pain: the moderating effect of sex. BMC Musculoskelet Disord. (2021) 22(1):640. doi: 10.1186/s12891-021-04511-2

PubMed Abstract | Crossref Full Text | Google Scholar

14. Seligowski AV, Webber TK, Marvar PJ, Ressler KJ, Philip NS. Involvement of the brain-heart axis in the link between PTSD and cardiovascular disease. Depress Anxiety. (2022) 39:663–74. doi: 10.1002/da.23271

PubMed Abstract | Crossref Full Text | Google Scholar

15. Craft RM, Mogil JS, Aloisi AM. Sex differences in pain and analgesia: the role of gonadal hormones. Eur J Pain. (2004) 8(5):397–411. doi: 10.1016/j.ejpain.2004.01.003

PubMed Abstract | Crossref Full Text | Google Scholar

16. Gregus AM, Levine IS, Eddinger KA, Yaksh TL, Buczynski MW. Sex differences in neuroimmune and glial mechanisms of pain. Pain. (2021) 162(8):2186–200. doi: 10.1097/j.pain.0000000000002215

PubMed Abstract | Crossref Full Text | Google Scholar

17. Institute of Medicine. The Health of Lesbian, Gay, Bisexual, and Transgender People: Building a Foundation for Better Understanding. Washington, DC: National Academies Press (2011).

Google Scholar

18. Institute of Medicine. Relieving Pain in America: A Blueprint for Transforming Prevention, Care, Education, and Research. Washington, DC: The National Academies Press (2011).

Google Scholar

19. Roberts TK, Fantz CR. Barriers to quality health care for the transgender population. Clin Biochem. (2014) 47(10–11):983–7. doi: 10.1016/j.clinbiochem.2014.02.009

PubMed Abstract | Crossref Full Text | Google Scholar

20. Lau F, Antonio M, Davison K, Queen R, Devor A. A rapid review of gender, sex, and sexual orientation documentation in electronic health records. J Am Med Inform Assoc. (2020) 27(11):1774–83. doi: 10.1093/jamia/ocaa158

PubMed Abstract | Crossref Full Text | Google Scholar

21. World Professional Association for Transgender Health. Standards of Care for the Health of Transsexual, Transgender, and Gender Nonconforming People [7th version]. (2012).

Google Scholar

22. Dakić T. New perspectives on transgender health in the forthcoming 11. Psychiatr Danub. (2020) 32(2):145–50. doi: 10.24869/psyd.2020.145

Crossref Full Text | Google Scholar

23. WHO releases new International Classification of Diseases (ICD 11) [press release]. (2018).

Google Scholar

24. Ipsos. LGBT+ PRIDE 2021 GLOBAL SURVEY: A 27-country Ipsos survey. (2021). p. 1–39.

Google Scholar

25. Baker KaH M. Sexual Orientation and Gender Identity Data Collection in the Behavioral Risk Factor Surveillance System Washington, DC: Center for American Progress; 2016. Available online at: https://www.americanprogress.org/article/sexual-orientation-and-gender-identity-data-collection-in-the-behavioral-risk-factor-surveillance-system/ (Accessed October 10, 2021).

Google Scholar

26. Goodman M, Adams N, Corneil T, Kreukels B, Motmans J, Coleman E. Size and distribution of transgender and gender nonconforming populations: a narrative review. Endocrinol Metab Clin N Am. (2019) 48(2):303–21. doi: 10.1016/j.ecl.2019.01.001

Crossref Full Text | Google Scholar

27. Meerwijk EL, Sevelius JM. Transgender population size in the United States: a meta-regression of population-based probability samples. Am J Public Health. (2017) 107(2):e1–8. doi: 10.2105/AJPH.2016.303578

PubMed Abstract | Crossref Full Text | Google Scholar

28. Herman JL FA, Brown TN, Wilson B, Conron KJ. Age of Individuals Who Identify as Transgender in the United States. Los Angeles: The Williams Institute, University of California (2017).

Google Scholar

29. Flores AR, Meyer IH, Langton L, Herman JL. Gender identity disparities in criminal victimization: national crime victimization survey, 2017–2018. Am J Public Health. (2021) 111(4):726–9. doi: 10.2105/AJPH.2020.306099

PubMed Abstract | Crossref Full Text | Google Scholar

30. Lett E, Everhart A. Considerations for transgender population health research based on US national surveys. Ann Epidemiol. (2022) 65:65–71. doi: 10.1016/j.annepidem.2021.10.009

PubMed Abstract | Crossref Full Text | Google Scholar

31. Johns MM, Lowry R, Andrzejewski J, Barrios LC, Demissie Z, McManus T, et al. Transgender identity and experiences of violence victimization, substance use, suicide risk, and sexual risk behaviors among high school students—19 states and large urban school districts, 2017. MMWR Morb Mortal Wkly Rep. (2019) 68(3):67–71. doi: 10.15585/mmwr.mm6803a3

PubMed Abstract | Crossref Full Text | Google Scholar

32. Lagos D, Compton D. Evaluating the use of a two-step gender identity measure in the 2018 general social survey. Demography. (2021) 58(2):763–72. doi: 10.1215/00703370-8976151

PubMed Abstract | Crossref Full Text | Google Scholar

33. Centers for Disease Control and Prevention. HIV Infection, Risk, Prevention, and Testing Behaviors Among Transgender Women—National HIV Behavioral Surveillance 7 U.S. Cities, 2019–2020. HIV Surveillance Special Report 27; (2021).

Google Scholar

34. Bosse JD, Leblanc RG, Jackman K, Bjarnadottir RI. Benefits of implementing and improving collection of sexual orientation and gender identity data in electronic health records. Comput Inform Nurs. (2018) 36(6):267–74. doi: 10.1097/CIN.0000000000000417

PubMed Abstract | Crossref Full Text | Google Scholar

35. Abboud C, Duveau A, Bouali-Benazzouz R, Massé K, Mattar J, Brochoire L, et al. Animal models of pain: diversity and benefits. J Neurosci Methods. (2021) 348:108997. doi: 10.1016/j.jneumeth.2020.108997

PubMed Abstract | Crossref Full Text | Google Scholar

36. Grant JM, M L, Tanis J. Injustice at Every Turn: A Report of the National Transgender Discrimination Survey. Washington: National Center for Transgender Equality and National Gay and Lesbian Task Force; (2011)).

Google Scholar

37. Mogil JS. Animal models of pain: progress and challenges. Nat Rev Neurosci. (2009) 10(4):283–94. doi: 10.1038/nrn2606

PubMed Abstract | Crossref Full Text | Google Scholar

38. Collin L, Reisner SL, Tangpricha V, Goodman M. Prevalence of transgender depends on the “case” definition: a systematic review. J Sex Med. (2016) 13(4):613–26. doi: 10.1016/j.jsxm.2016.02.001

PubMed Abstract | Crossref Full Text | Google Scholar

39. Blosnich JR, Cashy J, Gordon AJ, Shipherd JC, Kauth MR, Brown GR, et al. Using clinician text notes in electronic medical record data to validate transgender-related diagnosis codes. J Am Med Inform Assoc. (2018) 25(7):905–8. doi: 10.1093/jamia/ocy022

PubMed Abstract | Crossref Full Text | Google Scholar

40. Berkley KJ. Sex differences in pain. Behav Brain Sci. (1997) 20(3):371–80.; discussion 435-513. doi: 10.1017/S0140525X97221485

PubMed Abstract | Crossref Full Text | Google Scholar

41. Saylor D, Steiner TJ. The global burden of headache. Semin Neurol. (2018) 38(2):182–90. doi: 10.1055/s-0038-1647245

PubMed Abstract | Crossref Full Text | Google Scholar

42. Rollman GB, Lautenbacher S. Sex differences in musculoskeletal pain. Clin J Pain. (2001) 17(1):20–4. doi: 10.1097/00002508-200103000-00004

PubMed Abstract | Crossref Full Text | Google Scholar

43. Fernandes G, Arruda MA, Bigal ME, Camparis CM, Gonçalves DAG. Painful temporomandibular disorder is associated with migraine in adolescents: a case-control study. J Pain. (2019) 20(10):1155–63. doi: 10.1016/j.jpain.2019.03.010

PubMed Abstract | Crossref Full Text | Google Scholar

44. Bueno CH, Pereira DD, Pattussi MP, Grossi PK, Grossi ML. Gender differences in temporomandibular disorders in adult populational studies: a systematic review and meta-analysis. J Oral Rehabil. (2018) 45(9):720–9. doi: 10.1111/joor.12661

PubMed Abstract | Crossref Full Text | Google Scholar

45. Martínez-Lavín M. Fibromyalgia in women: somatisation or stress-evoked, sex-dimorphic neuropathic pain? Clin Exp Rheumatol. (2021) 39(2):422–5. doi: 10.55563/clinexprheumatol/0c7d6v

Crossref Full Text | Google Scholar

46. Marcus DA. Fibromyalgia: diagnosis and treatment options. Gend Med. (2009) 6(Suppl 2):139–51. doi: 10.1016/j.genm.2009.01.004

PubMed Abstract | Crossref Full Text | Google Scholar

47. Kim JR, Kim HA. Molecular mechanisms of sex-related differences in arthritis and associated pain. Int J Mol Sci. (2020) 21(21):7938. doi: 10.3390/ijms21217938

Crossref Full Text | Google Scholar

48. Tarannum S, Leung Y-Y, Johnson SR, Widdifield J, Strand V, Rochon P, et al. Sex- and gender-related differences in psoriatic arthritis. Nat Rev Rheumatol. (2022) 18(9):513–26. doi: 10.1038/s41584-022-00810-7

PubMed Abstract | Crossref Full Text | Google Scholar

49. Ellerkamp V, Rall KK, Schaefer J, Brucker S, Fuchs J. Techniques of primary vaginoplasty in young adults with differences of sex development and female identification. J Clin Med. (2022) 11(13):3688. doi: 10.3390/jcm11133688

PubMed Abstract | Crossref Full Text | Google Scholar

50. Soret P, Le Dantec C, Desvaux E, Foulquier N, Chassagnol B, Hubert S, et al. A new molecular classification to drive precision treatment strategies in primary Sjögren’s syndrome. Nat Commun. (2021) 12(1):3523. doi: 10.1038/s41467-021-23472-7

PubMed Abstract | Crossref Full Text | Google Scholar

51. Chronic Pain Research Alliance. Impact of Chronic Overlapping Pain Conditions on Public Health and the Urgent Need for Safe and Effective Treatment: 2015 Analysis and policy recommendations. (2015). p. 1–47.

Google Scholar

52. Wei DY, Yuan Ong JJ, Goadsby PJ. Cluster headache: epidemiology, pathophysiology, clinical features, and diagnosis. Ann Indian Acad Neurol. (2018) 21(Suppl 1):S3–8. doi: 10.4103/aian.AIAN_349_17

PubMed Abstract | Crossref Full Text | Google Scholar

53. Levit D, Yaish I, Shtrozberg S, Aloush V, Greenman Y, Ablin JN. Pain and transition: evaluating fibromyalgia in transgender individuals. Clin Exp Rheumatol. (2021) 39 Suppl 130(3):27–32. doi: 10.55563/clinexprheumatol/pq0qp6

PubMed Abstract | Crossref Full Text | Google Scholar

54. Motmans J, Meier P, Ponnet K, T’Sjoen G. Female and male transgender quality of life: socioeconomic and medical differences. J Sex Med. (2012) 9(3):743–50. doi: 10.1111/j.1743-6109.2011.02569.x

PubMed Abstract | Crossref Full Text | Google Scholar

55. Aloisi AM, Bachiocco V, Costantino A, Stefani R, Ceccarelli I, Bertaccini A, et al. Cross-sex hormone administration changes pain in transsexual women and men. Pain. (2007) 132(Suppl 1):S60–S7. doi: 10.1016/j.pain.2007.02.006

PubMed Abstract | Crossref Full Text | Google Scholar

56. Strath LJ, Sorge RE, Owens MA, Gonzalez CE, Okunbor JI, White DM, et al. Sex and gender are not the same: why identity is important for people living with HIV and chronic pain. J Pain Res. (2020) 13:829–35. doi: 10.2147/JPR.S248424

PubMed Abstract | Crossref Full Text | Google Scholar

57. Pringsheim T, Gooren L. Migraine prevalence in male to female transsexuals on hormone therapy. Neurology. (2004) 63(3):593. doi: 10.1212/01.WNL.0000130338.62037.CC

PubMed Abstract | Crossref Full Text | Google Scholar

58. Weitzer D. How to provide effective pain management to LGBTQ individuals. Pract Pain Manag. (2019) 19(6). Available online at: https://www.medcentral.com/pain/how-provide-effective-pain-management-lgbtq-individuals (Accessed April 4, 2021).

Google Scholar

59. Strangio C. Understanding trans pain: Medium; (201). Available online at: https://chase-strangio.medium.com/understanding-trans-pain-aa41a91548d4 (Accessed October 10, 2021).

Google Scholar

60. Goetsch MF, Ribbink PJA. Penetrative genital pain in transgender men using testosterone: a survey study. Am J Obstet Gynecol. (2022) 226(2):264–70. doi: 10.1016/j.ajog.2021.09.019

PubMed Abstract | Crossref Full Text | Google Scholar

61. Rosendale N, Wong JO, Flatt JD, Whitaker E. Sexual and gender minority health in neurology: a scoping review. JAMA Neurol. (2021) 78(6):747–54. doi: 10.1001/jamaneurol.2020.5536

PubMed Abstract | Crossref Full Text | Google Scholar

62. Coleman E, Bockting W, Botzer M, Cohen-Kettenis P, DeCuypere G, Feldman J, et al. Standards of care for the health of transsexual, transgender, and gender-nonconforming people, version 7. Int J Transgend. (2012) 13(4):165–232. doi: 10.1080/15532739.2011.700873

Crossref Full Text | Google Scholar

63. Feldman J, Safer J. Hormone therapy in adults: suggested revisions to the sixth version of the standards of Care. Int J Transgend. (2009) 11(3):146–82. doi: 10.1080/15532730903383757

Crossref Full Text | Google Scholar

64. Hranilovich JA, Kaiser EA, Pace A, Barber M, Ziplow J. Headache in transgender and gender-diverse patients: a narrative review. Headache. (2021) 61(7):1040–50. doi: 10.1111/head.14171

PubMed Abstract | Crossref Full Text | Google Scholar

65. MacGregor EA, van den Brink AM. Transgender and migraine. In: Massen van den Brink A, MacGregor E, editors. Gender and Migraine Headache. Switzerland: Springer Nature (2019). p. 113–27.

