Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Aging Neurosci., 01 December 2025

Sec. Alzheimer's Disease and Related Dementias

Volume 17 - 2025 | https://doi.org/10.3389/fnagi.2025.1632236

The locus coeruleus, a blue spot for early diagnosis and prognosis of Alzheimer’s disease

  • 1Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, United States
  • 2Department of Neurological Surgery, School of Medicine, University of California, Davis, Davis, CA, United States
  • 3Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
  • 4Institute for Brain and Cognition, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran

Intact functioning of the locus coeruleus-noradrenergic (LC-NA) system is pivotal in the control of numerous central processes, and damage to these systems leads to a wide range of nervous system disorders. The LC, as the main source of noradrenaline (NA) in the mammalian brain, was the first central nervous system (CNS) modulatory structure to be anatomically and biochemically characterized. LC-NA release exerts both excitatory and inhibitory effects on target areas. Over the past few decades, LC damage has been causatively identified as a common factor in CNS diseases, notably neurodegenerative diseases. Moreover, LC damage is the likely manifestation of common pathophysiological processes, thus elevating the LC as a diagnostic and therapeutic target, especially in Parkinson’s and Alzheimer’s diseases (PD & AD). This review also addresses why LC neurons, compared to other areas in particular, are highly vulnerable and sensitive to damage—such as specific anatomical features, tau phosphorylation, and high neuronal energy requirements—will be described in this review article. Finally, we explore whether these known LC vulnerabilities might be leveraged towards improved early diagnostic and prognostic biomarkers for AD.

1 Introduction

The activity of the brain’s neuromodulatory systems affect arousal, mood, attention, and motivation and consequently influence the behavior. The locus coeruleus (LC; “blue spot” in Latin) is the main hub of central noradrenaline (NA) which, as a key neuromodulator, affects forebrain-related cognitive functions and behaviors, including working memory, selective attention, decision-making, and adaptive behavior (Mather and Harley, 2016). Considering that several neurodegenerative and psychiatric disorders in humans are tightly related to disruption of NA signaling, it exemplifies the clinical importance of the LC-NA system (Sara, 2009; Feinstein et al., 2016). Other recent review articles explore technological advances that either allow a deeper functional understanding of the LC (Poe et al., 2020), or advanced imaging of the LC and other neuromodulatory areas (Engels-Dominguez et al., 2023; Galgani et al., 2020). Here, we provide updated references to both of these areas and focus on the very early stages of Alzheimer’s that impact the LC.

Discovery of the LC dates back nearly 240 years. The preeminent comparative anatomist and neuroanatomist of the 18th century, Félix Vicq d’Azyr, was the first to depict a dark blue-ish area at the base of the fourth ventricle in axial sections of the human brain drawn by Alexandre Briceau and published in 1786 [Planche XXIX, Figure III, area 12; (Vicq d’Azyr, 1786; Tubbs et al., 2011)]. In 1809, German anatomist Johann Christian Reil offered a descriptive location (“wo der vördere Schenkel mit der Area zusammenstößt”) of a darker tissue (“schimmert ein Strich von Schwarzer Substanz durch, die bloß mit dem Epithelium bedeck ist.”) (Reil, 1809). The translation offered in Chandler et al. (2019) is reminiscent of d’Azyr’s drawings: “In the angle at which the anterior shank [the Pedunculus cerebellaris superior] comes together with the adjoining area, a stripe of black substance shimmers through, only covered by the epithelium.” Writing in Latin in 1812 and using the alternate spelling “loculi caerulei” (Swanson, 2015), the brothers Joseph and Karl Wenzel described an area at the base of the fourth ventricle as ‘oblong, narrow bluish place [that] immediately strikes the eye at first sight’ (original Latin on p. 168: “angustus et caeruleus locus primo statim obtutu oculos ferit”) (Wenzel and Karl, 1812). In 1909, Jacobsohn described one main and two ancillary “nucleus pigmentosus pontis” but clearly identified the “triangular” main division in the corner of the fourth ventricle as “nucleus pigmentosus loci coerulei” worthy of particular attention due to its “brown[−ish] Pigment” (Jacobsohn, 1909). Additional details about the early history of the LC can be found in (Maeda, 2000). Importantly, the prominent dark, brown, or blue color, depending upon the preparation, is due to the accumulation of neuromelanin, formed through the oxidation and polymerization of catecholamines and their metabolites, principally dopamine and norepinephrine. It is this feature that attracted so many early neuroanatomists to the LC, which is hypothesized to be one of the reasons for its sensitivity to neurodegeneration (Sulzer et al., 2018), and potentially its use as an early biomarker of neurodegeneration in Alzheimer’s disease (AD).

1.1 Functional anatomy of the LC nucleus

The bilateral nucleus of the LC is located on the ventrolateral surface, as stated above ‘in the corner,’ of the fourth ventricle in the pons (Williams et al., 1984). Dimensions of this tiny nucleus in humans are estimated to be 12.0–17.0 mm, 2.5 mm, and 2.0 mm in length, width, and height, respectively (Fernandes et al., 2012). Neuronal nodes located in the brainstem containing NA are called “noradrenergic cell groups.” They include A1-A7, with the A6, locus coeruleus, being the largest among them (Dahlström and Fuxe, 1964). This tiny nucleus consists of compact cells, estimated to be around 45,000–50,000 in both hemispheres of the healthy adult human brain (Sharma et al., 2010), and although some variation has been noted, older individuals tend to have fewer LC neurons, which is more pronounced in those with dementia (German et al., 1988; Mouton et al., 1994; Kelly et al., 2017). The LC is composed of neurons that are mainly medium-sized cells with a fusiform and polar morphology, and smaller-sized neurons of various shapes (Chan-Palay and Asan, 1989; Patt and Gerhard, 1993). The medium sized neurons are projection neurons that suffer the most dendritic, axonal and cell body losses in neurodegenerative diseases (Williams et al., 1984; Chen et al., 2022). On a broader, “dendroarchitectonic” level, the LC is generally considered part of set of brainstem and pontine nuclei referred to as the “isodendritic core” contained within the reticular system as termed by Enrique Ramón-Moliner (Geser et al., 2022). Of the three neuronal types identified by Ramón-Moliner (allo- and idio-dendritic being the other two), the isodendritic neurons have the least dendritic complexity and thus could be hypothesized to be the most phylogenetically conserved form of neurons present in mammals (Ramon-Moliner and Nauta, 1966). Similar to other areas of the isodendritic core, the LC innervates throughout the neuraxis with volume transmission, and it is this combination of anatomical features that has been proposed to make this entire class of neurons particularly vulnerable to tauopathies (Gambardella et al., 2017; Lew et al., 2021) and AD pathogenesis (Giorgi et al., 2017; Theofilas et al., 2015). The LC itself is a mostly homogenous nucleus in which most neurons release NA (Szabadi, 2013; Taylor and Westlund, 2017), with simultaneous expression of a variety of neuropeptides, including neuropeptide Y, somatostatin, and cholecystokinin in a subset of neurons (Benarroch, 2018). Later studies on LC have asked how a small, homogeneous structure can respond to various sensory stimuli, modify neuronal actions in separate brain areas, and induce different behaviors. These studies have highlighted some heterogeneity within the LC-NA system (Schwarz and Luo, 2015; Chandler et al., 2014).