Google Scholar

66. Markle JG, Fish EN. SeXX matters in immunity. Trends Immunol. (2014) 35(3):97–104. doi: 10.1016/j.it.2013.10.006

PubMed Abstract | Crossref Full Text | Google Scholar

67. Salgado E, Romera-Baurés M, Beltran-Catalan E, Naredo E, Carreira PE, Garcia-Vivar M, et al. Inmune-mediated inflammatory rheumatic diseases in transgender people: a scoping review. Semin Arthritis Rheum. (2022) 52(151920). doi: 10.1016/j.semarthrit.2021.10.004

Crossref Full Text | Google Scholar

68. Dance A. Why the sexes don't feel pain the same way. Nature. (2019) 567(7749):448–50. doi: 10.1038/d41586-019-00895-3

PubMed Abstract | Crossref Full Text | Google Scholar

69. Yu S, Chen C, Pan Y, Kurz MC, Datner E, Hendry PL, et al. Genes known to escape X chromosome inactivation predict co-morbid chronic musculoskeletal pain and posttraumatic stress symptom development in women following trauma exposure. Am J Med Genet B Neuropsychiatr Genet. (2019) 180(6):415–27. doi: 10.1002/ajmg.b.32706

PubMed Abstract | Crossref Full Text | Google Scholar

70. Roque RA, O'Reilly-Shah V, Lorello GR, Adams T, Cladis F. Transgender patient care: a prospective survey of pediatric anesthesiologist attitudes and knowledge. Can J Anaesth. (2021) 68(11):1723–6. doi: 10.1007/s12630-021-02089-w

PubMed Abstract | Crossref Full Text | Google Scholar

71. Roque RA. Transgender pediatric surgical patients-important perioperative considerations. Paediatr Anaesth. (2020) 30(5):520–8. doi: 10.1111/pan.13845

PubMed Abstract | Crossref Full Text | Google Scholar

72. Sayeem M, Carter B, Phulwani P, Zempsky WT. Gender dysphoria and chronic pain in youth. Pediatrics. (2021) 148(4). doi: 10.1542/peds.2021-050128

PubMed Abstract | Crossref Full Text | Google Scholar

73. Coleman E, Radix AE, Bouman WP, Brown GR, de Vries ALC, Deutsch MB, et al. Standards of care for the health of transgender and gender diverse people, version 8. Int J Transgend Health. (2022) 23(Suppl 1):S1–S259. doi: 10.1080/26895269.2022.2100644

PubMed Abstract | Crossref Full Text | Google Scholar

74. Chaya BF, Berman ZP, Boczar D, Trilles J, Siringo NV, Diep GK, et al. Gender affirmation surgery on the rise: analysis of trends and outcomes. LGBT Health. (2022) 9(8):582–8. doi: 10.1089/lgbt.2021.0224

Crossref Full Text | Google Scholar

75. Morrison SD, Vyas KS, Motakef S, Gast KM, Chung MT, Rashidi V, et al. Facial feminization: systematic review of the literature. Plast Reconstr Surg. (2016) 137(6):1759–70. doi: 10.1097/PRS.0000000000002171

PubMed Abstract | Crossref Full Text | Google Scholar

76. Tirrell AR, Abu El Hawa AA, Bekeny JC, Chang BL, Del Corral G. Facial feminization surgery: a systematic review of perioperative surgical planning and outcomes. Plast Reconstr Surg Glob Open. (2022) 10(3):e4210. doi: 10.1097/GOX.0000000000004210

PubMed Abstract | Crossref Full Text | Google Scholar

77. Uhlman K, Gormley J, Churchill I, Huynh M, Leveille CF, McRae MH, et al. Outcomes in facial feminization surgery: a systematic review. Facial Plast Surg Aesthet Med. (2022). doi: 10.1089/fpsam.2021.0293

PubMed Abstract | Crossref Full Text | Google Scholar

78. Oles N, Darrach H, Landford W, Garza M, Twose C, Park CS, et al. Gender affirming surgery: a comprehensive, systematic review of all peer-reviewed literature and methods of assessing patient-centered outcomes (part 1: breast/chest, face, and voice). Ann Surg. (2022) 275(1):e52–66. doi: 10.1097/SLA.0000000000004728

PubMed Abstract | Crossref Full Text | Google Scholar

79. Salim A, Poh M. Gender-affirming penile inversion vaginoplasty. Clin Plast Surg. (2018) 45(3):343–50. doi: 10.1016/j.cps.2018.04.001

PubMed Abstract | Crossref Full Text | Google Scholar

80. Moisés da Silva GV, Lobato MIR, Silva DC, Schwarz K, Fontanari AMV, Costa AB, et al. Male-to-female gender-affirming surgery: 20-year review of technique and surgical results. Front Surg. (2021) 8:639430. doi: 10.3389/fsurg.2021.639430

Crossref Full Text | Google Scholar

81. Hadj-Moussa M, Agarwal S, Ohl DA, Kuzon WM. Masculinizing genital gender confirmation surgery. Sex Med Rev. (2019) 7(1):141–55. doi: 10.1016/j.sxmr.2018.06.004

PubMed Abstract | Crossref Full Text | Google Scholar

82. Al-Tamimi M, Pigot GL, Elfering L, Özer M, de Haseth K, van de Grift TC, et al. Genital gender-affirming surgery in transgender men in The Netherlands from 1989 to 2018: the evolution of surgical care. Plast Reconstr Surg. (2020) 145(1):153e–61e. doi: 10.1097/PRS.0000000000006385

PubMed Abstract | Crossref Full Text | Google Scholar

83. Oles N, Darrach H, Landford W, Garza M, Twose C, Park CS, et al. Gender affirming surgery: a comprehensive, systematic review of all peer-reviewed literature and methods of assessing patient-centered outcomes (part 2: genital reconstruction). Ann Surg. (2022) 275(1):e67–74. doi: 10.1097/SLA.0000000000004717

PubMed Abstract | Crossref Full Text | Google Scholar

84. Dubin S, Lane M, Morrison S, Radix A, Belkind U, Vercler C, et al. Medically assisted gender affirmation: when children and parents disagree. J Med Ethics. (2020) 46(5):295–9. doi: 10.1136/medethics-2019-105567

PubMed Abstract | Crossref Full Text | Google Scholar

85. Boskey ER, Johnson JA, Harrison C, Marron JM, Abecassis L, Scobie-Carroll A, et al. Ethical issues considered when establishing a pediatrics gender surgery center. Pediatrics. (2019) 143(6):e20183053. doi: 10.1542/peds.2018-3053

PubMed Abstract | Crossref Full Text | Google Scholar

86. Kraemer B, Delsignore A, Schnyder U, Hepp U. Body image and transsexualism. Psychopathology. (2008) 41(2):96–100. doi: 10.1159/000111554

PubMed Abstract | Crossref Full Text | Google Scholar

87. Owen-Smith AA, Gerth J, Sineath RC, Barzilay J, Becerra-Culqui TA, Getahun D, et al. Association between gender confirmation treatments and perceived gender congruence, body image satisfaction, and mental health in a cohort of transgender individuals. J Sex Med. (2018) 15(4):591–600. doi: 10.1016/j.jsxm.2018.01.017

PubMed Abstract | Crossref Full Text | Google Scholar

88. Lee MS, Almazan AN, Nambudiri VE, Keuroghlian AS. Association between gender-affirming hair removal and mental health outcomes. JAMA Dermatol. (2021) 157(9):1120–2. doi: 10.1001/jamadermatol.2021.2551

PubMed Abstract | Crossref Full Text | Google Scholar

89. Bradford NJ, Rider GN, Spencer KG. Hair removal and psychological well-being in transfeminine adults: associations with gender dysphoria and gender euphoria. J Dermatolog Treat. (2021) 32(6):635–42. doi: 10.1080/09546634.2019.1687823

PubMed Abstract | Crossref Full Text | Google Scholar

90. Nolan IT, Kuhner CJ, Dy GW. Demographic and temporal trends in transgender identities and gender confirming surgery. Transl Androl Urol. (2019) 8(3):184–90. doi: 10.21037/tau.2019.04.09

PubMed Abstract | Crossref Full Text | Google Scholar

91. Olson-Kennedy J, Warus J, Okonta V, Belzer M, Clark LF. Chest reconstruction and chest dysphoria in transmasculine minors and young adults: comparisons of nonsurgical and postsurgical cohorts. JAMA Pediatr. (2018) 172(5):431–6. doi: 10.1001/jamapediatrics.2017.5440

PubMed Abstract | Crossref Full Text | Google Scholar

92. American Society of Plastic Surgeons. Plastic Surgery Statistics Report. (2020).

Google Scholar

93. Williams M, Jezior J. Management of combat-related urological trauma in the modern era. Nat Rev Urol. (2013) 10(9):504–12. doi: 10.1038/nrurol.2013.148

PubMed Abstract | Crossref Full Text | Google Scholar

94. Goldman C, Shaw N, du Plessis D, Myers JB, van der Merwe A, Venkatesan K. Gunshot wounds to the penis and scrotum: a narrative review of management in civilian and military settings. Transl Androl Urol. (2021) 10(6):2596–608. doi: 10.21037/tau-20-1175

PubMed Abstract | Crossref Full Text | Google Scholar

95. Peña A, Levitt MA, Hong A, Midulla P. Surgical management of cloacal malformations: a review of 339 patients. J Pediatr Surg. (2004) 39(3):470–9.; discussion -9. doi: 10.1016/j.jpedsurg.2003.11.033

Crossref Full Text | Google Scholar

96. Wernick JA, Busa S, Matouk K, Nicholson J, Janssen A. A systematic review of the psychological benefits of gender-affirming surgery. Urol Clin North Am. (2019) 46(4):475–86. doi: 10.1016/j.ucl.2019.07.002

PubMed Abstract | Crossref Full Text | Google Scholar

97. Jerome RR, Randhawa MK, Kowalczyk J, Sinclair A, Monga I. Sexual satisfaction after gender affirmation surgery in transgender individuals. Cureus. (2022) 14(7):e27365. doi: 10.7759/cureus.27365

PubMed Abstract | Crossref Full Text | Google Scholar

98. Park RH, Liu YT, Samuel A, Gurganus M, Gampper TJ, Corbett ST, et al. Long-term outcomes after gender-affirming surgery: 40-year follow-up study. Ann Plast Surg. (2022) 89(4):431–6. doi: 10.1097/SAP.0000000000003233

PubMed Abstract | Crossref Full Text | Google Scholar

99. Eftekhar Ardebili M, Janani L, Khazaei Z, Moradi Y, Baradaran HR. Quality of life in people with transsexuality after surgery: a systematic review and meta-analysis. Health Qual Life Outcomes. (2020) 18(1):264. doi: 10.1186/s12955-020-01510-0

PubMed Abstract | Crossref Full Text | Google Scholar

100. Passos TS, Teixeira MS, Almeida-Santos MA. Quality of life after gender affirmation surgery: a systematic review and network meta-analysis. Sex Res Soc Policy. (2020) 17(2):252–62. doi: 10.1007/s13178-019-00394-0

Crossref Full Text | Google Scholar

101. Javier C, Crimston CR, Barlow FK. Surgical satisfaction and quality of life outcomes reported by transgender men and women at least one year post gender-affirming surgery: a systematic literature review. Int J Transgend Health. (2022) 23(3):255–73. doi: 10.1080/26895269.2022.2038334

PubMed Abstract | Crossref Full Text | Google Scholar

102. Dhejne C, Lichtenstein P, Boman M, Johansson ALV, Långström N, Landén M. Long-term follow-up of transsexual persons undergoing sex reassignment surgery: cohort study in Sweden. PLoS One. (2011) 6(2):e16885. doi: 10.1371/journal.pone.0016885

PubMed Abstract | Crossref Full Text | Google Scholar

103. Morrison SD, Perez MG, Carter CK, Crane CN. Pre- and post-operative care with associated intra-operative techniques for phalloplasty in female-to-male patients. Urol Nurs. (2015) 35(3):134–8. doi: 10.7257/1053-816X.2015.35.3.134

PubMed Abstract | Crossref Full Text | Google Scholar

104. Remington AC, Morrison SD, Massie JP, Crowe CS, Shakir A, Wilson SC, et al. Outcomes after phalloplasty: do transgender patients and multiple urethral procedures carry a higher rate of complication? Plast Reconstr Surg. (2018) 141(2):220e–9e. doi: 10.1097/PRS.0000000000004061

PubMed Abstract | Crossref Full Text | Google Scholar

105. Horbach SE, Bouman MB, Smit JM, Özer M, Buncamper ME, Mullender MG. Outcome of vaginoplasty in male-to-female transgenders: a systematic review of surgical techniques. J Sex Med. (2015) 12(6):1499–512. doi: 10.1111/jsm.12868

PubMed Abstract | Crossref Full Text | Google Scholar

106. Bustos SS, Bustos VP, Mascaro A, Ciudad P, Forte AJ, Del Corral G, et al. Complications and patient-reported outcomes in transfemale vaginoplasty: an updated systematic review and meta-analysis. Plast Reconstr Surg Glob Open. (2021) 9(3):e3510. doi: 10.1097/GOX.0000000000003510

PubMed Abstract | Crossref Full Text | Google Scholar

107. Haben CM. Masculinization laryngoplasty. Otolaryngol Clin North Am. (2022) 55(4):757–65. doi: 10.1016/j.otc.2022.04.011

PubMed Abstract | Crossref Full Text | Google Scholar

108. Thomas JP. Feminization laryngoplasty—a comprehensive approach to reducing the size of the larynx and pharynx. Otolaryngol Clin North Am. (2022) 55(4):739–48. doi: 10.1016/j.otc.2022.05.002

PubMed Abstract | Crossref Full Text | Google Scholar

109. De Cuypere G, Elaut E, Heylens G, Van Maele G, Selvaggi G, T’Sjoen G, et al. Long-term follow-up: psychosocial outcome of Belgian transsexuals after sex reassignment surgery. Sexologies. (2006) 15(2):126–33. doi: 10.1016/j.sexol.2006.04.002

Crossref Full Text | Google Scholar

110. Wierckx K, Van Caenegem E, Elaut E, Dedecker D, Van de Peer F, Toye K, et al. Quality of life and sexual health after sex reassignment surgery in transsexual men. J Sex Med. (2011) 8(12):3379–88. doi: 10.1111/j.1743-6109.2011.02348.x

PubMed Abstract | Crossref Full Text | Google Scholar

111. Lindqvist EK, Sigurjonsson H, Möllermark C, Rinder J, Farnebo F, Lundgren TK. Quality of life improves early after gender reassignment surgery in transgender women. Eur J Plast Surg. (2017) 40(3):223–6. doi: 10.1007/s00238-016-1252-0

PubMed Abstract | Crossref Full Text | Google Scholar

112. Gong Y, Tan Q, Qin Q, Wei C. Prevalence of postmastectomy pain syndrome and associated risk factors: a large single-institution cohort study. Medicine (Baltimore). (2020) 99(20):e19834. doi: 10.1097/MD.0000000000019834

PubMed Abstract | Crossref Full Text | Google Scholar

113. Ahmed A, Bhatnagar S, Rana SP, Ahmad SM, Joshi S, Mishra S. Prevalence of phantom breast pain and sensation among postmastectomy patients suffering from breast cancer: a prospective study. Pain Pract. (2014) 14(2):E17–28. doi: 10.1111/papr.12089

PubMed Abstract | Crossref Full Text | Google Scholar

114. Rogowsky LC, Illmann CF, Isaac KV. Chronic pain in breast cancer patients post mastectomy with alloplastic reconstruction: a scoping review. Eur J Cancer Care (Engl). (2022) 31(4):e13631. doi: 10.1111/ecc.13631

PubMed Abstract | Crossref Full Text | Google Scholar

115. Jung BF, Ahrendt GM, Oaklander AL, Dworkin RH. Neuropathic pain following breast cancer surgery: proposed classification and research update. Pain. (2003) 104(1-2):1–13. doi: 10.1016/S0304-3959(03)00241-0

PubMed Abstract | Crossref Full Text | Google Scholar

116. Pogatzki-Zahn E, Kutschar P, Nestler N, Osterbrink J. A prospective multicentre study to improve postoperative pain: identification of potentialities and problems. PLoS One. (2015) 10(11):e0143508. doi: 10.1371/journal.pone.0143508

PubMed Abstract | Crossref Full Text | Google Scholar

117. Miaskowski C, Paul SM, Cooper B, West C, Levine JD, Elboim C, et al. Identification of patient subgroups and risk factors for persistent arm/shoulder pain following breast cancer surgery. Eur J Oncol Nurs. (2014) 18(3):242–53. doi: 10.1016/j.ejon.2013.12.002

PubMed Abstract | Crossref Full Text | Google Scholar

118. Verdecchia N, Grunwaldt L, Visoiu M. Pain outcomes following mastectomy or bilateral breast reduction for transgender and nontransgender patients who received pectoralis nerve blocks. Paediatr Anaesth. (2020) 30(9):1049–50. doi: 10.1111/pan.13969

PubMed Abstract | Crossref Full Text | Google Scholar

119. Robinson KA, Duncan S, Austrie J, Fleishman A, Tobias A, Hopwood RA, et al. Opioid consumption after gender-affirming mastectomy and two other breast surgeries. J Surg Res. (2020) 251:33–7. doi: 10.1016/j.jss.2019.12.043

PubMed Abstract | Crossref Full Text | Google Scholar

120. Lang CL, Day DL, Klit A, Mejdahl MK, Holmgaard R. Low risk of persistent pain, sensory disturbances, and complications following mastectomy after gender-affirming surgery. Transgend Health. (2021) 6(4):188–93. doi: 10.1089/trgh.2020.0070

PubMed Abstract | Crossref Full Text | Google Scholar

121. Tran BNN, Epstein S, Singhal D, Lee BT, Tobias AM, Ganor O. Gender affirmation surgery: a synopsis using American College of surgeons national surgery quality improvement program and national inpatient sample databases. Ann Plast Surg. (2018) 80(4):S229–35. doi: 10.1097/SAP.0000000000001350

Crossref Full Text | Google Scholar

122. MacKinnon KR, Grober E, Krakowsky Y. Lost in transition: addressing the absence of quality surgical outcomes data in gender-affirming surgeries. Can Urol Assoc J. (2020) 14(6):157–8. doi: 10.5489/cuaj.6610

PubMed Abstract | Crossref Full Text | Google Scholar

123. Potter E, Sivagurunathan M, Armstrong K, Barker LC, Du Mont J, Lorello GR, et al. Patient reported symptoms and adverse outcomes seen in Canada’s first vaginoplasty postoperative care clinic. Neurourol Urodyn. (2023) 42(2):523–9. doi: 10.1002/nau.25132

PubMed Abstract | Crossref Full Text | Google Scholar

124. Ding C, Khondker A, Goldenberg MG, Kwong JCC, Lajkosz K, Potter E, et al. Urinary complications after penile inversion vaginoplasty in transgender women: systematic review and meta-analysis. Can Urol Assoc J. (2022) 17(4):121–8. doi: 10.5489/cuaj.8108

Crossref Full Text | Google Scholar

125. Li VY, Demzik A, Snyder L, Ogunleye AA, Wang A, Figler BD. Genital gender affirming surgery. Am Surg. (2022) 88(12):2817–22. doi: 10.1177/00031348221109479

PubMed Abstract | Crossref Full Text | Google Scholar

126. Calotta NA, Kuzon W, Dellon AL, Monstrey S, Coon D. Sensibility, sensation, and nerve regeneration after reconstructive genital surgery: evolving concepts in neurobiology. Plast Reconstr Surg. (2021) 147(6):995e–1003e. doi: 10.1097/PRS.0000000000007969

PubMed Abstract | Crossref Full Text | Google Scholar

127. Belfer I, Schreiber KL, Shaffer JR, Shnol H, Blaney K, Morando A, et al. Persistent postmastectomy pain in breast cancer survivors: analysis of clinical, demographic, and psychosocial factors. J Pain. (2013) 14(10):1185–95. doi: 10.1016/j.jpain.2013.05.002

PubMed Abstract | Crossref Full Text | Google Scholar

128. Waddington H, Wilkes R. The effects of intercostal nerve blocks on post-operative pain outcomes following mastectomy and wide local excision. Int J Surg. (2018) 55:S18. doi: 10.1016/j.ijsu.2018.05.059

Crossref Full Text | Google Scholar

129. Agarwal RR, Wallace AM, Madison SJ, Morgan AC, Mascha EJ, Ilfeld BM. Single-injection thoracic paravertebral block and postoperative analgesia after mastectomy: a retrospective cohort study. J Clin Anesth. (2015) 27(5):371–4. doi: 10.1016/j.jclinane.2015.04.003

PubMed Abstract | Crossref Full Text | Google Scholar

130. Hembree WC, Cohen-Kettenis PT, Gooren L, Hannema SE, Meyer WJ, Murad MH, et al. Endocrine treatment of gender-dysphoric/gender-incongruent persons: an endocrine society clinical practice guideline. J Clin Endocrinol Metab. (2017) 102(11):3869–903. doi: 10.1210/jc.2017-01658

PubMed Abstract | Crossref Full Text | Google Scholar

131. Becasen JS, Denard CL, Mullins MM, Higa DH, Sipe TA. Estimating the prevalence of HIV and sexual behaviors among the US transgender population: a systematic review and meta-analysis, 2006–2017. Am J Public Health. (2019) 109(1):e1–8. doi: 10.2105/AJPH.2018.304727

PubMed Abstract | Crossref Full Text | Google Scholar

132. Kuhns LM, Reisner SL, Mimiaga MJ, Gayles T, Shelendich M, Garofalo R. Correlates of PrEP indication in a multi-site cohort of young HIV-uninfected transgender women. AIDS Behav. (2016) 20(7):1470–7. doi: 10.1007/s10461-015-1182-z

PubMed Abstract | Crossref Full Text | Google Scholar

133. Kuper LE, Nussbaum R, Mustanski B. Exploring the diversity of gender and sexual orientation identities in an online sample of transgender individuals. J Sex Res. (2012) 49(2-3):244–54. doi: 10.1080/00224499.2011.596954

PubMed Abstract | Crossref Full Text | Google Scholar

134. Chan KJ, Jolly D, Liang JJ, Weinand JD, Safer JD. Estrogen levels do not rise with testosterone treatment for transgender men. Endocr Pract. (2018) 24(4):329–33. doi: 10.4158/EP-2017-0203

PubMed Abstract | Crossref Full Text | Google Scholar

135. Iwamoto SJ, Defreyne J, Rothman MS, Van Schuylenbergh J, Van de Bruaene L, Motmans J, et al. Health considerations for transgender women and remaining unknowns: a narrative review. Ther Adv Endocrinol Metab. (2019) 10:2042018819871166. doi: 10.1177/2042018819871166

PubMed Abstract | Crossref Full Text | Google Scholar

136. Iwamoto SJ, T’Sjoen G, Safer JD, Davidge-Pitts CJ, Wierman ME, Glodowski MB, et al. Letter to the editor: “progesterone is important for transgender women’s therapy-applying evidence for the benefits of progesterone in ciswomen”. J Clin Endocrinol Metab. (2019) 104(8):3127–8. doi: 10.1210/jc.2019-00249

PubMed Abstract | Crossref Full Text | Google Scholar

137. Gooren LJ, Sungkaew T, Giltay EJ, Guadamuz TE. Cross-sex hormone use, functional health and mental well-being among transgender men (toms) and transgender women (kathoeys) in Thailand. Cult Health Sex. (2015) 17(1):92–103. doi: 10.1080/13691058.2014.950982

PubMed Abstract | Crossref Full Text | Google Scholar

138. Grimstad FW, Boskey E, Grey M. New-onset abdominopelvic pain after initiation of testosterone therapy among trans-masculine persons: a community-based exploratory survey. LGBT Health. (2020) 7(5):248–53. doi: 10.1089/lgbt.2019.0258

PubMed Abstract | Crossref Full Text | Google Scholar

139. Vedovo F, Di Blas L, Aretusi F, Falcone M, Perin C, Pavan N, et al. Physical, mental and sexual health among transgender women: a comparative study among operated transgender and cisgender women in a national tertiary referral network. J Sex Med. (2021) 18(5):982–9. doi: 10.1016/j.jsxm.2021.02.006

PubMed Abstract | Crossref Full Text | Google Scholar

140. Ailani J. Updates on management of headache in women and transgender women. Curr Opin Neurol. (2021) 34(3):339–43. doi: 10.1097/WCO.0000000000000926

PubMed Abstract | Crossref Full Text | Google Scholar

141. National Institure of Diabetes and Digestive and Kidney Diseases. Livertox: Clinical and Research Information in Drug-Induced Liver Injury. Bethesda, MD: National Institute of Health (2012).

Google Scholar

142. Sachedina A, Todd N. Dysmenorrhea, endometriosis and chronic pelvic pain in adolescents. J Clin Res Pediatr Endocrinol. (2020) 12(Suppl 1):7–17. doi: 10.4274/jcrpe.galenos.2019.2019.S0217

PubMed Abstract | Crossref Full Text | Google Scholar

143. Dlugi AM, Miller JD, Knittle J. Lupron depot (leuprolide acetate for depot suspension) in the treatment of endometriosis: a randomized, placebo-controlled, double-blind study. Lupron study group. Fertil Steril. (1990) 54(3):419–27. doi: 10.1016/S0015-0282(16)53755-8

PubMed Abstract | Crossref Full Text | Google Scholar

144. Baum M, Hackshaw A, Houghton J, Rutqvist , Fornander T, Nordenskjold B, et al. Adjuvant goserelin in pre-menopausal patients with early breast cancer: results from the ZIPP study. Eur J Cancer. (2006) 42(7):895–904. doi: 10.1016/j.ejca.2005.12.013

PubMed Abstract | Crossref Full Text | Google Scholar

145. Freystaetter G, Fischer K, Orav EJ, Egli A, Theiler R, Münzer T, et al. Total serum testosterone and western Ontario and McMaster universities osteoarthritis index pain and function among older men and women with severe knee osteoarthritis. Arthritis Care Res (Hoboken). (2020) 72(11):1511–8. doi: 10.1002/acr.24074

PubMed Abstract | Crossref Full Text | Google Scholar

146. Bartley EJ, Palit S, Kuhn BL, Kerr KL, Terry EL, DelVentura JL, et al. Natural variation in testosterone is associated with hypoalgesia in healthy women. Clin J Pain. (2015) 31(8):730–9. doi: 10.1097/AJP.0000000000000153

PubMed Abstract | Crossref Full Text | Google Scholar

147. Evans SF, Kwok Y, Solterbeck A, Pyragius C, Hull ML, Hutchinson MR, et al. The relationship between androgens and days per month of period pain, pelvic pain, headache, and TLR4 responsiveness of peripheral blood mononuclear cells in young women with dysmenorrhoea. J Pain Res. (2021) 14:585–99. doi: 10.2147/JPR.S279253

PubMed Abstract | Crossref Full Text | Google Scholar

148. Aloisi AM, Ceccarelli I, Fiorenzani P, De Padova AM, Massafra C. Testosterone affects formalin-induced responses differently in male and female rats. Neurosci Lett. (2004) 361(1-3):262–4. doi: 10.1016/j.neulet.2003.12.023

PubMed Abstract | Crossref Full Text | Google Scholar

149. Aloisi AM, Affaitati G, Ceccarelli I, Fiorenzani P, Lerza R, Rossi C, et al. Estradiol and testosterone differently affect visceral pain-related behavioural responses in male and female rats. Eur J Pain. (2010) 14(6):602–7. doi: 10.1016/j.ejpain.2009.10.016

PubMed Abstract | Crossref Full Text | Google Scholar

150. Blair JA, McGee H, Bhatta S, Palm R, Casadesus G. Hypothalamic-pituitary-gonadal axis involvement in learning and memory and Alzheimer’s disease: more than “just” estrogen. Front Endocrinol (Lausanne). (2015) 6:45. doi: 10.3389/fendo.2015.00045

PubMed Abstract | Crossref Full Text | Google Scholar

151. Traish A, Bolanos J, Nair S, Saad F, Morgentaler A. Do androgens modulate the pathophysiological pathways of inflammation? Appraising the contemporary evidence. J Clin Med. (2018) 7(12):549. doi: 10.3390/jcm7120549

PubMed Abstract | Crossref Full Text | Google Scholar

152. Zwickl S, Burchill L, Wong AFQ, Leemaqz SY, Cook T, Angus LM, et al. Pelvic pain in transgender people using testosterone therapy. LGBT Health. (2023) 10(3):179–90. doi: 10.1089/lgbt.2022.0187

PubMed Abstract | Crossref Full Text | Google Scholar

153. Johansen JA, Breedlove SM, Jordan CL. Androgen receptor expression in the levator ani muscle of male mice. J Neuroendocrinol. (2007) 19(10):823–6. doi: 10.1111/j.1365-2826.2007.01592.x

PubMed Abstract | Crossref Full Text | Google Scholar

154. Maurer AJ, Lissounov A, Knezevic I, Candido KD, Knezevic NN. Pain and sex hormones: a review of current understanding. Pain Manag. (2016) 6(3):285–96. doi: 10.2217/pmt-2015-0002

PubMed Abstract | Crossref Full Text | Google Scholar

155. Hassan S, Muere A, Einstein G. Ovarian hormones and chronic pain: a comprehensive review. Pain. (2014) 155(12):2448–60. doi: 10.1016/j.pain.2014.08.027

PubMed Abstract | Crossref Full Text | Google Scholar

156. Iacovides S, Avidon I, Baker FC. Does pain vary across the menstrual cycle? A review. Eur J Pain. (2015) 19(10):1389–405. doi: 10.1002/ejp.714

PubMed Abstract | Crossref Full Text | Google Scholar

157. Ceccarelli I, Fiorenzani P, Grasso G, Lariviere WR, Massafra C, Massai L, et al. Estrogen and mu-opioid receptor antagonists counteract the 17 beta-estradiol-induced licking increase and interferon-gamma reduction occurring during the formalin test in male rats. Pain. (2004) 111(1-2):181–90. doi: 10.1016/j.pain.2004.06.005

PubMed Abstract | Crossref Full Text | Google Scholar

158. Craft RM. Modulation of pain by estrogens. Pain. (2007) 132(Suppl 1):S3–S12. doi: 10.1016/j.pain.2007.09.028

PubMed Abstract | Crossref Full Text | Google Scholar

159. Weiser MJ, Foradori CD, Handa RJ. Estrogen receptor beta in the brain: from form to function. Brain Res Rev. (2008) 57(2):309–20. doi: 10.1016/j.brainresrev.2007.05.013

PubMed Abstract | Crossref Full Text | Google Scholar

160. Unger CA. Hormone therapy for transgender patients. Transl Androl Urol. (2016) 5(6):877–84. doi: 10.21037/tau.2016.09.04

PubMed Abstract | Crossref Full Text | Google Scholar

161. Abdel-Salam OME, Baiuomy AR, Nada SA. Effect of spironolactone on pain responses in mice. EXCLI J. (2010) 9:46–57. doi: 10.1016/j.ejphar.2010.08.050

PubMed Abstract | Crossref Full Text | Google Scholar

162. Jaggi AS, Singh N. Differential effect of spironolactone in chronic constriction injury and vincristine-induced neuropathic pain in rats. Eur J Pharmacol. (2010) 648(1-3):102–9. doi: 10.1016/j.ejphar.2010.08.050

PubMed Abstract | Crossref Full Text | Google Scholar

163. Böhm R, Westermann P, Gleim M, Cascorbi I, Gruenewald M, Herdegen T, et al. High-dose spironolactone lacks effectiveness in treatment of fibromyalgia (RCT). Eur J Pain. (2021) 25(8):1739–50. doi: 10.1002/ejp.1784

Crossref Full Text | Google Scholar

164. Mcmurdo ME, Sumukadas D, Donnan PT, Cvoro V, Rauchhaus P, Argo I, et al. Spironolactone for people age 70 years and older with osteoarthritic knee pain: a proof-of-concept trial. Arthritis Care Res (Hoboken). (2016) 68(5):716–21. doi: 10.1002/acr.22724

PubMed Abstract | Crossref Full Text | Google Scholar

165. Berman SM, Naliboff BD, Suyenobu B, Labus JS, Stains J, Bueller JA, et al. Sex differences in regional brain response to aversive pelvic visceral stimuli. Am J Physiol Regul Integr Comp Physiol. (2006) 291(2):R268–76. doi: 10.1152/ajpregu.00065.2006

PubMed Abstract | Crossref Full Text | Google Scholar

166. Kern MK, Jaradeh S, Arndorfer RC, Jesmanowicz A, Hyde J, Shaker R. Gender differences in cortical representation of rectal distension in healthy humans. Am J Physiol Gastrointest Liver Physiol. (2001) 281(6):G1512–23. doi: 10.1152/ajpgi.2001.281.6.G1512

PubMed Abstract | Crossref Full Text | Google Scholar

167. Moulton EA, Keaser ML, Gullapalli RP, Maitra R, Greenspan JD. Sex differences in the cerebral BOLD signal response to painful heat stimuli. Am J Physiol Regul Integr Comp Physiol. (2006) 291(2):R257–67. doi: 10.1152/ajpregu.00084.2006

PubMed Abstract | Crossref Full Text | Google Scholar

168. Smith MD, Jones LS, Wilson MA. Sex differences in hippocampal slice excitability: role of testosterone. Neuroscience. (2002) 109(3):517–30. doi: 10.1016/S0306-4522(01)00490-0