NA is one of the first neurotransmitters identified in the CNS, which was discovered and introduced by the Swedish physiologist Ulf von Euler in the 1940s (Von Euler, 1946). Apart from the studies that two Japanese researchers published during World War II, the first study about LC function was done by Russell in 1955 (Maeda, 2000). NA is released by LC neurons in two ways, synaptic and non-synaptic (volume) transmission. The latter involves the diffusion of NA into the extracellular space and the paracrine effects on neurons, glial cells, and capillaries (Waterhouse et al., 1977). The LC-NA system’s effects are mainly exerted by different types of NA receptors and their distribution in the LC target areas. Noradrenoceptors belong to three families of G protein-coupled receptors, including alpha-1, alpha-2, and beta, which operate with different mechanisms and each one has several subtypes (Benarroch, 2018). Various studies have reported multiple effects of NA on membrane potential, cell excitability, intracellular signaling cascades, and synaptic plasticity in target regions of the LC projections (Sara, 2009; Feinstein et al., 2016).

LC neurons, despite their small number, project throughout the neuraxis with the exception of the striatum, nucleus accumbens, substantia nigra, and globus pallidus. In addition, based on more recent studies, these neurons receive direct input from more than one hundred brain regions (Schwarz and Luo, 2015; Schwarz et al., 2015). For this reason, the LC plays a key role in regulating various functions such as the autonomic system, vigilance, sleep/wake cycle, sensory processing, attention, memory and learning, pain processing, and drug dependence (Poe et al., 2020; Szabadi, 2013; Suárez-Pereira et al., 2022; Alaee et al., 2022; Farahani et al., 2021; Farahani et al., 2023; Ahmadi-Soleimani et al., 2020; Samuels and Szabadi, 2008; Foote and Berridge, 2019; Aston-Jones and Waterhouse, 2016).

1.2 Physiology of the LC nucleus

LC neurons are electrically connected via gap junctions that respond to environmental stimuli (de Carvalho et al., 2014), and their electrophysiological activity is modulated by the synaptic release of various neurotransmitters. LC activity is sculpted by both subcortical inputs—for example, the orexinergic inputs from the lateral hypothalamus and corticotropin-releasing peptide from the central nuclei of the amygdala—as well as descending glutamatergic projections from the prefrontal and anterior cingulate cortices (Gompf et al., 2010). Furthermore, NA released from LC neurons provides feedback inhibition via alpha-2 autoreceptors, which are expressed in the cell bodies and dendrites (Benarroch, 2018; Benarroch, 2009).

In addition to the above-mentioned extent of LC projections, their distribution in the cortex is also heterogeneous. For example, in humans the most extensive innervations of the LC project to the somatosensory and motor cortices and associative regions including prefrontal and parietal cortices, and the temporal cortex, which is critically involved in memory formation (Lewis and Morrison, 1989; Gaspar et al., 1989). Using these connections, LC can contribute to alertness, response to stress, attention, and memory.

LC neurons fire action potentials in two distinct patterns, tonic and phasic. By switching between these two modes, different behavioral states are regulated. Basal tonic activity is characterized by continuous high-frequency (2–5 Hz) discharge in the awake and alert state. In quiet wakefulness, LC neurons fire regularly and at a low frequency of approximately 1 Hz, while they fire bursts in response to alerting stimuli. From drowsiness to slow-wave sleep, their firing frequency decreases, and LC neuronal activity is silent during REM sleep until the transition back to wakefulness. This ability to change firing patterns is crucial in maintaining the organism’s behavioral adaptation to environmental changes (Sara, 2009; Aston-Jones and Cohen, 2005).

During selective attention, the LC nucleus may temporarily stop its tonic firing and respond to the desired stimulus with phasic firing. This transient phasic firing pattern moderately elevates NA concentrations which in turn facilitates working memory formation mainly by acting on high affinity alpha-2 receptors. On the other hand, high concentrations of NA, resulting from augmentation of tonic firing, which is thought to occur during stress, activate medium-affinity adrenoceptors, alpha-1, and may cause defects in the function of the prefrontal cortex (Berridge and Waterhouse, 2003; Arnsten et al., 2012).

The role of the LC in the stress response is first manifested by the effect of NA on the beta receptors of the basolateral part of the amygdala. In addition, the inputs of the central nucleus of the amygdala to the LC are involved in increasing LC activity following stress. Indeed, laboratory evidence shows that the stress-induced activity of orexinergic neurons of the hypothalamus activates the orexin type 1 receptor in the LC. NA also contributes to the manifestations of autonomic system activity, such as heart rate and blood pressure. The LC is involved in the processing and modulation of pain through its connections with pain-processing centers such as the dorsal horn of the spinal cord, sensory nuclei of the trigeminal nerve, and the thalamus (Szabadi, 2013; Samuels and Szabadi, 2008; Barsegyan et al., 2014).

These afferent, efferent and intra-LC pathways may impact symptomatology throughout the course of AD. For example, the amygdala, another brain area displaying early susceptibility to AD pathology, influences network connectivity to various cortical regions, and also subcortical structures such as the LC, leading to neuropsychiatric symptoms throughout the course of AD (Stouffer et al., 2024). Numerous pharmacotherapies have been proposed for neuropsychiatric symptoms in AD (Pless et al., 2023). Amongst these symptoms, primarily apathy and anxiety in AD are likely to benefit most from noradrenergic treatment regimens (David et al., 2022; Liu et al., 2018; La et al., 2019); however, depression (Herrmann et al., 2004) and to some degree psychosis (Orlando et al., 2023) associated with AD have been shown to benefit from pharmacotherapies that target multiple monoaminergic systems, including noradrenergic receptors. For instance, tricyclic antidepressants that target norepinephrine and serotonin for depression, and atypical antipsychotics for psychosis, though the latter is highly controversial due to the negative side effects in older individuals. Since multiple noradrenergic target sites are coordinated by LC activity, LC degeneration is expected to affect larger-scale network reconfiguration, and thus functional connectivity studies might inform future drug intervention strategies. Although functional connectivity mapping of the LC has been hindered by its relatively small size (Serra et al., 2018), proxies to LC connectivity such as pupil diameter could be developed for AD research studies and patient diagnostics instead of LC fMRI to overcome the limitations of its small size (Wu et al., 2025). As discussed below, higher spatial resolution fMRI studies should begin to answer to which degree LC connectivity correlates with early disease biomarkers such as APOE4 (Um et al., 2024) as well as symptomatic ramifications of AD, as has already been demonstrated for the role of the LC in depressive symptoms (Dai et al., 2023) and agitation (Liu et al., 2025) in AD, and in ageing more generally (Jacobs et al., 2018; Mijalkov et al., 2025; Vereb et al., 2023).