PubMed Abstract | Crossref Full Text | Google Scholar

169. Amandusson Å, Blomqvist A. Estrogenic influences in pain processing. Front Neuroendocrinol. (2013) 34(4):329–49. doi: 10.1016/j.yfrne.2013.06.001

PubMed Abstract | Crossref Full Text | Google Scholar

170. Kawata M. Roles of steroid hormones and their receptors in structural organization in the nervous system. Neurosci Res. (1995) 24(1):1–46. doi: 10.1016/0168-0102(96)81278-8

PubMed Abstract | Crossref Full Text | Google Scholar

171. Marcus DA. Interrelationships of neurochemicals, estrogen, and recurring headache. Pain. (1995) 62(2):129–39. doi: 10.1016/0304-3959(95)00052-T

PubMed Abstract | Crossref Full Text | Google Scholar

172. Smith MJ, Adams LF, Schmidt PJ, Rubinow DR, Wassermann EM. Effects of ovarian hormones on human cortical excitability. Ann Neurol. (2002) 51(5):599–603. doi: 10.1002/ana.10180

PubMed Abstract | Crossref Full Text | Google Scholar

173. Vincent K, Warnaby C, Stagg CJ, Moore J, Kennedy S, Tracey I. Brain imaging reveals that engagement of descending inhibitory pain pathways in healthy women in a low endogenous estradiol state varies with testosterone. Pain. (2013) 154(4):515–24. doi: 10.1016/j.pain.2012.11.016

PubMed Abstract | Crossref Full Text | Google Scholar

174. Alvarez P, Bogen O, Levine JD. Role of nociceptor estrogen receptor GPR30 in a rat model of endometriosis pain. Pain. (2014) 155(12):2680–6. doi: 10.1016/j.pain.2014.09.035

PubMed Abstract | Crossref Full Text | Google Scholar

175. Fehrenbacher JC, Loverme J, Clarke W, Hargreaves KM, Piomelli D, Taylor BK. Rapid pain modulation with nuclear receptor ligands. Brain Res Rev. (2009) 60(1):114–24. doi: 10.1016/j.brainresrev.2008.12.019

PubMed Abstract | Crossref Full Text | Google Scholar

176. Lu YC, Chen CW, Wang SY, Wu FS. 17Beta-estradiol Mediates the sex difference in capsaicin-induced nociception in rats. J Pharmacol Exp Ther. (2009) 331(3):1104–10. doi: 10.1124/jpet.109.158402

PubMed Abstract | Crossref Full Text | Google Scholar

177. Jiang P, Kong Y, Zhang XB, Wang W, Liu CF, Xu TL. Glycine receptor in rat hippocampal and spinal cord neurons as a molecular target for rapid actions of 17-beta-estradiol. Mol Pain. (2009) 5:2. doi: 10.1186/1744-8069-5-2

PubMed Abstract | Crossref Full Text | Google Scholar

178. Lagrange AH, Wagner EJ, Rønnekleiv OK, Kelly MJ. Estrogen rapidly attenuates a GABAB response in hypothalamic neurons. Neuroendocrinology. (1996) 64(2):114–23. doi: 10.1159/000127106

PubMed Abstract | Crossref Full Text | Google Scholar

179. Lagrange AH, Ronnekleiv OK, Kelly MJ. Modulation of G protein-coupled receptors by an estrogen receptor that activates protein kinase A. Mol Pharmacol. (1997) 51(4):605–12. doi: 10.1124/mol.51.4.605

PubMed Abstract | Crossref Full Text | Google Scholar

180. Castelli MP, Fadda P, Casu A, Spano MS, Casti A, Fratta W, et al. Male and female rats differ in brain cannabinoid CB1 receptor density and function and in behavioural traits predisposing to drug addiction: effect of ovarian hormones. Curr Pharm Des. (2014) 20(13):2100–13. doi: 10.2174/13816128113199990430

PubMed Abstract | Crossref Full Text | Google Scholar

181. Kumar A, Storman EM, Liu NJ, Gintzler AR. Estrogens suppress spinal endomorphin 2 release in female rats in phase with the estrous cycle. Neuroendocrinology. (2015) 102(1-2):33–43. doi: 10.1159/000430817

PubMed Abstract | Crossref Full Text | Google Scholar

182. Kelly MJ, Lagrange AH, Wagner EJ, Rønnekleiv OK. Rapid effects of estrogen to modulate G protein-coupled receptors via activation of protein kinase A and protein kinase C pathways. Steroids. (1999) 64(1-2):64–75. doi: 10.1016/S0039-128X(98)00095-6

PubMed Abstract | Crossref Full Text | Google Scholar

183. Craft RM, Ulibarri C. Sexual differentiation of rat reproductive versus opioid antinociceptive systems. Gend Med. (2009) 6(Suppl 2):208–24. doi: 10.1016/j.genm.2009.03.003

PubMed Abstract | Crossref Full Text | Google Scholar

184. Craft RM, Ulibarri C, Leitl MD, Sumner JE. Dose- and time-dependent estradiol modulation of morphine antinociception in adult female rats. Eur J Pain. (2008) 12(4):472–9. doi: 10.1016/j.ejpain.2007.07.014

PubMed Abstract | Crossref Full Text | Google Scholar

185. Stoffel EC, Ulibarri CM, Folk JE, Rice KC, Craft RM. Gonadal hormone modulation of mu, kappa, and delta opioid antinociception in male and female rats. J Pain. (2005) 6(4):261–74. doi: 10.1016/j.jpain.2004.12.006

PubMed Abstract | Crossref Full Text | Google Scholar

186. Loyd DR, Wang X, Murphy AZ. Sex differences in micro-opioid receptor expression in the rat midbrain periaqueductal gray are essential for eliciting sex differences in morphine analgesia. J Neurosci. (2008) 28(52):14007–17. doi: 10.1523/JNEUROSCI.4123-08.2008

PubMed Abstract | Crossref Full Text | Google Scholar

187. Jokinen V, Lilius T, Laitila J, Niemi M, Kambur O, Kalso E, et al. Do diuretics have antinociceptive actions: studies of spironolactone, eplerenone, furosemide and chlorothiazide, individually and with oxycodone and morphine. Basic Clin Pharmacol Toxicol. (2017) 120(1):38–45. doi: 10.1111/bcpt.12634

PubMed Abstract | Crossref Full Text | Google Scholar

188. Lilius TO, Jokinen V, Neuvonen MS, Väänänen AJ, Niemi M, Rauhala PV, et al. The mineralocorticoid receptor antagonist spironolactone enhances morphine antinociception. Eur J Pain. (2014) 18(3):386–95. doi: 10.1002/j.1532-2149.2013.00371.x

PubMed Abstract | Crossref Full Text | Google Scholar

189. Loyd DR, Murphy AZ. The role of the periaqueductal gray in the modulation of pain in males and females: are the anatomy and physiology really that different? Neural Plast. (2009) 2009:462879. doi: 10.1155/2009/462879

PubMed Abstract | Crossref Full Text | Google Scholar

190. Kelly MJ, Qiu J, Rønnekleiv OK. Estrogen modulation of G-protein-coupled receptor activation of potassium channels in the central nervous system. Ann N Y Acad Sci. (2003) 1007:6–16. doi: 10.1196/annals.1286.001

PubMed Abstract | Crossref Full Text | Google Scholar

191. Eckersell CB, Popper P, Micevych PE. Estrogen-induced alteration of mu-opioid receptor immunoreactivity in the medial preoptic nucleus and medial amygdala. J Neurosci. (1998) 18(10):3967–76. doi: 10.1523/JNEUROSCI.18-10-03967.1998

PubMed Abstract | Crossref Full Text | Google Scholar

192. Liu CC, Veldhuijzen DS, Ohara S, Winberry J, Greenspan JD, Lenz FA. Spatial attention to thermal pain stimuli in subjects with visual spatial hemi-neglect: extinction, mislocalization and misidentification of stimulus modality. Pain. (2011) 152(3):498–506. doi: 10.1016/j.pain.2010.10.017

PubMed Abstract | Crossref Full Text | Google Scholar

193. Correale J, Arias M, Gilmore W. Steroid hormone regulation of cytokine secretion by proteolipid protein-specific CD4+ T cell clones isolated from multiple sclerosis patients and normal control subjects. J Immunol. (1998) 161(7):3365–74. doi: 10.4049/jimmunol.161.7.3365

PubMed Abstract | Crossref Full Text | Google Scholar

194. Doyle HH, Eidson LN, Sinkiewicz DM, Murphy AZ. Sex differences in microglia activity within the periaqueductal gray of the rat: a potential mechanism driving the dimorphic effects of morphine. J Neurosci. (2017) 37(12):3202–14. doi: 10.1523/JNEUROSCI.2906-16.2017

PubMed Abstract | Crossref Full Text | Google Scholar

195. Doyle HH, Murphy AZ. Sex-dependent influences of morphine and its metabolites on pain sensitivity in the rat. Physiol Behav. (2018) 187:32–41. doi: 10.1016/j.physbeh.2017.11.030

PubMed Abstract | Crossref Full Text | Google Scholar

196. Sorge RE, LaCroix-Fralish ML, Tuttle AH, Sotocinal SG, Austin JS, Ritchie J, et al. Spinal cord toll-like receptor 4 mediates inflammatory and neuropathic hypersensitivity in male but not female mice. J Neurosci. (2011) 31(43):15450–4. doi: 10.1523/JNEUROSCI.3859-11.2011

PubMed Abstract | Crossref Full Text | Google Scholar

197. Gabel F, Hovhannisyan V, Andry V, Goumon Y. Central metabolism as a potential origin of sex differences in morphine antinociception but not induction of antinociceptive tolerance in mice. Br J Pharmacol. (2022) 180(7):843–61. doi: 10.1111/bph.15792

PubMed Abstract | Crossref Full Text | Google Scholar

198. Knodell RG, Allen RC, Kyner WT. Effects of ethinyl estradiol on pharmacokinetics of meperidine and pentobarbital in the rat. J Pharmacol Exp Ther. (1982) 221(1):1–6. Available online at: https://jpet.aspetjournals.org/content/221/1/1.long (Accessed April 22, 2021).7062276

PubMed Abstract | Google Scholar

199. Li J, Xie M, Wang X, Ouyang X, Wan Y, Dong G, et al. Sex hormones regulate cerebral drug metabolism via brain miRNAs: down-regulation of brain CYP2D by androgens reduces the analgesic effects of tramadol. Br J Pharmacol. (2015) 172(19):4639–54. doi: 10.1111/bph.13206

PubMed Abstract | Crossref Full Text | Google Scholar

200. Soldin OP, Chung SH, Mattison DR. Sex differences in drug disposition. J Biomed Biotechnol. (2011) 2011:187103. doi: 10.1155/2011/187103

PubMed Abstract | Crossref Full Text | Google Scholar

201. South SM, Wright AW, Lau M, Mather LE, Smith MT. Sex-related differences in antinociception and tolerance development following chronic intravenous infusion of morphine in the rat: modulatory role of testosterone via morphine clearance. J Pharmacol Exp Ther. (2001) 297(1):446–57. Available online at: https://jpet.aspetjournals.org/content/297/1/446 (Accessed April 22, 2021).11259573

PubMed Abstract | Google Scholar

202. Mawhinney LJ, Mabourakh D, Lewis MC. Gender-specific differences in the central nervous system’s response to anesthesia. Transl Stroke Res. (2013) 4(4):462–75. doi: 10.1007/s12975-012-0229-y

PubMed Abstract | Crossref Full Text | Google Scholar

203. Fullerton EF, Karom MC, Streicher JM, Young LJ, Murphy AZ. Age-induced changes in μ-opioid receptor signaling in the midbrain periaqueductal gray of male and female rats. J Neurosci. (2022) 42(32):6232–42. doi: 10.1523/JNEUROSCI.0355-22.2022

PubMed Abstract | Crossref Full Text | Google Scholar

204. Turban JL, King D, Carswell JM, Keuroghlian AS. Pubertal suppression for transgender youth and risk of suicidal ideation. Pediatrics. (2020) 145(2). doi: 10.1542/peds.2019-1725

PubMed Abstract | Crossref Full Text | Google Scholar

205. Klein DA, Emerick JE, Sylvester JE, Vogt KS. Disorders of puberty: an approach to diagnosis and management. Am Fam Physician. (2017) 96(9):590–9. Available online at: https://www.aafp.org/pubs/afp/issues/2017/1101/p590.html (Accessed April 22, 2021).29094880

PubMed Abstract | Google Scholar

206. Magiakou MA, Manousaki D, Papadaki M, Hadjidakis D, Levidou G, Vakaki M, et al. The efficacy and safety of gonadotropin-releasing hormone analog treatment in childhood and adolescence: a single center, long-term follow-up study. J Clin Endocrinol Metab. (2010) 95(1):109–17. doi: 10.1210/jc.2009-0793

PubMed Abstract | Crossref Full Text | Google Scholar

207. Gallagher JS, Missmer SA, Hornstein MD, Laufer MR, Gordon CM, DiVasta AD. Long-term effects of gonadotropin-releasing hormone agonists and add-back in adolescent endometriosis. J Pediatr Adolesc Gynecol. (2018) 31(4):376–81. doi: 10.1016/j.jpag.2018.03.004

PubMed Abstract | Crossref Full Text | Google Scholar

208. Lopez CM, Solomon D, Boulware SD, Christison-Lagay ER. Trends in the use of puberty blockers among transgender children in the United States. J Pediatr Endocrinol Metab. (2018) 31(6):665–70. doi: 10.1515/jpem-2018-0048

PubMed Abstract | Crossref Full Text | Google Scholar

209. Zaliznyak M, Bresee C, Garcia MM. Age at first experience of gender dysphoria among transgender adults seeking gender-affirming surgery. JAMA Netw Open. (2020) 3(3):e201236. doi: 10.1001/jamanetworkopen.2020.1236

PubMed Abstract | Crossref Full Text | Google Scholar

210. T’Sjoen G, Arcelus J, De Vries ALC, Fisher AD, Nieder TO, Özer M, et al. European Society for sexual medicine position statement “assessment and hormonal management in adolescent and adult trans people, with attention for sexual function and satisfaction”. J Sex Med. (2020) 17(4):570–84. doi: 10.1016/j.jsxm.2020.01.012

Crossref Full Text | Google Scholar

211. Rew L, Young CC, Monge M, Bogucka R. Review: puberty blockers for transgender and gender diverse youth-a critical review of the literature. Child Adolesc Ment Health. (2021) 26(1):3–14. doi: 10.1111/camh.12437

PubMed Abstract | Crossref Full Text | Google Scholar

212. Hisle-Gorman E, Schvey NA, Adirim TA, Rayne AK, Susi A, Roberts TA, et al. Mental healthcare utilization of transgender youth before and after affirming treatment. J Sex Med. (2021) 18(8):1444–54. doi: 10.1016/j.jsxm.2021.05.014

PubMed Abstract | Crossref Full Text | Google Scholar

213. de Vries AL, McGuire JK, Steensma TD, Wagenaar EC, Doreleijers TA, Cohen-Kettenis PT. Young adult psychological outcome after puberty suppression and gender reassignment. Pediatrics. (2014) 134(4):696–704. doi: 10.1542/peds.2013-2958

PubMed Abstract | Crossref Full Text | Google Scholar

214. Costa R, Dunsford M, Skagerberg E, Holt V, Carmichael P, Colizzi M. Psychological support, puberty suppression, and psychosocial functioning in adolescents with gender dysphoria. J Sex Med. (2015) 12(11):2206–14. doi: 10.1111/jsm.13034

PubMed Abstract | Crossref Full Text | Google Scholar

215. Wiepjes CM, Nota NM, de Blok CJM, Klaver M, de Vries ALC, Wensing-Kruger SA, et al. The Amsterdam cohort of gender dysphoria study (1972–2015): trends in prevalence, treatment, and regrets. J Sex Med. (2018) 15(4):582–90. doi: 10.1016/j.jsxm.2018.01.016