2 Vulnerability of LC neurons

Cell body abnormalities and loss in Parkinson’s disease (PD) were first noted in the 1950s, around the same time as comparable alterations were observed in postmortem aged and AD tissue (Clewett et al., 2016; Gesi et al., 2000). After that, LC degeneration and the subsequent reduction of NA in the CNS have been reported in a wide range of additional neurological disorders and neurodegenerative diseases, including anxiety, depression, sleep disorders, post-traumatic stress disorder, Huntington disease, and multiple sclerosis (MS), which is primarily a neuroinflammatory disorder rather than a classic neurodegenerative disease, as well as AD (Bielau et al., 2012; Chandley et al., 2013; George et al., 2013; Leiman and Hämmerer, 2024; Mann et al., 1982; Pietrzak et al., 2013; Polak et al., 2011; Rajda et al., 2006; Zhu et al., 2015; Zoukos et al., 1992; Zweig et al., 1992). A large part of the studies conducted on LC degeneration have been done in relation to AD. Histological evidence indicates that the number of LC neurons decreases by 60% in AD patients (Mann et al., 1982; Tomlinson et al., 1981; Perry et al., 1981) and up to 80% in PD patients (Zarow et al., 2003). It is important to note that the LC shares similar vulnerability to AD pathology and a number of other neurodegenerative diseases, including PD, and that other nodes of the isodendritic core share vulnerabilities that are seen in the LC. A number of review articles provide information about these issues that are outside of the scope of this review (Theofilas et al., 2015; Ehrenberg et al., 2023; Grinberg et al., 2011).

LC neurodegeneration is significantly correlated with the number of plaques, the duration and severity of dementia; it is substantially greater than other subcortical nuclei such as the substantia nigra, and is seen in earlier stages of the disease (Zarow et al., 2003; Bondareff et al., 1987; Grudzien et al., 2007). Concomitantly, the loss of LC via experimental lesions leads to greater amyloid deposits and increased neuroinflammation in LC projection areas (Heneka et al., 2006; Heneka et al., 2010). Therefore, both the onset and progression of AD pathogenesis are intertwined with LC degeneration (Beardmore et al., 2021). In this section we discuss the possible reasons for the vulnerability of these neurons. We later turn our attention to how these known vulnerabilities can serve as prognostic tools in the care and treatment of AD patients.

2.1 LC degeneration during aging

LC degeneration is a normal part of aging, though it is accelerated in AD. Progressively through the fourth decade of life, 25%–40% of pigmented LC neurons are lost, most prominently in the rostral part of this nucleus (Chan-Palay and Asan, 1989; Mann et al., 1982; Vijayashankar and Brody, 1979; Manaye et al., 1995), and this starts earlier in men compared with women (Wree et al., 1980). Not only do LC neuronal numbers decrease by ~25%, the level of NA in the brain also is reduced by 40% between the 4th and 9th decades of life (Marien et al., 2004), and this can be recapitulated in rodents, where the reduction is generally less prominent than in humans (Leslie et al., 1985).

LC neurons are sensitive to chronic inflammation and microglial activity, events that occur naturally during aging, yet are exacerbated in neurodegenerative diseases (Bardou et al., 2014). This is also the case with the effects of NA on microglia in LC target regions, where beta-amyloid clearance is attenuated when NA levels are lower in older subjects (Kong et al., 2010; Le et al., 2025). Therefore, in those treatments that target microglial phagocytosis, the level of NA is viewed as an important regulatory factor in that LC neurodegeneration could exacerbate AD progression (Heneka et al., 2010). NA itself is neuroprotective and anti-inflammatory by inducing expression of inhibitor kappa B-alpha (IκB-α) in neurons, and nitric oxide synthase type 2 (NOS2) in glial cells; while also reducing tumor necrosis factor alpha (TNF-α) expression in microglia and interferon gamma (Ifγ) in astrocytes following inflammation (Sara, 2009). Other key neuroprotective proteins are also negatively affected by the reduction in NA in target areas (Biagioni et al., 2024). Finally, capillary blood flow and water permeability are regulated by NA, and the same exacerbating effect of LC neurodegeneration can therefore be found in AD and PD compared to normal aging in terms of tight junction disruption at the blood–brain barrier (BBB) and blood volume / oxygen demand homeostasis (Mann, 1983; Kalinin et al., 2006; Bekar et al., 2012).

Age-related degeneration of the LC causes deficits in attention, episodic and working memory, and cognition (Mather, 2018; Hämmerer et al., 2018; Dahl et al., 2019), and quite possibly through the mechanisms described above. Since AD occurs in the context of aging, AD could be seen as one of a continuum of events that broadly fall under the category of aging. It is therefore important to identify what the key differences are between normal aging of the brain and AD.

2.2 Phosphorylation of tau protein

Tau protein phosphorylation is known as one of the primary causes of LC degeneration during aging and disease (Omoluabi et al., 2025; Satoh and Iijima, 2019). Tau proteins are a group of six highly soluble protein isoforms that play a role in maintaining the stability of microtubules in axons and are abundantly expressed in CNS neurons (Bachmann et al., 2021; Barbier et al., 2019). Tau proteins are normally phosphorylated to preserve microtubule function; however, under pathological conditions, these proteins become hyperphosphorylated, insoluble, and inefficient. Hyperphosphorylated tau exhibits a reduced affinity for microtubules, leading to destabilization of the axonal cytoskeleton and impaired intracellular transport of organelles and vesicles. This disruption hampers synaptic function and contributes to neuronal degeneration observed in aging and neurodegenerative diseases (Rodriguez-Martin et al., 2013). Recent studies have shown that hyperphosphorylated tau accumulates in neurons, causing neurite degeneration, loss of viable synapses, and behavioral abnormalities, even in the absence of fibrillar tau structures (Watamura et al., 2025). The hyperphosphorylated tau itself does not immediately cause neuronal damage, instead accumulating over decades in a soluble “pretangle” state prior to the first signs of aggregated neurofibrillary tangles, tauopathy or any clinical manifestations of mild cognitive impairment. This process occurs in the LC long before any other brain regions (Giorgi et al., 2019; Braak et al., 2011). In the later stages, the presence of amyloid exacerbates tauopathy and this in turn may mediate Aβ neurotoxicity in the cortex in humans and also in Aβ-overproducing transgenic rat model (Cohen et al., 2013). Interestingly, it has been shown that the accumulation of the phosphorylated tau protein co-localizes with active caspase enzymes in LC neurons. Since caspase enzymes are involved in programmed cell death, it is possible that the accumulation of the phosphorylated tau protein in the LC neurons plays a role in their degeneration (Wai et al., 2009). These findings suggest that the LC nucleus contributes to the onset and progression of AD (Bueicheku et al., 2024; Ehrenberg et al., 2017).

During the course of the progressing degeneration, compensatory mechanisms—such as high NA turnover (Hoogendijk et al., 1999) and LC hyperactivity (Weinshenker, 2018; Kelberman et al., 2023) in both humans and rodent models of AD—become increasingly dominant. This model of LC hyperactivity predicts that as establishment of hyperphosphorylated tau within the LC results in increased neuronal activity causing pre-MCI (“prodromal”) symptoms, such as anxiety, hyperarousal (Kelberman et al., 2023; Kelberman et al., 2022), and in humans, aggression (Bachman and Rabins, 2006; Ballard and Corbett, 2013; Scarmeas et al., 2007). As the disease progresses, phosphorylated tau proteins form neurofibrillary tangles (NFT) in the LC, intensifying cell loss, and MCI, eventually resulting in AD diagnosis (Orlando et al., 2023). Kelly et al. have shown that in AD, tau pathology of LC neurons decreased the expression of the genes related to mitochondrial function (Kelly et al., 2017). Experimentally, LC neuronal activity transitions from hyper- to hypo-activity in the later stages of AD (Kelberman et al., 2023), and chemogenetic activation of the LC reverses cognitive deficits, indicating a causative relationship (Rorabaugh et al., 2017). Combined, evidence from human and animal models indicates that tau pathology within the LC is a critical driver of Alzheimer’s disease onset and progression. Early hyperphosphorylation of tau alters LC neuronal activity, contributing to prodromal symptoms such as anxiety and hyperarousal. With disease advancement, tau aggregates into neurofibrillary tangles, accelerating LC cell loss and cognitive decline. These findings highlight the close interplay between tau accumulation and LC dysfunction in shaping the trajectory of Alzheimer’s disease.

2.3 High metabolic demand and oxidative stress in LC neurons

Metabolic demands are placed on LC neurons due to their involvement in regulating a significant number of basic physiological functions, including arousal, attention, stress responses, cardiovascular regulation, and sleep–wake cycles, which require sustained spontaneous activity in both normal and stressful conditions. As we explained in part 1–2, LC neurons can adjust their firing across a roughly tenfold range (from less than 1 Hz up to ~5 Hz) depending on behavioral state and external stimuli, allowing flexible modulation of arousal and attention. In stressful situations, firing rate increases or phasic bursts occur to facilitate adaptive behavioral responses. Pacemaker activity, due to the presence of L- and T-type voltage-gated calcium channels, permits maintenance of activity even after the interruption of glutamatergic and GABAergic inputs (Matschke et al., 2015). Consequent high levels of intracellular Ca2+ will, in turn, promote the mitochondrial oxidative stress of these neurons (Sanchez-Padilla et al., 2014). And high levels of oxidative stress make neurons susceptible to DNA damage (Wang and Michaelis, 2010), which is probably one of the causes of the selective vulnerability of LC neurons (Zhan et al., 2019). Also, the activation of the mitochondrial-dependent programmed cell death pathway and caspase 9 enzyme has been reported in the LC neurons of PD and Lewy body dementia (Kawamoto et al., 2014) as well as AD patients (Rohn et al., 2002).

Another metabolic demand on LC neurons is related to their morphology. LC neurons have elongated, diffuse and unmyelinated projections that extend throughout the CNS. Subsequently, these neurons require more metabolic support than neurons with shorter axon lengths and more sparse projections (Sara, 2009). Oxidative stress and neurotoxicity might be more consequential in the LC than in other areas, and therapeutic protection of LC neurons might even be an avenue to delay or mitigate AD onset.

2.4 Widespread proximity of LC neurons to CNS capillaries

Anatomical investigations have revealed that the blood supply of the LC is distinct from most other brain regions except the paraventricular and supraoptic nuclei (Samuels and Szabadi, 2008). The results of an old study conducted in 1940 in non-human primates revealed that the cell body of each LC neuron was surrounded by two or more capillaries (Finley and Cobb, 1940). Additionally, LC neurons send projections to the vast majority of CNS capillaries and the astrocytic end-feet forming the BBB. Considering the total number of LC neurons and the total length of the capillaries in the normal human brain, Pamphlett has hypothesized that each LC neuron innervates 20 m of capillaries on average (Pamphlett, 2014).

These special features enable the LC to maintain the BBB by releasing noradrenaline (NA), which acts on both endothelial cells and astrocytes to regulate barrier integrity, tight junction protein expression, and endothelial pinocytotic activity (Sobral et al., 2025; Verkhratsky et al., 2023). Additionally, NA released by LC neurons can selectively increase blood flow in the most active regions employed during vigilance, supporting higher metabolic demands during vigilance (Bekar et al., 2012). Lastly, additional vulnerabilities for the LC lie in its anatomical location, in the “corner” of the base of the fourth ventricle. This may lead to neuronal exposure to neurotoxins and inflammatory cytokines present in the cerebrospinal fluid stream (Mravec et al., 2014; Zorec et al., 2017).