PubMed Abstract | Crossref Full Text | Google Scholar

216. Skinner DC, Albertson AJ, Navratil A, Smith A, Mignot M, Talbott H, et al. Effects of gonadotrophin-releasing hormone outside the hypothalamic-pituitary-reproductive axis. J Neuroendocrinol. (2009) 21(4):282–92. doi: 10.1111/j.1365-2826.2009.01842.x

PubMed Abstract | Crossref Full Text | Google Scholar

217. De Sanctis V, Soliman AT, Di Maio S, Soliman N, Elsedfy H. Long-term effects and significant adverse drug reactions (ADRs) associated with the use of gonadotropin-releasing hormone analogs (GnRHa) for central precocious puberty: a brief review of literature. Acta Biomed. (2019) 90(3):345–59. doi: 10.23750/abm.v90i3.8736

PubMed Abstract | Crossref Full Text | Google Scholar

218. Klink D, Caris M, Heijboer A, van Trotsenburg M, Rotteveel J. Bone mass in young adulthood following gonadotropin-releasing hormone analog treatment and cross-sex hormone treatment in adolescents with gender dysphoria. J Clin Endocrinol Metab. (2015) 100(2):E270–5. doi: 10.1210/jc.2014-2439

PubMed Abstract | Crossref Full Text | Google Scholar

219. De Pedro KT, Gilreath TD, Jackson C, Esqueda MC. Substance use among transgender students in California public middle and high schools. J Sch Health. (2017) 87(5):303–9. doi: 10.1111/josh.12499

PubMed Abstract | Crossref Full Text | Google Scholar

220. Klein SL, Flanagan KL. Sex differences in immune responses. Nat Rev Immunol. (2016) 16(10):626–38. doi: 10.1038/nri.2016.90

PubMed Abstract | Crossref Full Text | Google Scholar

221. Ziegler SM, Altfeld M. Human immunodeficiency virus 1 and type I interferons-where sex makes a difference. Front Immunol. (2017) 8:1224. doi: 10.3389/fimmu.2017.01224

PubMed Abstract | Crossref Full Text | Google Scholar

222. Scully EP, Haverfield J, Ursin RL, Tannenbaum C, Klein SL. Considering how biological sex impacts immune responses and COVID-19 outcomes. Nat Rev Immunol. (2020) 20(7):442–7. doi: 10.1038/s41577-020-0348-8

PubMed Abstract | Crossref Full Text | Google Scholar

223. Channappanavar R, Fett C, Mack M, Ten Eyck PP, Meyerholz DK, Perlman S. Sex-based differences in susceptibility to severe acute respiratory syndrome coronavirus infection. J Immunol. (2017) 198(10):4046–53. doi: 10.4049/jimmunol.1601896

PubMed Abstract | Crossref Full Text | Google Scholar

224. Cerri S, Mus L, Blandini F. Parkinson’s disease in women and men: what’s the difference? J Parkinsons Dis. (2019) 9(3):501–15. doi: 10.3233/JPD-191683

PubMed Abstract | Crossref Full Text | Google Scholar

225. Adams C, Kumar R. The effect of estrogen in a man with Parkinson’s disease and a review of its therapeutic potential. Int J Neurosci. (2013) 123(10):741–2. doi: 10.3109/00207454.2013.797903

PubMed Abstract | Crossref Full Text | Google Scholar

226. Chen Q, Zhang W, Sadana N, Chen X. Estrogen receptors in pain modulation: cellular signaling. Biol Sex Differ. (2021) 12(1):22. doi: 10.1186/s13293-021-00364-5

PubMed Abstract | Crossref Full Text | Google Scholar

227. Lee JY, Choi HY, Ju BG, Yune TY. Estrogen alleviates neuropathic pain induced after spinal cord injury by inhibiting microglia and astrocyte activation. Biochim Biophys Acta Mol Basis Dis. (2018) 1864(7):2472–80. doi: 10.1016/j.bbadis.2018.04.006

PubMed Abstract | Crossref Full Text | Google Scholar

228. Vacca V, Marinelli S, Pieroni L, Urbani A, Luvisetto S, Pavone F. 17beta-estradiol Counteracts neuropathic pain: a behavioural, immunohistochemical, and proteomic investigation on sex-related differences in mice. Sci Rep. (2016) 6:18980. doi: 10.1038/srep18980

PubMed Abstract | Crossref Full Text | Google Scholar

229. Loiola RA, Wickstead ES, Solito E, McArthur S. Estrogen promotes pro-resolving microglial behavior and phagocytic cell clearance through the actions of annexin A1. Front Endocrinol (Lausanne). (2019) 10:420. doi: 10.3389/fendo.2019.00420

PubMed Abstract | Crossref Full Text | Google Scholar

230. Levin ER. Extranuclear steroid receptors are essential for steroid hormone actions. Annu Rev Med. (2015) 66:271–80. doi: 10.1146/annurev-med-050913-021703

PubMed Abstract | Crossref Full Text | Google Scholar

231. Maglione A, Rolla S, Mercanti SF, Cutrupi S, Clerico M. The adaptive immune system in multiple sclerosis: an estrogen-mediated point of view. Cells. (2019) 8(10):1280. doi: 10.3390/cells8101280

PubMed Abstract | Crossref Full Text | Google Scholar

232. Hao S, Zhao J, Zhou J, Zhao S, Hu Y, Hou Y. Modulation of 17beta-estradiol on the number and cytotoxicity of NK cells in vivo related to MCM and activating receptors. Int Immunopharmacol. (2007) 7(13):1765–75. doi: 10.1016/j.intimp.2007.09.017

PubMed Abstract | Crossref Full Text | Google Scholar

233. Yovel G, Shakhar K, Ben-Eliyahu S. The effects of sex, menstrual cycle, and oral contraceptives on the number and activity of natural killer cells. Gynecol Oncol. (2001) 81(2):254–62. doi: 10.1006/gyno.2001.6153

PubMed Abstract | Crossref Full Text | Google Scholar

234. Stopińska-Głuszak U, Waligóra J, Grzela T, Głuszak M, Jóźwiak J, Radomski D, et al. Effect of estrogen/progesterone hormone replacement therapy on natural killer cell cytotoxicity and immunoregulatory cytokine release by peripheral blood mononuclear cells of postmenopausal women. J Reprod Immunol. (2006) 69(1):65–75. doi: 10.1016/j.jri.2005.07.006

Crossref Full Text | Google Scholar

235. Groer M, El-Badri N, Djeu J, Harrington M, Van Eepoel J. Suppression of natural killer cell cytotoxicity in postpartum women. Am J Reprod Immunol. (2010) 63(3):209–13. doi: 10.1111/j.1600-0897.2009.00788.x

PubMed Abstract | Crossref Full Text | Google Scholar

236. Davies AJ, Kim HW, Gonzalez-Cano R, Choi J, Back SK, Roh SE, et al. Natural killer cells degenerate intact sensory afferents following nerve injury. Cell. (2019) 176(4):716–28.e18. doi: 10.1016/j.cell.2018.12.022

PubMed Abstract | Crossref Full Text | Google Scholar

237. Davies AJ, Rinaldi S, Costigan M, Oh SB. Cytotoxic immunity in peripheral nerve injury and pain. Front Neurosci. (2020) 14:142. doi: 10.3389/fnins.2020.00142

PubMed Abstract | Crossref Full Text | Google Scholar

238. Das B, Dobrowolski C, Luttge B, Valadkhan S, Chomont N, Johnston R, et al. Estrogen receptor-1 is a key regulator of HIV-1 latency that imparts gender-specific restrictions on the latent reservoir. Proc Natl Acad Sci U S A. (2018) 115(33):E7795–E804. doi: 10.1073/pnas.1803468115

PubMed Abstract | Crossref Full Text | Google Scholar

239. Réu P, Khosravi A, Bernard S, Mold JE, Salehpour M, Alkass K, et al. The lifespan and turnover of microglia in the human brain. Cell Rep. (2017) 20(4):779–84. doi: 10.1016/j.celrep.2017.07.004

Crossref Full Text | Google Scholar

240. Lin CR, Rosendale N, Deeb W. Expanding sexual and gender minority research in movement disorders: more than awareness and acceptance. Parkinsonism Relat Disord. (2021) 87:162–5. doi: 10.1016/j.parkreldis.2021.05.019

PubMed Abstract | Crossref Full Text | Google Scholar

241. Bruno KA, Morales-Lara AC, Bittencourt EB, Siddiqui H, Bommarito G, Patel J, et al. Sex differences in comorbidities associated with Sjögren’s disease. Front Med (Lausanne). (2022) 9:958670. doi: 10.3389/fmed.2022.958670

PubMed Abstract | Crossref Full Text | Google Scholar

242. Goebel A, Andersson D, Helyes Z, Clark JD, Dulake D, Svensson C. The autoimmune aetiology of unexplained chronic pain. Autoimmun Rev. (2022) 21(3):103015. doi: 10.1016/j.autrev.2021.103015

PubMed Abstract | Crossref Full Text | Google Scholar

243. Eltumi HG, Tashani OA. Effect of age, sex and gender on pain sensitivity: a narrative review. Open Pain Journal. (2017) 10:44–55. doi: 10.2174/1876386301710010044

Crossref Full Text | Google Scholar

244. LaCroix-Fralish ML, Tawfik VL, DeLeo JA. The organizational and activational effects of sex hormones on tactile and thermal hypersensitivity following lumbar nerve root injury in male and female rats. Pain. (2005) 114(1-2):71–80. doi: 10.1016/j.pain.2004.12.006

PubMed Abstract | Crossref Full Text | Google Scholar

245. Gioiosa L, Chen X, Watkins R, Umeda EA, Arnold AP. Sex chromosome complement affects nociception and analgesia in newborn mice. J Pain. (2008) 9(10):962–9. doi: 10.1016/j.jpain.2008.06.001

PubMed Abstract | Crossref Full Text | Google Scholar

246. Hagiwara H, Funabashi T, Mitsushima D, Kimura F. Effects of neonatal testosterone treatment on sex differences in formalin-induced nociceptive behavior in rats. Neurosci Lett. (2007) 412(3):264–7. doi: 10.1016/j.neulet.2006.11.020

PubMed Abstract | Crossref Full Text | Google Scholar

247. Ellermeier W, Westphal W. Gender differences in pain ratings and pupil reactions to painful pressure stimuli. Pain. (1995) 61(3):435–9. doi: 10.1016/0304-3959(94)00203-Q

PubMed Abstract | Crossref Full Text | Google Scholar

248. Miclescu A, Essemark M, Astermark M, Gkatziani P, Straatmann A, Butler S, et al. Prolonged time of after-sensation after experimental pain stimuli despite efficient conditioned pain modulation in patients with chronic neuropathic pain after traumatic nerve injuries in upper extremity. Pain Rep. (2021) 6(1):e908. doi: 10.1097/PR9.0000000000000908

PubMed Abstract | Crossref Full Text | Google Scholar

249. Unruh AM. Gender variations in clinical pain experience. Pain. (1996) 65(2-3):123–67. doi: 10.1016/0304-3959(95)00214-6

PubMed Abstract | Crossref Full Text | Google Scholar

250. Casale R, Atzeni F, Bazzichi L, Beretta G, Costantini E, Sacerdote P, et al. Pain in women: a perspective review on a relevant clinical issue that deserves prioritization. Pain Ther. (2021) 10(1):287–314. doi: 10.1007/s40122-021-00244-1

PubMed Abstract | Crossref Full Text | Google Scholar

251. Mogil JS. Sex differences in pain and pain inhibition: multiple explanations of a controversial phenomenon. Nat Rev Neurosci. (2012) 13(12):859–66. doi: 10.1038/nrn3360

PubMed Abstract | Crossref Full Text | Google Scholar

252. Fish EN. The X-files in immunity: sex-based differences predispose immune responses. Nat Rev Immunol. (2008) 8(9):737–44. doi: 10.1038/nri2394

PubMed Abstract | Crossref Full Text | Google Scholar

253. Schurz H, Salie M, Tromp G, Hoal EG, Kinnear CJ, Möller M. The X chromosome and sex-specific effects in infectious disease susceptibility. Hum Genomics. (2019) 13(1):2. doi: 10.1186/s40246-018-0185-z

PubMed Abstract | Crossref Full Text | Google Scholar

254. Lefèvre N, Corazza F, Valsamis J, Delbaere A, De Maertelaer V, Duchateau J, et al. The number of X chromosomes influences inflammatory cytokine production following toll-like receptor stimulation. Front Immunol. (2019) 10:1052. doi: 10.3389/fimmu.2019.01052

Crossref Full Text | Google Scholar

255. Raznahan A, Parikshak NN, Chandran V, Blumenthal JD, Clasen LS, Alexander-Bloch AF, et al. Sex-chromosome dosage effects on gene expression in humans. Proc Natl Acad Sci U S A. (2018) 115(28):7398–403. doi: 10.1073/pnas.1802889115

PubMed Abstract | Crossref Full Text | Google Scholar

256. Meester I, Manilla-Muñoz E, León-Cachón RBR, Paniagua-Frausto GA, Carrión-Alvarez D, Ruiz-Rodríguez CO, et al. SeXY chromosomes and the immune system: reflections after a comparative study. Biol Sex Differ. (2020) 11(1):3. doi: 10.1186/s13293-019-0278-y

PubMed Abstract | Crossref Full Text | Google Scholar

257. Kohn LA, Kohn DB. Gene therapies for primary immune deficiencies. Front Immunol. (2021) 12:648951. doi: 10.3389/fimmu.2021.648951

PubMed Abstract | Crossref Full Text | Google Scholar

258. Syrett CM, Sindhava V, Hodawadekar S, Myles A, Liang G, Zhang Y, et al. Loss of Xist RNA from the inactive X during B cell development is restored in a dynamic YY1-dependent two-step process in activated B cells. PLoS Genet. (2017) 13(10):e1007050. doi: 10.1371/journal.pgen.1007050

PubMed Abstract | Crossref Full Text | Google Scholar

259. Syrett CM, Sindhava V, Sierra I, Dubin AH, Atchison M, Anguera MC. Diversity of epigenetic features of the inactive X-chromosome in NK cells, dendritic cells, and macrophages. Front Immunol. (2018) 9:3087. doi: 10.3389/fimmu.2018.03087

PubMed Abstract | Crossref Full Text | Google Scholar

260. Syrett CM, Paneru B, Sandoval-Heglund D, Wang J, Banerjee S, Sindhava V, et al. Altered X-chromosome inactivation in T cells may promote sex-biased autoimmune diseases. JCI Insight. (2019) 4(7):e126751. doi: 10.1172/jci.insight.126751

PubMed Abstract | Crossref Full Text | Google Scholar

261. Dinno A. Homicide rates of transgender individuals in the United States: 2010–2014. Am J Public Health. (2017) 107(9):1441–7. doi: 10.2105/AJPH.2017.303878

PubMed Abstract | Crossref Full Text | Google Scholar

262. Richman AD. Concurrent social disadvantages and chronic inflammation: the intersection of race and ethnicity, gender, and socioeconomic Status. J Racial Ethn Health Disparities. (2018) 5(4):787–97. doi: 10.1007/s40615-017-0424-3

PubMed Abstract | Crossref Full Text | Google Scholar

263. Diamond LM, Dehlin AJ, Alley J. Systemic inflammation as a driver of health disparities among sexually-diverse and gender-diverse individuals. Psychoneuroendocrinology. (2021) 129:105215. doi: 10.1016/j.psyneuen.2021.105215

PubMed Abstract | Crossref Full Text | Google Scholar

264. Steptoe A, Hamer M, Chida Y. The effects of acute psychological stress on circulating inflammatory factors in humans: a review and meta-analysis. Brain Behav Immun. (2007) 21(7):901–12. doi: 10.1016/j.bbi.2007.03.011

PubMed Abstract | Crossref Full Text | Google Scholar

265. Fuchs F, Damm J, Gerstberger R, Roth J, Rummel C. Activation of the inflammatory transcription factor nuclear factor interleukin-6 during inflammatory and psychological stress in the brain. J Neuroinflammation. (2013) 10:140. doi: 10.1186/1742-2094-10-140