Because LC neurons are constantly and intensively exposed to blood flow, they are more likely to be affected by potential toxins carried in blood, even at low concentrations, than other areas of the brain. This vulnerability has been hypothesized by Giorgi et al. (2020). Generally, two major groups of toxins can affect the LC nucleus. The first group consists of transfer toxicants that, after passing through the BBB using norepinephrine transporters (NET) located in the axon terminals of LC neurons, enter them. Then they reach the LC neurons’ soma by retrograde transport (Font et al., 1982; Gilsbach et al., 2006). The sensitivity of LC neurons to transfer toxicants depends on the presence of NET that is abundantly expressed in these neurons (Bonisch and Bruss, 2006). DSP4 is a selective neurotoxin for the LC-NA system in rodent and bird brains that selectively destroys LC neurons and is used in laboratory studies to lesion the LC. It was discovered in 1973 that DSP4 can easily cross the BBB. It selectively binds to NET, irreversibly inhibits it, enters the cell and nucleolus, and destroys the axon and cell body (Ross and Stenfors, 2015). It must be mentioned that there is substantial variation in the amount of damage caused to the LC axon terminal relative to the cell bodies in various species (Ross and Stenfors, 2015), though a major contributor to the observed lack of consistency may be due to experimental design (Szot et al., 2010). These differences are not confined to the LC-NA and are also found in the degree to which DSP4 affects the serotonergic system in various species. Yet the specificity to LC-NA over non-coerulear NA terminals persists (Fornai et al., 1996), suggesting a particular vulnerability of the LC-NA system even in species where it is not as pronounced. DSP4-like substances are found in cigarette smoke, car exhaust, and some plants. On the other hand, it has been found that toxins and environmental pollution can reduce LC neuronal NA reuptake (Pamphlett, 2014; Ross and Stenfors, 2015). The second group is intracellular toxins that pass through the BBB like the first group, but they use cotransmitters’ transporters in addition to NET for entering the axon terminals of LC neurons. Also, intracellular toxins can directly enter the cell body of LC neurons using NET in the membrane of the soma (Pamphlett, 2014). Heavy metals such as mercury, lead, silver, and bismuth belong to this group and accumulate inside the cytoplasm of the LC cells. Pamphlett et al. have detected LC neurons filled with environmental toxins and heavy metals with variable concentrations in the human brain (Pamphlett, 2014; Pamphlett and Kum Jew, 2014; Pamphlett et al., 2020), yet in human AD the population of LC neurons enriched in heavy metals were not the same as those enriched in hyperphosphorylated tau (Pamphlett and Kum Jew, 2015). The authors suggest a model by which the toxic metals interact with neuromelanin, discussed below, and the neurofibrillary tangles resulting from hyperphosphorylated tau, as discussed above, both interact to reduce NA transmission and BBB function.

2.5 Neuromelanin

Most LC neurons contain neuromelanin (NM), which is a by-product of catecholamine synthesis oxidation. NM can have both neuroprotective and neurotoxic effects. In natural conditions, NM can chelate environmental toxins, including heavy metals such as iron, cadmium, copper, and mercury, and reduce their toxic effects (Zecca et al., 2008; Zucca et al., 2017). NM also removes excess catecholamines inside the cells (Font et al., 1982). But the stress resulting from aging, disease or the accumulation of toxins prevents the chelating effect of NM and causes toxicity (Giorgi et al., 2020). In this way, since NM granules do not have an enclosed membrane, these accumulated toxins may be released in the cell cytoplasm (Doublea et al., 2008) and upon cell death are released into the extracellular space to propagate neurodegeneration to neighboring neurons (Zucca et al., 2017). Dopaminergic neurons in the substantia nigra also contain NM; however, in AD, the substantia nigra exhibits more resistance to neuronal loss than the LC, while the opposite is seen in PD, where greater neuronal loss in the substantia nigra is found than in the LC nucleus. Therefore, NM cannot be the only vulnerability factor, though it may interact with other factors leading to neurodegeneration in diseases such as AD (Iannitelli and Weinshenker, 2023).

The presence of NM in LC neurons is useful for identifying these cells after death and also in vivo imaging by NM-sensitive magnetic resonance imaging (NM-MRI), which can be correlated with cognitive function (Beardmore et al., 2021; Hämmerer et al., 2018). However, the MRI signal is not exclusively due to NM, as other factors such as lipids, proteins, water, etc. may also contribute.

2.6 Decreased expression of LC co-transmitters and trophic factors

Differences in expression of co-transmitters and receptors in the LC of AD patients have been hypothesized to be either neuroprotective or to expose these cells to vulnerabilities. For instance, though overall LC neurodegeneration was observed in one study, LC neurons co-expressing galanin were seemingly unaffected, as their numbers were roughly the same between AD and normal older human controls. Thus indicating a protective effect of galanin expression or signaling (Miller et al., 1999). On the other hand, a study by Adori and colleagues discovered a reduction of somatostatin receptor function in the LC nucleus of AD patients, which could be involved in the vulnerability of the LC (Ádori et al., 2015).

It has also been observed that the loss of afferent projections to the LC in AD has compounding effects on disease symptoms. For instance, lateral hypothalamic orexin B inputs significantly decreased with age in macaques, and this decrease was significantly correlated with the decrease in the expression of the tyrosine hydroxylase enzyme gene (Downs et al., 2007). This is even more pronounced in human AD (Oh et al., 2019). Indeed, a similar study in orexin (hypocretin)-deficient mice showed that chronic sleep fragmentation-induced Aβ42 accumulation was relatively reduced by the absence of orexin in the hippocampus, but this protective effect was not observed in the LC (Nick et al., 2022). The combined effect of orexinergic and LC neurodegeneration thereby contribute to the characteristic sleep deficiencies in AD (Parhizkar and Holtzman, 2025) which in turn worsen AD progression (Wang and Holtzman, 2020).

It has been determined that the growth and survival of LC neurons depends upon various neurotrophins that can be expressed in LC neurons or provided by retrograde transport from its target areas. The expression of these elements is controlled by NA (Remy et al., 2001). Therefore, changes in the function of the LC or NA levels can affect the expression of the trophic factors. Similarly, retrograde transport of trophic factors is critical for neuronal development and survival (Ito and Enomoto, 2016) and given the diversity of the LC targets, any pathological conditions in these structures might also contribute to LC neurodegeneration.

3 Clinical impact and prognostic value of LC vulnerabilities

According to the World Health Organization, AD and other dementias affect an estimated 55 million people (World Health Organization, n.d.) and somewhere between 69 and 315 million more people may be living with preclinical or prodromal symptoms (Gustavsson et al., 2023). This suggests that a staggering 384 million people may benefit from early diagnosis and prognosis of the disease and underscores the need for accurate, early diagnosis. In the US, Medicare pays for 80% of doctors’ visits, which can amount to around $26,000 per year for some of the newer AD treatments. While early diagnoses may incur more up-front costs (i.e., earlier treatment initiation), economic models suggest that later savings could be achieved by, for instance, contributing to a delay of patient institutionalization (Foldes et al., 2018; Jutkowitz et al., 2023; Weimer and Sager, 2009). Finally, given the similarities of the LC-NA system involvement in certain types of epileptic seizures (Giorgi et al., 2004) and PD (Gesi et al., 2000), advances in the field of AD could also inform clinical practice in a wide array of other diseases.

3.1 Correlation of Alzheimer’s Braak stages with LC dysfunction

Motivated by the observation that, though end-stage postmortem AD had very clear anatomical characteristics, the earlier stages did not, the husband-wife team of Heiko and Eva Braak described a series of morphological stages throughout AD progression in 1991 (Braak and Braak, 1991). These stages are a semiquantitative measure of the severity of tau-based neurofibrillary tangle (NFT), which is the main neuropathological hallmark of AD. They describe the NFT pathology from least to greatest within the medial temporal lobe memory circuit in AD using the Bielschowsky silver stain to visualize NFTs in the frontal, temporal, parietal, and entorhinal cortex, as well as the hippocampus. The Braak stages were based upon the distribution and severity of NFT pathology: Stages I and II indicate NFTs mainly confined to the entorhinal region. Stages III and IV point towards the involvement of limbic regions such as the hippocampus. Stages V and VI signify moderate to severe involvement of the neocortex (Braak and Braak, 1991). According to the Braak stages, LC is the first brain area to show specific alterations, many years before the clinical onset of AD (Braak et al., 2011), involving “pretangle” tau as discussed above. Cassidy et al. investigated the association between LC MRI signals and AD patients’ Braak stage, which was extrapolated from tau PET scans. The findings show the direct relation between LC integrity and Braak stages (Cassidy et al., 2022). Consistent with this, stereological analyses of human postmortem brainstems were performed to estimate LC volume and neuronal population in healthy and AD patients. The results revealed that as the Braak stage increases by 1 unit, the LC volume decreases by 8.4% (Theofilas et al., 2017) and the decline in neural density correlates with cognitive decline (Wilson et al., 2013). Further explorations of AD have disclosed that the accumulation of hyperphosphorylated tau in LC neurons leads to their degeneration as the Braak stages progress (Van Egroo et al., 2023; Jacobs et al., 2022). These are potential implications of LC pathology as an early-stage biomarker for the diagnosis of AD.

3.2 LC degeneration and memory deficits in Alzheimer’s disease

The Braaks were initially hesitant to correlate their anatomical findings with disease symptomatology given the high inter-individual variability, particularly in early stages of the disease. However, the functional connections of the LC with several brain structures involved in learning and memory, including the cortical regions and hippocampus, make this line of inquiry stand out. The decline in noradrenergic transmission is consistent with the cognitive impairment seen in AD. Apart from NA, LC neurons also produce dopamine, and they can corelease both catecholamines to influence synaptic plasticity and memory in the hippocampus (Ciampa et al., 2022). In vivo research on the integrity of the LC consistently shows an association with memory deficits (Jacobs et al., 2022; Dahl et al., 2022). Dahl et al. have recently put forth the hypothesis, based on rodent (Dahl et al., 2023b) and human (Dahl et al., 2023a) studies, that the declining catecholaminergic input to the hippocampus in both normal and AD aging leads to decreased synaptic plasticity and subsequent decline in memory performance.

3.3 The need for earlier Alzheimer’s diagnoses

A new class of AD medications, monoclonal antibodies directed against Aβ, has shown limited delay of disease progression in ~30% of patients (Boxer and Sperling, 2023). This may be due to the clinical trial inclusion criteria: participants needed to show clinical symptoms and signs of deteriorating cognitive performance. Since accelerated Aβ deposition and initial neurodegeneration is likely to have begun up to 10 years prior to symptom onset, preventive interventions rather than mitigation of disease progression has been proposed (Jucker and Walker, 2023). Longitudinal studies in preclinical / asymptomatic subjects, such as the AHEAD 3–45, are already underway (Rafii et al., 2023). Thus, the need for early AD diagnosis was also highlighted in a US Food and Drug Administration guidance for industry proposal (Food and Drug Administration, n.d.).

Prognoses and interventions beginning long before the onset of symptoms could also be directed at processes influenced by the LC such as the sleep/wake cycle. Sleep EEG is a potential biomarker of the relative “brain age” of individuals with the same chronological age (Sun et al., 2019). Chronic sleep disruption is a risk factor for AD (Parhizkar and Holtzman, 2025; Wang and Holtzman, 2020; Bubu et al., 2017; Himali et al., 2023; Neylan and Walsh, 2024), and prolonged sleep durations may protect cognitive function in the prodromal stage of the disease in some prospective AD patients (Baril et al., 2024), which is a high-priority area of research (Andre et al., 2025). Possibly, a combination of AD biomarkers in conjunction with circadian and sleep measures could even assess individuals’ probability of progressing to AD (Yang et al., 2024). In light of this, it is interesting to speculate whether the sleep disruption could be caused by prodromal LC hyperactivity (Mayer et al., 2024). Nocturnal awakenings are associated with decreases in LC integrity as measured by MRI in AD patients (Van Egroo et al., 2021), suggesting that longitudinal studies of LC integrity, sleep/wake alterations prior to AD symptom onset may offer early prognostic value (Van Egroo et al., 2022). In later stages, so-called “sundowning,” occurs in ~20–45% of people with AD and has been linked to LC neurodegeneration (Yang et al., 2024; Garcia-Rill, 2019; Oh et al., 2022), and hyperactivity (Fitzgerald, 2021). Counteracting sleep loss itself could even one day be used to delay disease onset. The consensus view is that while Aβ and tau accumulate in the interstitial fluid during wakefulness, they are cleared during the deepest stage of non-REM sleep, slow-wave sleep (Holth et al., 2019), and that this precedes cognitive decline and is related to autonomic regulation (Chen et al., 2023). The neuroprotective role of non-REM sleep in AD pathology has also been confirmed in humans in both correlational (Zavecz et al., 2023) and pharmacological (Lucey et al., 2023; Zhou and Tang, 2022) studies, further opening up the possibility of shunting the deleterious effects of LC overactivation on Aβ removal during slow-wave sleep (La et al., 2019; Ashford, 2019), prior to symptom onset.

While currently available blood biomarkers provide fast, inexpensive and accurate test results for those with MCI (Li et al., 2022; Brand et al., 2022; Ashton et al., 2024; Barthelemy et al., 2024), the sensitivity and specificity of these tests are often considerably lower in presymptomatic individuals. On the other hand, there has been tremendous progress recently in advancing LC imaging in vivo in humans as a way to diagnose neurodegeneration and earlier AD prognosis (Kelberman et al., 2020). The potential for the longitudinal prediction of future cognitive abilities based on catecholaminergic size in MRI has recently been demonstrated (Dahl et al., 2023a) and thus posits that the LC is not only a target of AD symptom intervention but also an early diagnostic/prognostic center for the development of AD (Dahl et al., 2023b). In the following section, we review LC imaging in relation to AD diagnosis.

3.4 Can LC imaging improve Alzheimer’s diagnoses?

Structural MRI scans with higher resolution are minimally invasive and widely available compared to other techniques such as PET, so appear to be an ideal method to study the LC. Aggregation of hyperphosphorylated tau protein results in reduced LC integrity in the neurodegenerative disease state (Jacobs et al., 2022; Dahl et al., 2022). Betts and colleagues showed that lower LC contrast, in the rostral and middle thirds of the LC, is correlated with CSF Aβ levels (Betts et al., 2019; Betts et al., 2019). These findings are in agreement with the Dahl et al. study highlighting the importance of rostral parts of the LC in memory performance (Dahl et al., 2019). Structural covariance of the LC with the whole brain is also correlated to subjective and objective memory loss (Fu et al., 2021; Tang et al., 2023). Together, this suggests that LC contrast studies could be used as a diagnostic tool to assess the AD onset and progression.