PubMed Abstract | Crossref Full Text | Google Scholar

266. Kuebler U, Zuccarella-Hackl C, Arpagaus A, Wolf JM, Farahmand F, von Känel R, et al. Stress-induced modulation of NF-κB activation, inflammation-associated gene expression, and cytokine levels in blood of healthy men. Brain Behav Immun. (2015) 46:87–95. doi: 10.1016/j.bbi.2014.12.024

PubMed Abstract | Crossref Full Text | Google Scholar

267. Slavich GM, Cole SW. The emerging field of human social genomics. Clin Psychol Sci. (2013) 1(3):331–48. doi: 10.1177/2167702613478594

PubMed Abstract | Crossref Full Text | Google Scholar

268. Hendricks ML, Testa RJ. A conceptual framework for clinical work with transgender and gender nonconforming clients: an adaptation of the minority stress model. Prof Psychol Res Pr. (2012) 43:460–7. doi: 10.1037/a0029597

Crossref Full Text | Google Scholar

269. Holt-Lunstad J, Smith TB, Baker M, Harris T, Stephenson D. Loneliness and social isolation as risk factors for mortality: a meta-analytic review. Perspect Psychol Sci. (2015) 10(2):227–37. doi: 10.1177/1745691614568352

PubMed Abstract | Crossref Full Text | Google Scholar

270. DuBois LZ, Powers S, Everett BG, Juster RP. Stigma and diurnal cortisol among transitioning transgender men. Psychoneuroendocrinology. (2017) 82:59–66. doi: 10.1016/j.psyneuen.2017.05.008

PubMed Abstract | Crossref Full Text | Google Scholar

271. Schistad EI, Stubhaug A, Furberg AS, Engdahl BL, Nielsen CS. C-reactive protein and cold-pressor tolerance in the general population: the tromso study. Pain. (2017) 158(7):1280–8. doi: 10.1097/j.pain.0000000000000912

PubMed Abstract | Crossref Full Text | Google Scholar

272. Pace A, Barber M, Ziplow J, Hranilovich JA, Kaiser EA. Gender minority stress, psychiatric comorbidities, and the experience of migraine in transgender and gender-diverse individuals: a narrative review. Curr Pain Headache Rep. (2021) 25(12):82. doi: 10.1007/s11916-021-00996-7

PubMed Abstract | Crossref Full Text | Google Scholar

273. Dubois LZ. Associations between transition-specific stress experience, nocturnal decline in ambulatory blood pressure, and C-reactive protein levels among transgender men. Am J Hum Biol. (2012) 24(1):52–61. doi: 10.1002/ajhb.22203

PubMed Abstract | Crossref Full Text | Google Scholar

274. Ryu S, Fan L. The relationship between financial worries and psychological distress among U.S. Adults. J Fam Econ Issues. (2023) 44(1):16–33. doi: 10.1007/s10834-022-09820-9

PubMed Abstract | Crossref Full Text | Google Scholar

275. Barzilay R, Moore TM, Greenberg DM, DiDomenico GE, Brown LA, White LK, et al. Resilience, COVID-19-related stress, anxiety and depression during the pandemic in a large population enriched for healthcare providers. Transl Psychiatry. (2020) 10(1):291. doi: 10.1038/s41398-020-00982-4

PubMed Abstract | Crossref Full Text | Google Scholar

276. Coombs NC, Meriwether WE, Caringi J, Newcomer SR. Barriers to healthcare access among U.S. Adults with mental health challenges: a population-based study. SSM Popul Health. (2021) 15:100847. doi: 10.1016/j.ssmph.2021.100847

PubMed Abstract | Crossref Full Text | Google Scholar

277. Wolfson JA, Garcia T, Leung CW. Food insecurity is associated with depression, anxiety, and stress: evidence from the early days of the COVID-19 pandemic in the United States. Health Equity. (2021) 5(1):64–71. doi: 10.1089/heq.2020.0059

PubMed Abstract | Crossref Full Text | Google Scholar

278. Downing JM, Przedworski JM. Health of transgender adults in the U.S., 2014–2016. Am J Prev Med. (2018) 55(3):336–44. doi: 10.1016/j.amepre.2018.04.045

PubMed Abstract | Crossref Full Text | Google Scholar

279. Puckett JA, Matsuno E, Dyar C, Mustanski B, Newcomb ME. Mental health and resilience in transgender individuals: what type of support makes a difference? J Fam Psychol. (2019) 33(8):954–64. doi: 10.1037/fam0000561

PubMed Abstract | Crossref Full Text | Google Scholar

280. McQuillan MT, Kuhns LM, Miller AA, McDade T, Garofalo R. Gender minority stress, support, and inflammation in transgender and gender-nonconforming youth. Transgend Health. (2021) 6(2):91–100. doi: 10.1089/trgh.2020.0019

PubMed Abstract | Crossref Full Text | Google Scholar

281. Meanley S, Flores DD, Listerud L, Chang CJ, Feinstein BA, Watson RJ. The interplay of familial warmth and LGBTQ+ specific family rejection on LGBTQ+ adolescents’ self-esteem. J Adolesc. (2021) 93:40–52. doi: 10.1016/j.adolescence.2021.10.002

PubMed Abstract | Crossref Full Text | Google Scholar

282. Wood EP, Cook SH. Father support is protective against the negative effects of perceived discrimination on CRP among sexual minorities but not heterosexuals. Psychoneuroendocrinology. (2019) 110:104368. doi: 10.1016/j.psyneuen.2019.06.019

PubMed Abstract | Crossref Full Text | Google Scholar

283. Cooper M. Family Values: Between Neoliberalism And The New Social Conservatism. New York: Zone Books (2017).

Google Scholar

284. Movement Advancement Project. Where We Call Home: Transgender People in Rural America. (2019).

Google Scholar

285. Apkarian VA, Hashmi JA, Baliki MN. Pain and the brain: specificity and plasticity of the brain in clinical chronic pain. Pain. (2011) 152(3 Suppl):S49–64. doi: 10.1016/j.pain.2010.11.010

PubMed Abstract | Crossref Full Text | Google Scholar

286. Bushnell MC, Ceko M, Low LA. Cognitive and emotional control of pain and its disruption in chronic pain. Nat Rev Neurosci. (2013) 14(7):502–11. doi: 10.1038/nrn3516

PubMed Abstract | Crossref Full Text | Google Scholar

287. Villemure C, Bushnell MC. Mood influences supraspinal pain processing separately from attention. J Neurosci. (2009) 29(3):705–15. doi: 10.1523/JNEUROSCI.3822-08.2009

PubMed Abstract | Crossref Full Text | Google Scholar

288. Loggia ML, Schweinhardt P, Villemure C, Bushnell MC. Effects of psychological state on pain perception in the dental environment. J Can Dent Assoc. (2008) 74(7):651–6. Available online at: https://www.cda-adc.ca/jcda/vol-74/issue-7/651.html (Accessed May 1, 2021).18789200

PubMed Abstract | Google Scholar

289. Roy M, Peretz I, Rainville P. Emotional valence contributes to music-induced analgesia. Pain. (2008) 134(1-2):140–7. doi: 10.1016/j.pain.2007.04.003

PubMed Abstract | Crossref Full Text | Google Scholar

290. Casey CY, Greenberg MA, Nicassio PM, Harpin ER, Hubbard D. Transition from acute to chronic pain and disability: a model including cognitive, affective, and trauma factors. PAIN. (2008) 134(1):69–79. doi: 10.1016/j.pain.2007.03.032

PubMed Abstract | Crossref Full Text | Google Scholar

291. Apkarian AV, Baliki MN, Farmer MA. Predicting transition to chronic pain. Curr Opin Neurol. (2013) 26(4):360–7. doi: 10.1097/WCO.0b013e32836336ad

PubMed Abstract | Crossref Full Text | Google Scholar

292. Fisher AD, Ristori J, Castellini G, Sensi C, Cassioli E, Prunas A, et al. Psychological characteristics of Italian gender dysphoric adolescents: a case-control study. J Endocrinol Invest. (2017) 40(9):953–65. doi: 10.1007/s40618-017-0647-5

PubMed Abstract | Crossref Full Text | Google Scholar

293. George SZ, Dannecker EA, Robinson ME. Fear of pain, not pain catastrophizing, predicts acute pain intensity, but neither factor predicts tolerance or blood pressure reactivity: an experimental investigation in pain-free individuals. Eur J Pain. (2006) 10(5):457–65. doi: 10.1016/j.ejpain.2005.06.007

PubMed Abstract | Crossref Full Text | Google Scholar

294. Dersh J, Polatin PB, Gatchel RJ. Chronic pain and psychopathology: research findings and theoretical considerations. Psychosom Med. (2002) 64(5):773–86. doi: 10.1097/01.psy.0000024232.11538.54

PubMed Abstract | Crossref Full Text | Google Scholar

295. Progovac AM, Tran NM, Mullin BO, De Mello Libardi Maia J, Creedon TB, Dunham E, et al. Elevated rates of violence victimization and suicide attempt among transgender and gender diverse patients in an urban, safety net health system. World Med Health Policy. (2021) 13(2):176–98. doi: 10.1002/wmh3.403

Crossref Full Text | Google Scholar

296. Messinger AM, Guadalupe-Diaz XL, Kurdyla V. Transgender polyvictimization in the U.S. Transgender survey. J Interpers Violence. (2021) 37(19–20):NP18810–36. doi: 10.1177/08862605211039250

Crossref Full Text | Google Scholar

297. Safer JD, Coleman E, Feldman J, Garofalo R, Hembree W, Radix A, et al. Barriers to healthcare for transgender individuals. Curr Opin Endocrinol Diabetes Obes. (2016) 23(2):168–71. doi: 10.1097/MED.0000000000000227

PubMed Abstract | Crossref Full Text | Google Scholar

298. Bockting WO, Miner MH, Swinburne Romine RE, Hamilton A, Coleman E. Stigma, mental health, and resilience in an online sample of the US transgender population. Am J Public Health. (2013) 103(5):943–51. doi: 10.2105/AJPH.2013.301241

PubMed Abstract | Crossref Full Text | Google Scholar

299. Khobzi RN, BG R, Robb T, Anna T, Kyle S, Matthias K. Depression in male-to-female transgender ontarians: results from the trans PULSE project. Can J Community Ment Health. (2011) 30(2):113–33. doi: 10.7870/cjcmh-2011-0020

Crossref Full Text | Google Scholar

300. Reisner SL, White JM, Mayer KH, Mimiaga MJ. Sexual risk behaviors and psychosocial health concerns of female-to-male transgender men screening for STDs at an urban community health center. AIDS Care. (2014) 26(7):857–64. doi: 10.1080/09540121.2013.855701

PubMed Abstract | Crossref Full Text | Google Scholar

301. Wilchins R. The first national survey on transviolence. GenderPAC. (1997). Available online at: https://rinj.org/documents//research/transgender_violence.pdf (Accessed February 2, 2022).

Google Scholar

302. Hassan AN, Le Foll B, Imtiaz S, Rehm J. The effect of post-traumatic stress disorder on the risk of developing prescription opioid use disorder: results from the national epidemiologic survey on alcohol and related conditions III. Drug Alcohol Depend. (2017) 179:260–6. doi: 10.1016/j.drugalcdep.2017.07.012

PubMed Abstract | Crossref Full Text | Google Scholar

303. Barr SM, Snyder KE, Adelson JL, Budge SL. Posttraumatic stress in the trans community: the roles of anti-transgender bias, non-affirmation, and internalized transphobia. Psychol Sex Orientat Gend Divers. (2021) 9(4):410–21. doi: 10.1037/sgd0000500

Crossref Full Text | Google Scholar

304. Hughto JMW, Restar AJ, Wolfe HL, Gordon LK, Reisner SL, Biello KB, et al. Opioid pain medication misuse, concomitant substance misuse, and the unmet behavioral health treatment needs of transgender and gender diverse adults. Drug Alcohol Depend. (2021) 222:108674. doi: 10.1016/j.drugalcdep.2021.108674

PubMed Abstract | Crossref Full Text | Google Scholar

305. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders (DSM-5®). Washington, DC: American Psychiatric Pub (2013).

Google Scholar

306. Cohen-Kettenis PT, Pfäfflin F. The DSM diagnostic criteria for gender identity disorder in adolescents and adults. Arch Sex Behav. (2010) 39(2):499–513. doi: 10.1007/s10508-009-9562-y

PubMed Abstract | Crossref Full Text | Google Scholar

307. Becker I, Nieder TO, Cerwenka S, Briken P, Kreukels BP, Cohen-Kettenis PT, et al. Body image in young gender dysphoric adults: a European Multi-Center Study. Arch Sex Behav. (2016) 45(3):559–74. doi: 10.1007/s10508-015-0527-z

PubMed Abstract | Crossref Full Text | Google Scholar

308. Davis SA, Colton Meier S. Effects of testosterone treatment and chest reconstruction surgery on mental health and sexuality in female-to-male transgender people. Int J Sex Health. (2014) 26:113–28. doi: 10.1080/19317611.2013.833152

Crossref Full Text | Google Scholar

309. Dutton L, Koenig K, Fennie K. Gynecologic care of the female-to-male transgender man. J Midwifery Womens Health. (2008) 53(4):331–7. doi: 10.1016/j.jmwh.2008.02.003

PubMed Abstract | Crossref Full Text | Google Scholar

310. van de Grift TC, Cohen-Kettenis PT, Steensma TD, De Cuypere G, Richter-Appelt H, Haraldsen IR, et al. Body satisfaction and physical appearance in gender dysphoria. Arch Sex Behav. (2016) 45(3):575–85. doi: 10.1007/s10508-015-0614-1

PubMed Abstract | Crossref Full Text | Google Scholar

311. Pulice-Farrow L, Cusack CE, Galupo MP. “Certain parts of my body don’t belong to me”: trans individuals’ descriptions of body-specific gender dysphoria. Sex Res Soc Policy. (2019) 17:654–67. doi: 10.1007/s13178-019-00423-y

Crossref Full Text | Google Scholar

312. Walling DP, Goodwin JM, Cole CM. Dissociation in a transsexual population. J Sex Educ Ther. (1998) 23(2):121–3. doi: 10.1080/01614576.1998.11074218

Crossref Full Text | Google Scholar

313. Nierula B, Martini M, Matamala-Gomez M, Slater M, Sanchez-Vives MV. Seeing an embodied virtual hand is analgesic contingent on colocation. J Pain. (2017) 18(6):645–55. doi: 10.1016/j.jpain.2017.01.003

PubMed Abstract | Crossref Full Text | Google Scholar

314. Hegedüs G, Darnai G, Szolcsányi T, Feldmann Á, Janszky J, Kállai J. The rubber hand illusion increases heat pain threshold. Eur J Pain. (2014) 18(8):1173–81. doi: 10.1002/j.1532-2149.2014.00466.x

Crossref Full Text | Google Scholar

315. Pięta M, Rzeszutek M, Gasik R. Body image, pain and level of resources among arthritis patients: the moderating role of gender. Psychol Health Med. (2020) 25:802–11. doi: 10.1080/13548506.2019.1668566

Crossref Full Text | Google Scholar

316. Osumi M, Imai R, Ueta K, Nobusako S, Morioka S. Negative body image associated with changes in the visual body appearance increases pain perception. PLoS One. (2014) 9(9):e107376. doi: 10.1371/journal.pone.0107376

PubMed Abstract | Crossref Full Text | Google Scholar

317. Ramachandran VS, McGeoch PD. Phantom penises in transsexuals. J Conscious Stud. (2008) 15(1):5–16. Available online at: https://psycnet.apa.org/record/2008-15313-001 (Accessed February 2, 2022).

Google Scholar

318. Ramachandran VS, McGeoch PD. Occurrence of phantom genitalia after gender reassignment surgery. Med Hypotheses. (2007) 69(5):1001–3. doi: 10.1016/j.mehy.2007.02.024

PubMed Abstract | Crossref Full Text | Google Scholar

319. Straayer CRK, editor. Transformations Conference, Psychotherapy Center for Gender and Sexuality, Institute for Contemporary Psychology. New York: Institute for Contemporary Psychotherapy (2021).