It should be noted that LC imaging parameters are also negatively correlated with normal aging (Betts et al., 2017) and aging-related behavioral changes (Liu et al., 2019; Liu et al., 2020) as well as brain correlates of cognition in later life (Bachman et al., 2021). Though when comparing to subjects who later display cognitive decline, no differences are noted in the cohort of cognitively intact individuals (Giorgi et al., 2022). Nevertheless, the specific alterations seen in AD patients’ LC imaging correlate well with posthumous LC cell counts (Theofilas et al., 2017; Betts et al., 2019; Keren et al., 2015).

Diffusion tensor imaging provides a detailed, microstructural view of the brain by quantifying the diffusion of water and other fluids, allowing imaging of axon fibers (Basser et al., 1994). Using DTI we can obtain more information about LC axonal dysfunctions such as myelination, integrity or size of wide and diffuse projections of the LC to its target regions (Kelberman et al., 2020; Lenglet, 2015). Moreover, DTI can reliably detect differences in the integrity of the LC projections related to resilience to sleep deprivation (Quan et al., 2024) and ageing (Svendsen et al., 1986). Recently, a reduction in LC integrity was found in older individuals, and this was correlated with a decline in memory performance (Langley et al., 2020). And a reduction in fiber integrity of the LC cortical projections was found in AD patients (Sun et al., 2020), which is likely due to LC cell loss rather than fiber denervation (Lin et al., 2024; Quattrini et al., 2024). As advances in resolution continue to be made, for instance via MR relaxometry (Bae et al., 2025), early microstructural changes in the LC are becoming a reality.

Functional MRI (fMRI) displays regional and temporal changes in brain activity through changes in blood flow, though it is complicated for a variety of reasons. First, since LC neurons fire using two separate patterns, tonic and phasic, each may recruit specific functional connectivities. In addition, these neurons do not always fire synchronously. Therefore, attention should be paid to technical analysis considerations when using LC fMRI (Kelberman et al., 2020). A recent advance in imaging resolution and data analysis first developed using structural MRI (Hämmerer et al., 2018) and then fMRI (Ludwig et al., 2024), with the former study showing a correlation between older age LC integrity and salient memories, and the latter study showing age-related increases in LC activity during learning tasks. Another recent study has determined “LC-to-pons” parameters that reliably distinguish between AD and age-matched controls (Dordevic et al., 2017), and this “T1-weighted turbo spin echo” scan approach (Takahashi et al., 2015) has been shown to be longitudinally reliable as well (Tona et al., 2017). Beyond that, and contrary to the idea that LC-NA connection with the striatum is weak, resting-state fMRI has revealed that LC connectivity with the salience networks in older people declines (Lee et al., 2020; Liebe et al., 2020). Functional MRI studies have revealed that a stronger connection between LC and the parahippocampal gyrus is associated with better memory performance (Sara, 2009; Jacobs et al., 2015). Finally, ultra-high resolution 7 T fMRI has shown much promise in overcoming the size limitations of LC imaging. Studies of LC functional connectivity (FC) in ageing have shown linear decreases in FC between the LC and some areas, whereas non-linear relationships exist with others (Jacobs et al., 2018), an association of emotional memory and emotion regulation along the rostro-caudal extent of the LC (Vereb et al., 2023), and a slower memory decline in individuals ageing with higher memory capacity (Mijalkov et al., 2025).

Positron Emission Tomography (PET) visualizes metabolic and other physiological activity using radiotracers. PET is associated with greater costs and lower resolution compared to structural MRI, but its advantage is that it can be used to measure LC dysfunction in the terminal regions. Norepinephrine transporters, which are expressed in all parts of LC cells, can be assessed with PET in regions innervated by the LC to evaluate the LC fiber integrity (Arakawa et al., 2008; Brumberg et al., 2019; Chen et al., 2020; Ding et al., 2006; Moriguchi et al., 2017). Significant interactions of tau tracers with neuromelanin make direct PET tau imaging of the LC currently unadvisable (Betts et al., 2019; Krohn et al., 2023; Marquie et al., 2015). Also, using the metabolic activity of the LC as measured via [18F]fluorodeoxyglucose (FDG) as a biomarker for Alzheimer’s is still somewhat uncertain in that one study found no correlation (Liu et al., 2021), though another found increased activity in the preclinical stages correlated with attenuated memory decline (Koops et al., 2025). Importantly, however, lower LC integrity as measured by LC-MRI is correlated with greater cortical tau pathology as measured by PET, as well A-β and cognitive measures (Jacobs et al., 2022), and lower regional cortical glucose metabolism as measured by [18F]FDG in individuals with dementia and MCI (Aghakhanyan et al., 2023), and this is correlated with cognitive performance in patients with AD (Khosravi et al., 2019).

4 Conclusion

The unique neuronal architecture and morphology, physiology, activity and copious ascending and descending projections allow the LC to be involved in a wide variety of functions such as sleep and wakefulness, the stress response, attention, memory and sensory processing, etc. This also exposes the LC to vulnerabilities leading to neurodegeneration and propagation of neurodegenerative species to other brain areas. It is clear that the LC is one of the earliest affected areas in Alzheimer’s disease, and contemporary imaging techniques are now able to detect these foundational changes. These insights will be leveraged into expanded fundamental knowledge of AD initiation and progression, and further refinement should lead to earlier disease detection and/or treatment and lifestyle modification options.

Many factors can play a role in the damage of LC neurons, which show a decline during aging that can be correlated to the increased activity of microglia, the accumulation of phosphorylated tau protein, the accumulation of environmental toxins, and the reduction of trophic factors from the target areas of the LC. Therefore, additional damage to this nucleus during disease progression can be the result of the aggravation of these natural processes. This exacerbation may be the result of pathological and inflammatory events in the target areas of the LC nucleus, which reduce the production of trophic factors and sometimes produce toxic substances. On the other hand, toxins act selectively on LC neurons. LC damage has also been observed in many laboratory models of neurodegenerative diseases and large-scale animal studies are being conducted to understand the dimensions of these effects. In medicine, translational brain imaging studies are a big step in this field.

There are some caveats to note. Brain imaging is a very expensive and somewhat intrusive endeavor. While there is some hope that over time, techniques will be refined and standardized in a way that will lower costs and enhance access (Engels-Dominguez et al., 2024), the high fixed costs of imaging will likely not make any of these techniques routine outside of research settings anytime soon, if ever. Nonetheless, the demand for early testing—decades prior to symptom onset—amongst the general population is exceedingly high according to a recent Alzheimer’s Association survey (Alzheimer's Association Report, 2025). Imaging of the LC, with the early signs of AD reviewed here, can assist in this. Most of the studies cited in this review article state that longitudinal studies, and the combination of structural and functional imaging are needed to understand the progression of LC damage in AD better, sentiments echoed in a prior review article (Engels-Dominguez et al., 2023). If costs could be reduced and resolution increased with future advanced technologies, criteria could be developed to triage subjects for imaging in a wider population.