Google Scholar

320. Straayer C. Phantom penis: extrapolating neuroscience and employing imagination for trans male sexual embodiment. Stud Gend Sex. (2021) 21(4):251–79. doi: 10.1080/15240657.2020.1842075

Crossref Full Text | Google Scholar

321. Gong G, He Y, Evans AC. Brain connectivity: gender makes a difference. Neuroscientist. (2011) 17(5):575–91. doi: 10.1177/1073858410386492

PubMed Abstract | Crossref Full Text | Google Scholar

322. Karkazis K. The misuses of “biological sex”. Lancet. (2019) 394(10212):1898–9. doi: 10.1016/S0140-6736(19)32764-3

PubMed Abstract | Crossref Full Text | Google Scholar

323. Ingalhalikar M, Smith A, Parker D, Satterthwaite TD, Elliott MA, Ruparel K, et al. Sex differences in the structural connectome of the human brain. Proc Natl Acad Sci U S A. (2014) 111(2):823–8. doi: 10.1073/pnas.1316909110

PubMed Abstract | Crossref Full Text | Google Scholar

324. Di Noto PM, Newman L, Wall S, Einstein G. The hermunculus: what is known about the representation of the female body in the brain? Cerebral Cortex. (2013) 23(5):1005–13. doi: 10.1093/cercor/bhs005

PubMed Abstract | Crossref Full Text | Google Scholar

325. Case L. Discussion of “phantom penis: extrapolating neuroscience and employing imagination for trans male embodiment”. Stud Gend Sex. (2021) 21(4):307–11. doi: 10.1080/15240657.2020.1842068

Crossref Full Text | Google Scholar

326. Ramachandran VS, Hirstein W. The perception of phantom limbs. The D. O. Hebb lecture. Brain. (1998) 121(Pt 9):1603–30. doi: 10.1093/brain/121.9.1603

PubMed Abstract | Crossref Full Text | Google Scholar

327. Case LK, Brang D, Landazuri R, Viswanathan P, Ramachandran VS. Altered white matter and sensory response to bodily sensation in female-to-male transgender individuals. Arch Sex Behav. (2017) 46(5):1223–37. doi: 10.1007/s10508-016-0850-z

PubMed Abstract | Crossref Full Text | Google Scholar

328. Manzouri A, Kosidou K, Savic I. Anatomical and functional findings in female-to-male transsexuals: testing a new hypothesis. Cereb Cortex. (2017) 27(2):998–1010. doi: 10.1093/cercor/bhv278

PubMed Abstract | Crossref Full Text | Google Scholar

329. Tsakiris M, Hesse MD, Boy C, Haggard P, Fink GR. Neural signatures of body ownership: a sensory network for bodily self-consciousness. Cereb Cortex. (2007) 17(10):2235–44. doi: 10.1093/cercor/bhl131

PubMed Abstract | Crossref Full Text | Google Scholar

330. Matuz-Budai T, Lábadi B, Kohn E, Matuz A, Zsidó AN, Inhóf O, et al. Individual differences in the experience of body ownership are related to cortical thickness. Sci Rep. (2022) 12(1):808. doi: 10.1038/s41598-021-04720-8

PubMed Abstract | Crossref Full Text | Google Scholar

331. Feusner JD, Lidström A, Moody TD, Dhejne C, Bookheimer SY, Savic I. Intrinsic network connectivity and own body perception in gender dysphoria. Brain Imaging Behav. (2017) 11(4):964–76. doi: 10.1007/s11682-016-9578-6

PubMed Abstract | Crossref Full Text | Google Scholar

332. Horga G, Kaur T, Peterson BS. Annual research review: current limitations and future directions in MRI studies of child- and adult-onset developmental psychopathologies. J Child Psychol Psychiatry. (2014) 55(6):659–80. doi: 10.1111/jcpp.12185

PubMed Abstract | Crossref Full Text | Google Scholar

333. Frigerio A, Ballerini L, Valdés Hernández M. Structural, functional, and metabolic brain differences as a function of gender identity or sexual orientation: a systematic review of the human neuroimaging literature. Arch Sex Behav. (2021) 50:3329–52. doi: 10.1007/s10508-021-02005-9

PubMed Abstract | Crossref Full Text | Google Scholar

334. Altinay M, Anand A. Neuroimaging gender dysphoria: a novel psychobiological model. Brain Imaging Behav. (2020) 14(4):1281–97. doi: 10.1007/s11682-019-00121-8

PubMed Abstract | Crossref Full Text | Google Scholar

335. Guillamon A, Junque C, Gómez-Gil E. A review of the Status of brain structure research in transsexualism. Arch Sex Behav. (2016) 45(7):1615–48. doi: 10.1007/s10508-016-0768-5

PubMed Abstract | Crossref Full Text | Google Scholar

336. Kreukels BP, Guillamon A. Neuroimaging studies in people with gender incongruence. Int Rev Psychiatry. (2016) 28(1):120–8. doi: 10.3109/09540261.2015.1113163

PubMed Abstract | Crossref Full Text | Google Scholar

337. Mueller SC, De Cuypere G, T’Sjoen G. Transgender research in the 21st century: a selective critical review from a neurocognitive perspective. Am J Psychiatry. (2017) 174(12):1155–62. doi: 10.1176/appi.ajp.2017.17060626

PubMed Abstract | Crossref Full Text | Google Scholar

338. Smith ES, Junger J, Derntl B, Habel U. The transsexual brain–a review of findings on the neural basis of transsexualism. Neurosci Biobehav Rev. (2015) 59:251–66. doi: 10.1016/j.neubiorev.2015.09.008

PubMed Abstract | Crossref Full Text | Google Scholar

339. Joel D, Berman Z, Tavor I, Wexler N, Gaber O, Stein Y, et al. Sex beyond the genitalia: the human brain mosaic. Proc Natl Acad Sci U S A. (2015) 112(50):15468–73. doi: 10.1073/pnas.1509654112

PubMed Abstract | Crossref Full Text | Google Scholar

340. Kurth F, Jancke L, Luders E. Sexual dimorphism of broca’s region: more gray matter in female brains in brodmann areas 44 and 45. J Neurosci Res. (2016) 95(1-2):626–32. doi: 10.1002/jnr.23898

Crossref Full Text | Google Scholar

341. Ritchie SJ, Cox SR, Shen X, Lombardo MV, Reus LM, Alloza C, et al. Sex differences in the adult human brain: evidence from 5216 UK biobank participants. Cereb Cortex. (2018) 28(8):2959–75. doi: 10.1093/cercor/bhy109

PubMed Abstract | Crossref Full Text | Google Scholar

342. Ruigrok AN, Salimi-Khorshidi G, Lai MC, Baron-Cohen S, Lombardo MV, Tait RJ, et al. A meta-analysis of sex differences in human brain structure. Neurosci Biobehav Rev. (2014) 39:34–50. doi: 10.1016/j.neubiorev.2013.12.004

PubMed Abstract | Crossref Full Text | Google Scholar

343. De Bellis MD, Keshavan MS, Beers SR, Hall J, Frustaci K, Masalehdan A, et al. Sex differences in brain maturation during childhood and adolescence. Cereb Cortex. (2001) 11(6):552–7. doi: 10.1093/cercor/11.6.552

PubMed Abstract | Crossref Full Text | Google Scholar

344. Reiss AL, Abrams MT, Singer HS, Ross JL, Denckla MB. Brain development, gender and IQ in children. A volumetric imaging study. Brain. (1996) 119(Pt 5):1763–74. doi: 10.1093/brain/119.5.1763

PubMed Abstract | Crossref Full Text | Google Scholar

345. Allen JS, Damasio H, Grabowski TJ, Bruss J, Zhang W. Sexual dimorphism and asymmetries in the gray-white composition of the human cerebrum. Neuroimage. (2003) 18(4):880–94. doi: 10.1016/S1053-8119(03)00034-X

PubMed Abstract | Crossref Full Text | Google Scholar

346. Filipek PA, Richelme C, Kennedy DN, Caviness VS. The young adult human brain: an MRI-based morphometric analysis. Cereb Cortex. (1994) 4(4):344–60. doi: 10.1093/cercor/4.4.344

PubMed Abstract | Crossref Full Text | Google Scholar

347. Nopoulos P, Flaum M, O'Leary D, Andreasen NC. Sexual dimorphism in the human brain: evaluation of tissue volume, tissue composition and surface anatomy using magnetic resonance imaging. Psychiatry Res. (2000) 98(1):1–13. doi: 10.1016/S0925-4927(99)00044-X

PubMed Abstract | Crossref Full Text | Google Scholar

348. Passe TJ, Rajagopalan P, Tupler LA, Byrum CE, MacFall JR, Krishnan KR. Age and sex effects on brain morphology. Prog Neuropsychopharmacol Biol Psychiatry. (1997) 21(8):1231–7. doi: 10.1016/S0278-5846(97)00160-7

PubMed Abstract | Crossref Full Text | Google Scholar

349. Gur RC, Turetsky BI, Matsui M, Yan M, Bilker W, Hughett P, et al. Sex differences in brain gray and white matter in healthy young adults: correlations with cognitive performance. J Neurosci. (1999) 19(10):4065–72. doi: 10.1523/JNEUROSCI.19-10-04065.1999

PubMed Abstract | Crossref Full Text | Google Scholar

350. Goldstein JM, Seidman LJ, Horton NJ, Makris N, Kennedy DN, Caviness VS, et al. Normal sexual dimorphism of the adult human brain assessed by in vivo magnetic resonance imaging. Cereb Cortex. (2001) 11(6):490–7. doi: 10.1093/cercor/11.6.490

PubMed Abstract | Crossref Full Text | Google Scholar

351. Prendergast DM, Ardekani B, Ikuta T, John M, Peters B, DeRosse P, et al. Age and sex effects on corpus callosum morphology across the lifespan. Hum Brain Mapp. (2015) 36(7):2691–702. doi: 10.1002/hbm.22800

PubMed Abstract | Crossref Full Text | Google Scholar

352. Giedd JN, Vaituzis AC, Hamburger SD, Lange N, Rajapakse JC, Kaysen D, et al. Quantitative MRI of the temporal lobe, amygdala, and hippocampus in normal human development: ages 4-18 years. J Comp Neurol. (1996) 366(2):223–30. doi: 10.1002/(SICI)1096-9861(19960304)366:2%3C223::AID-CNE3%3E3.0.CO;2-7

PubMed Abstract | Crossref Full Text | Google Scholar

353. Marwha D, Halari M, Eliot L. Meta-analysis reveals a lack of sexual dimorphism in human amygdala volume. Neuroimage. (2017) 147:282–94. doi: 10.1016/j.neuroimage.2016.12.021

PubMed Abstract | Crossref Full Text | Google Scholar

354. Tan A, Ma W, Vira A, Marwha D, Eliot L. The human hippocampus is not sexually-dimorphic: meta-analysis of structural MRI volumes. Neuroimage. (2016) 124(Pt A):350–66. doi: 10.1016/j.neuroimage.2015.08.050

PubMed Abstract | Crossref Full Text | Google Scholar

355. McCarthy MM, Arnold AP. Reframing sexual differentiation of the brain. Nat Neurosci. (2011) 14(6):677–83. doi: 10.1038/nn.2834

PubMed Abstract | Crossref Full Text | Google Scholar

356. Massa MG, Correa SM. Sexes on the brain: sex as multiple biological variables in the neuronal control of feeding. Biochim Biophys Acta Mol Basis Dis. (2020) 1866(10):165840. doi: 10.1016/j.bbadis.2020.165840

PubMed Abstract | Crossref Full Text | Google Scholar

357. Eliot L, Ahmed A, Khan H, Patel J. Dump the “dimorphism": comprehensive synthesis of human brain studies reveals few male-female differences beyond size. Neurosci Biobehav Rev. (2021) 125:667–97. doi: 10.1016/j.neubiorev.2021.02.026

PubMed Abstract | Crossref Full Text | Google Scholar

358. Arif-Rahu M, Fisher D, Matsuda Y. Biobehavioral measures for pain in the pediatric patient. Pain Manag Nurs. (2012) 13(3):157–68. doi: 10.1016/j.pmn.2010.10.036

PubMed Abstract | Crossref Full Text | Google Scholar

359. Atlas LY, Wager TD. How expectations shape pain. Neurosci Lett. (2012) 520(2):140–8. doi: 10.1016/j.neulet.2012.03.039

PubMed Abstract | Crossref Full Text | Google Scholar

360. Brooks J, Tracey I. From nociception to pain perception: imaging the spinal and supraspinal pathways. J Anat. (2005) 207(1):19–33. doi: 10.1111/j.1469-7580.2005.00428.x

PubMed Abstract | Crossref Full Text | Google Scholar

361. Bingel U, Tracey I. Imaging CNS modulation of pain in humans. Physiology (Bethesda). (2008) 23:371–80. doi: 10.1152/physiol.00024.2008

PubMed Abstract | Crossref Full Text | Google Scholar

362. Basbaum AI, Jessel T. Pain. In: Kandel E, editors. Principles of Neural Sciences. New York: Appleton and Lange (2021) Available online at: https://neurology.mhmedical.com/content.aspx?bookid=3024&sectionid=254330205 (Accessed April 1, 2022).

Google Scholar

363. Becker S, Navratilova E, Nees F, Van Damme S. Emotional and motivational pain processing: current state of knowledge and perspectives in translational research. Pain Res Manag. (2018) 2018:5457870. doi: 10.1155/2018/5457870

PubMed Abstract | Crossref Full Text | Google Scholar

364. Martucci KT, Mackey SC. Neuroimaging of pain: human evidence and clinical relevance of central nervous system processes and modulation. Anesthesiology. (2018) 128(6):1241–54. doi: 10.1097/ALN.0000000000002137

PubMed Abstract | Crossref Full Text | Google Scholar

365. Becker S, Gandhi W, Schweinhardt P. Cerebral interactions of pain and reward and their relevance for chronic pain. Neurosci Lett. (2012) 520(2):182–7. doi: 10.1016/j.neulet.2012.03.013

PubMed Abstract | Crossref Full Text | Google Scholar

366. Bergeron D, Obaid S, Fournier-Gosselin MP, Bouthillier A, Nguyen DK. Deep brain stimulation of the posterior insula in chronic pain: a theoretical framework. Brain Sci. (2021) 11(5):639. doi: 10.3390/brainsci11050639

Crossref Full Text | Google Scholar

367. Coppieters I, Meeus M, Kregel J, Caeyenberghs K, De Pauw R, Goubert D, et al. Relations between brain alterations and clinical pain measures in chronic musculoskeletal pain: a systematic review. J Pain. (2016) 17(9):949–62. doi: 10.1016/j.jpain.2016.04.005

PubMed Abstract | Crossref Full Text | Google Scholar

368. Mackey S, Greely HT, Martucci KT. Neuroimaging-based pain biomarkers: definitions, clinical and research applications, and evaluation frameworks to achieve personalized pain medicine. Pain Rep. (2019) 4(4):e762. doi: 10.1097/PR9.0000000000000762

PubMed Abstract | Crossref Full Text | Google Scholar

369. Moody TD, Feusner JD, Reggente N, Vanhoecke J, Holmberg M, Manzouri A, et al. Predicting outcomes of cross-sex hormone therapy in transgender individuals with gender incongruence based on pre-therapy resting-state brain connectivity. Neuroimage Clin. (2021) 29:102517. doi: 10.1016/j.nicl.2020.102517

PubMed Abstract | Crossref Full Text | Google Scholar

370. Kurth F, Gaser C, Sánchez FJ, Luders E. Brain sex in transgender women is shifted towards gender identity. J Clin Med. (2022) 11(6):1582. doi: 10.3390/jcm11061582

PubMed Abstract | Crossref Full Text | Google Scholar

371. Starcevic A, Dakovic M, Radojicic Z, Filipovic B. A structural magnetic resonance imaging study in therapy-naïve transsexual individuals. Folia Morphol (Warsz). (2021) 80(2):442–7. doi: 10.5603/FM.a2020.0073

PubMed Abstract | Crossref Full Text | Google Scholar

372. Clemens B, Votinov M, Puiu AA, Schüppen A, Hüpen P, Neulen J, et al. Replication of previous findings? Comparing gray matter volumes in transgender individuals with gender incongruence and cisgender individuals. J Clin Med. (2021) 10(7):1454. doi: 10.3390/jcm10071454

Crossref Full Text | Google Scholar

373. Hagemann G, Ugur T, Schleussner E, Mentzel HJ, Fitzek C, Witte OW, et al. Changes in brain size during the menstrual cycle. PLoS One. (2011) 6(2):e14655. doi: 10.1371/journal.pone.0014655

PubMed Abstract | Crossref Full Text | Google Scholar

374. Kim TH, Kim B, Kim YR, Jeong CW, Lee YH. Gray matter differences associated with menopausal hormone therapy in menopausal women: a DARTEL-based VBM study. Sci Rep. (2023) 13(1):1401. doi: 10.1038/s41598-023-28673-2

PubMed Abstract | Crossref Full Text | Google Scholar

375. Schneider MA, Spritzer PM, Minuzzi L, Frey BN, Syan SK, Fighera TM, et al. Effects of estradiol therapy on resting-state functional connectivity of transgender women after gender-affirming related gonadectomy. Front Neurosci. (2019) 13:817. doi: 10.3389/fnins.2019.00817

PubMed Abstract | Crossref Full Text | Google Scholar

376. Beauregard M. Mind does really matter: evidence from neuroimaging studies of emotional self-regulation, psychotherapy, and placebo effect. Prog Neurobiol. (2007) 81(4):218–36. doi: 10.1016/j.pneurobio.2007.01.005

PubMed Abstract | Crossref Full Text | Google Scholar

377. Carvalho-Netto EF, Myers B, Jones K, Solomon MB, Herman JP. Sex differences in synaptic plasticity in stress-responsive brain regions following chronic variable stress. Physiol Behav. (2011) 104(2):242–7. doi: 10.1016/j.physbeh.2011.01.024

PubMed Abstract | Crossref Full Text | Google Scholar

378. Cutolo M, Sulli A, Capellino S, Villaggio B, Montagna P, Seriolo B, et al. Sex hormones influence on the immune system: basic and clinical aspects in autoimmunity. Lupus. (2004) 13(9):635–8. doi: 10.1191/0961203304lu1094oa

PubMed Abstract | Crossref Full Text | Google Scholar

379. Bhatia A, Sekhon HK, Kaur G. Sex hormones and immune dimorphism. ScientificWorldJournal. (2014) 2014:159150. doi: 10.1155/2014/159150

PubMed Abstract | Crossref Full Text | Google Scholar

380. Annibalini G, Agostini D, Calcabrini C, Martinelli C, Colombo E, Guescini M, et al. Effects of sex hormones on inflammatory response in male and female vascular endothelial cells. J Endocrinol Invest. (2014) 37(9):861–9. doi: 10.1007/s40618-014-0118-1

PubMed Abstract | Crossref Full Text | Google Scholar

381. Jiménez-Sánchez L, Hamilton OKL, Clancy U, Backhouse EV, Stewart CR, Stringer MS, et al. Sex differences in cerebral small vessel disease: a systematic review and meta-analysis. Front Neurol. (2021) 12:756887. doi: 10.3389/fneur.2021.756887

Crossref Full Text | Google Scholar

382. Pavlovic JM, Akcali D, Bolay H, Bernstein C, Maleki N. Sex-related influences in migraine. J Neurosci Res. (2017) 95(1-2):587–93. doi: 10.1002/jnr.23903

PubMed Abstract | Crossref Full Text | Google Scholar

383. Gupta A, Mayer EA, Fling C, Labus JS, Naliboff BD, Hong JY, et al. Sex-based differences in brain alterations across chronic pain conditions. J Neurosci Res. (2017) 95(1-2):604–16. doi: 10.1002/jnr.23856

PubMed Abstract | Crossref Full Text | Google Scholar

384. Chen X, Sachdev PS, Wen W, Anstey KJ. Sex differences in regional gray matter in healthy individuals aged 44-48 years: a voxel-based morphometric study. Neuroimage. (2007) 36(3):691–9. doi: 10.1016/j.neuroimage.2007.03.063

PubMed Abstract | Crossref Full Text | Google Scholar

385. Button KS, Ioannidis JP, Mokrysz C, Nosek BA, Flint J, Robinson ES, et al. Power failure: why small sample size undermines the reliability of neuroscience. Nat Rev Neurosci. (2013) 14(5):365–76. doi: 10.1038/nrn3475

PubMed Abstract | Crossref Full Text | Google Scholar

386. Amanzio M, Palermo S. Pain anticipation and nocebo-related responses: a descriptive Mini-review of functional neuroimaging studies in normal subjects and precious hints on pain processing in the context of neurodegenerative disorders. Front Pharmacol. (2019) 10:969. doi: 10.3389/fphar.2019.00969

PubMed Abstract | Crossref Full Text | Google Scholar

387. Khorashad BS, Manzouri A, Feusner JD, Savic I. Cross-sex hormone treatment and own-body perception: behavioral and brain connectivity profiles. Sci Rep. (2021) 11(1):2799. doi: 10.1038/s41598-020-80687-2

PubMed Abstract | Crossref Full Text | Google Scholar

388. Gómez Á, Cerdán S, Pérez-Laso C, Ortega E, Pásaro E, Fernández R, et al. Effects of adult male rat feminization treatments on brain morphology and metabolomic profile. Horm Behav. (2020) 125:104839. doi: 10.1016/j.yhbeh.2020.104839

Crossref Full Text | Google Scholar

389. Kranz GS, Hahn A, Kaufmann U, Tik M, Ganger S, Seiger R, et al. Effects of testosterone treatment on hypothalamic neuroplasticity in female-to-male transgender individuals. Brain Struct Funct. (2018) 223(1):321–8. doi: 10.1007/s00429-017-1494-z

PubMed Abstract | Crossref Full Text | Google Scholar

390. Kranz GS, Zhang BBB, Handschuh P, Ritter V, Lanzenberger R. Gender-affirming hormone treatment—a unique approach to study the effects of sex hormones on brain structure and function. Cortex. (2020) 129:68–79. doi: 10.1016/j.cortex.2020.04.005

PubMed Abstract | Crossref Full Text | Google Scholar

391. Breedlove SM. Sexual differentiation of the human nervous system. Annu Rev Psychol. (1994) 45:389–418. doi: 10.1146/annurev.ps.45.020194.002133

PubMed Abstract | Crossref Full Text | Google Scholar

392. Hines M. Gender development and the human brain. Annu Rev Neurosci. (2011) 34:69–88. doi: 10.1146/annurev-neuro-061010-113654

PubMed Abstract | Crossref Full Text | Google Scholar

393. Reardon PK, Clasen L, Giedd JN, Blumenthal J, Lerch JP, Chakravarty MM, et al. An allometric analysis of sex and sex chromosome dosage effects on subcortical anatomy in humans. J Neurosci. (2016) 36(8):2438–48. doi: 10.1523/JNEUROSCI.3195-15.2016

PubMed Abstract | Crossref Full Text | Google Scholar

394. Caes L, Roche M. Adverse early life experiences are associated with changes in pressure and cold pain sensitivity in young adults. Ann Palliat Med. (2020) 9(4):1366–9. doi: 10.21037/apm-20-914

PubMed Abstract | Crossref Full Text | Google Scholar

395. Waller R, Smith AJ, O’Sullivan PB, Slater H, Sterling M, Straker LM. The association of early life stressors with pain sensitivity and pain experience at 22 years. Pain. (2020) 161(1):220–9. doi: 10.1097/j.pain.0000000000001704

PubMed Abstract | Crossref Full Text | Google Scholar

396. Simon E, Zsidó AN, Birkás B, Csathó Á. Pain catastrophizing, pain sensitivity and fear of pain are associated with early life environmental unpredictability: a path model approach. BMC Psychol. (2022) 10(1):97. doi: 10.1186/s40359-022-00800-0

PubMed Abstract | Crossref Full Text | Google Scholar

397. Ahmed W, Pai M, Muhammad T, Maurya C, Mohanty P, Javed NB. Early life factors associated with the experiences of pain in later life: evidence from a population based study in India. BMC Public Health. (2023) 23(1):968. doi: 10.1186/s12889-023-15805-6

PubMed Abstract | Crossref Full Text | Google Scholar

398. French FS, Lubahn DB, Brown TR, Simental JA, Quigley CA, Yarbrough WG, et al. Molecular basis of androgen insensitivity. Recent Prog Horm Res. (1990) 46:1–38. doi: 10.1016/b978-0-12-571146-3.50005-5

PubMed Abstract | Crossref Full Text | Google Scholar

399. McCarthy MM. A new view of sexual differentiation of mammalian brain. J Comp Physiol A Neuroethol Sens Neural Behav Physiol. (2020) 206(3):369–78. doi: 10.1007/s00359-019-01376-8

PubMed Abstract | Crossref Full Text | Google Scholar

400. McCarthy MM. How it’s made: organisational effects of hormones on the developing brain. J Neuroendocrinol. (2010) 22(7):736–42. doi: 10.1111/j.1365-2826.2010.02021.x

PubMed Abstract | Crossref Full Text | Google Scholar

401. McCarthy MM, De Vries GJ, Forger NG. In: Pfaff DW, Joels M, editors. Hormones, Brain and Behavior. 3: Amsterdam: Elsevier; 2017. p. 5–32.

Google Scholar

402. McCarthy MM, Herold K, Stockman SL. Fast, furious and enduring: sensitive versus critical periods in sexual differentiation of the brain. Physiol Behav. (2018) 187:13–9. doi: 10.1016/j.physbeh.2017.10.030

PubMed Abstract | Crossref Full Text | Google Scholar

403. McCarthy MM, Pickett LA, VanRyzin JW, Kight KE. Surprising origins of sex differences in the brain. Horm Behav. (2015) 76:3–10. doi: 10.1016/j.yhbeh.2015.04.013

PubMed Abstract | Crossref Full Text | Google Scholar

404. Sisk CL, Foster DL. The neural basis of puberty and adolescence. Nat Neurosci. (2004) 7(10):1040–7. doi: 10.1038/nn1326

PubMed Abstract | Crossref Full Text | Google Scholar

405. Sisk CL, Zehr JL. Pubertal hormones organize the adolescent brain and behavior. Front Neuroendocrinol. (2005) 26(3-4):163–74. doi: 10.1016/j.yfrne.2005.10.003

PubMed Abstract | Crossref Full Text | Google Scholar

406. Arnold AP. A general theory of sexual differentiation. J Neurosci Res. (2017) 95(1-2):291–300. doi: 10.1002/jnr.23884

PubMed Abstract | Crossref Full Text | Google Scholar

407. Hohmann CF, Beard NA, Kari-Kari P, Jarvis N, Simmons Q. Effects of brief stress exposure during early postnatal development in Balb/CByJ mice: iI. Altered cortical morphology. Dev Psychobiol. (2012) 54(7):723–35. doi: 10.1002/dev.21028

PubMed Abstract | Crossref Full Text | Google Scholar

408. Sequeira MK, Bolton JL. Stressed microglia: neuroendocrine-neuroimmune interactions in the stress response. Endocrinology. (2023) 164(7):bqad088. doi: 10.1210/endocr/bqad088

PubMed Abstract | Crossref Full Text | Google Scholar

409. Bridges RS, Zarrow MX, Goldman BD, Denenberg VH. A developmental study of maternal responsiveness in the rat. Physiol Behav. (1974) 12(1):149–51. doi: 10.1016/0031-9384(74)90082-1

PubMed Abstract | Crossref Full Text | Google Scholar

410. Mennella JA, Moltz H. Infanticide in rats: male strategy and female counter-strategy. Physiol Behav. (1988) 42(1):19–28. doi: 10.1016/0031-9384(88)90254-5

PubMed Abstract | Crossref Full Text | Google Scholar

411. Mennella JA, Moltz H. Infanticide in the male rat: the role of the vomeronasal organ. Physiol Behav. (1988) 42(3):303–6. doi: 10.1016/0031-9384(88)90087-X

PubMed Abstract | Crossref Full Text | Google Scholar

412. Rosenblatt J, Mayer A, Giordano A. Hormonal basis during pregnancy for the onset of maternal behavior in the rat. Psychoneuroendocrinology. (1988) 13(1-2):29–46. doi: 10.1016/0306-4530(88)90005-4

PubMed Abstract | Crossref Full Text | Google Scholar

413. Tooley UA, Bassett DS, Mackey AP. Environmental influences on the pace of brain development. Nat Rev Neurosci. (2021) 22(6):372–84. doi: 10.1038/s41583-021-00457-5

PubMed Abstract | Crossref Full Text | Google Scholar

414. Merrill SM, Gladish N, Kobor MS. Social environment and epigenetics. Curr Top Behav Neurosci. (2019) 42:83–126. doi: 10.1007/7854_2019_114

PubMed Abstract | Crossref Full Text | Google Scholar

415. Rivi V, Rigillo G, Toscano Y, Benatti C, Blom JMC. Narrative review of the complex interaction between pain and trauma in children: a focus on biological memory, preclinical data, and epigenetic processes. Children (Basel). (2023) 10(7):1217. doi: 10.3390/children10071217

PubMed Abstract | Crossref Full Text | Google Scholar

416. Kodila ZN, Shultz SR, Yamakawa GR, Mychasiuk R. Critical windows: exploring the association between perinatal trauma, epigenetics, and chronic pain. Neuroscientist. (2023):10738584231176233. doi: 10.1177/10738584231176233

PubMed Abstract | Crossref Full Text | Google Scholar

417. National Institute of Health. NIH-Wide Strategic Plan, Fiscal Years 2021–2025. Washington, DC: National Institutes of Health (2021).

Google Scholar

418. National Institure of Health. Consideration of Sex as a Biological Variable in NIH-funded Research. Washington, DC (2015). Available online at: https://grants.nih.gov/grants/guide/notice-files/NOT-OD-15-102.html (Accessed November 3, 2021).

Google Scholar

419. National Institute of Health. Advancing Science for the Health of Women: The Trans-NIH strategic plan for women’s health research. Washington, DC: National Institute of Health (2019).

Google Scholar

420. National Institute of Health. Mechanisms, models, measurement, & management in pain research (R01 clinical trial optional). In: Department of Health and Human Services, editor. Washington, DC (2017). Available online at: https://grants.nih.gov/grants/guide/pa-files/PA-18-141.html (Accessed November 3, 2021).

Google Scholar

421. Burke SM, Manzouri AH, Dhejne C, Bergström K, Arver S, Feusner JD, et al. Testosterone effects on the brain in transgender men. Cereb Cortex. (2018) 28(5):1582–96. doi: 10.1093/cercor/bhx054

PubMed Abstract | Crossref Full Text | Google Scholar

422. Burke SM, Manzouri AH, Savic I. Structural connections in the brain in relation to gender identity and sexual orientation. Sci Rep. (2017) 7(1):17954. doi: 10.1038/s41598-017-17352-8

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: transgender (GLBT) issues, post operative pain, gender binary system, hormone replacement therapy (HRT), gender affirming care

Citation: Anger JT, Case LK, Baranowski AP, Berger A, Craft RM, Damitz LA, Gabriel R, Harrison T, Kaptein K, Lee S, Murphy AZ, Said E, Smith SA, Thomas DA, Valdés Hernández MdC, Trasvina V, Wesselmann U and Yaksh TL (2024) Pain mechanisms in the transgender individual: a review. Front. Pain Res. 5:1241015. doi: 10.3389/fpain.2024.1241015

Received: 15 June 2023; Accepted: 25 January 2024;
Published: 27 March 2024.

Edited by:

Kevin M. Hellman, NorthShore University HealthSystem, United States

Reviewed by:

Lesley Marson, Dignify Therapeutics LLC, United States
Scott Holmes, Harvard Medical School, United States

© 2024 Anger, Case, Baranowski, Berger, Craft, Damitz, Gabriel, Harrison, Kaptein, Lee, Murphy, Said, Smith, Thomas, Valdés Hernández, Trasvina, Wesselmann and Yaksh. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jennifer T. Anger janger@health.ucsd.edu Tony L. Yaksh tyaksh@health.ucsd.edu

Download