Author contributions

FF: Conceptualization, Writing – original draft, Writing – review & editing. CA: Conceptualization, Writing – review & editing. HA: Conceptualization, Writing – original draft, Writing – review & editing. HG: Conceptualization, Writing – original draft, Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Conflict of interest

The 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.

The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision.

Generative AI statement

The authors declare that no Gen AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

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.

References

Ádori, C., Glück, L., Barde, S., Yoshitake, T., Kovacs, G. G., Mulder, J., et al. (2015). Critical role of somatostatin receptor 2 in the vulnerability of the central noradrenergic system: new aspects on Alzheimer’s disease. Acta Neuropathol. 129, 541–563. doi: 10.1007/s00401-015-1394-3

PubMed Abstract | Crossref Full Text | Google Scholar

Aghakhanyan, G., Galgani, A., Vergallo, A., Lombardo, F., Martini, N., Baldacci, F., et al. (2023). Brain metabolic correlates of locus coeruleus degeneration in Alzheimer's disease: a multimodal neuroimaging study. Neurobiol. Aging 122, 12–21. doi: 10.1016/j.neurobiolaging.2022.11.002

PubMed Abstract | Crossref Full Text | Google Scholar

Ahmadi-Soleimani, S. M., Mianbandi, V., Azizi, H., Azhdari-Zarmehri, H., Ghaemi-Jandabi, M., Abbasi-Mazar, A., et al. (2020). Coregulation of sleep-pain physiological interplay by orexin system: an unprecedented review. Behav. Brain Res. 391:112650. doi: 10.1016/j.bbr.2020.112650

PubMed Abstract | Crossref Full Text | Google Scholar

Alaee, E., Farahani, F., Semnanian, S., and Azizi, H. (2022). Prenatal exposure to morphine enhances excitability in locus coeruleus neurons. J. Neural Transm. 129, 1049–1060. doi: 10.1007/s00702-022-02515-3

PubMed Abstract | Crossref Full Text | Google Scholar

Alzheimer's Association Report (2025). 2025 Alzheimer's disease facts and figures. Alzheimers Dement. 21:e70235. doi: 10.1002/alz.70235

PubMed Abstract | Crossref Full Text | Google Scholar

Andre, C., Stankeviciute, L., Michaelian, J. C., Antonsdottir, I. M., Benca, R. M., Coulthard, E. J., et al. (2025). International recommendations for sleep and circadian research in aging and Alzheimer's disease: a Delphi consensus study. Alzheimers Dement. 21:e70742. doi: 10.1002/alz.70742

PubMed Abstract | Crossref Full Text | Google Scholar

Arakawa, R., Okumura, M., Ito, H., Seki, C., Takahashi, H., Takano, H., et al. (2008). Quantitative analysis of norepinephrine transporter in the human brain using PET with (S,S)-18F-FMeNER-D2. J. Nucl. Med. 49, 1270–1276. doi: 10.2967/jnumed.108.051292

Crossref Full Text | Google Scholar

Arnsten, A. F. T., Wang, M. J., and Paspalas, C. D. (2012). Neuromodulation of thought: flexibilities and vulnerabilities in prefrontal cortical network synapses. Neuron 76, 223–239. doi: 10.1016/j.neuron.2012.08.038

PubMed Abstract | Crossref Full Text | Google Scholar

Ashford, J. W. (2019). Treatment of Alzheimer's disease: trazodone, sleep, serotonin, norepinephrine, and future directions. J. Alzheimers Dis. 67, 923–930. doi: 10.3233/JAD-181106

PubMed Abstract | Crossref Full Text | Google Scholar

Ashton, N. J., Brum, W. S., Di Molfetta, G., Benedet, A. L., Arslan, B., Jonaitis, E., et al. (2024). Diagnostic accuracy of a plasma phosphorylated tau 217 immunoassay for Alzheimer disease pathology. JAMA Neurol. 81, 255–263. doi: 10.1001/jamaneurol.2023.5319

PubMed Abstract | Crossref Full Text | Google Scholar

Aston-Jones, G., and Cohen, J. D. (2005). An integrative theory of locus coeruleus-norepinephrine function: adaptive gain and optimal performance. Annu. Rev. Neurosci. 28, 403–450. doi: 10.1146/annurev.neuro.28.061604.135709

PubMed Abstract | Crossref Full Text | Google Scholar

Aston-Jones, G., and Waterhouse, B. (2016). Locus coeruleus: from global projection system to adaptive regulation of behavior. Brain Res. 1645, 75–78. doi: 10.1016/j.brainres.2016.03.001

PubMed Abstract | Crossref Full Text | Google Scholar

Bachman, S. L., Dahl, M. J., Werkle-Bergner, M., Duzel, S., Forlim, C. G., Lindenberger, U., et al. (2021). Locus coeruleus MRI contrast is associated with cortical thickness in older adults. Neurobiol. Aging 100, 72–82. doi: 10.1016/j.neurobiolaging.2020.12.019

PubMed Abstract | Crossref Full Text | Google Scholar

Bachman, D., and Rabins, P. (2006). "Sundowning" and other temporally associated agitation states in dementia patients. Annu. Rev. Med. 57, 499–511. doi: 10.1146/annurev.med.57.071604.141451

PubMed Abstract | Crossref Full Text | Google Scholar

Bachmann, S., Bell, M., Klimek, J., and Zempel, H. (2021). Differential effects of the six human TAU isoforms: somatic retention of 2N-TAU and increased microtubule number induced by 4R-TAU. Front. Neurosci. 15:643115. doi: 10.3389/fnins.2021.643115

PubMed Abstract | Crossref Full Text | Google Scholar

Bae, J., Gong, Z., Mazucanti, C., Bilgel, M., Laporte, J. P., Faulkner, M. E., et al. (2025). Age-related differences in rostral-middle locus coeruleus microstructure: a critical role in cognitive decline revealed by magnetic resonance relaxometry. Alzheimers Res. Ther. 17:161. doi: 10.1186/s13195-025-01809-4

PubMed Abstract | Crossref Full Text | Google Scholar

Ballard, C., and Corbett, A. (2013). Agitation and aggression in people with Alzheimer's disease. Curr. Opin. Psychiatry 26, 252–259. doi: 10.1097/YCO.0b013e32835f414b

PubMed Abstract | Crossref Full Text | Google Scholar

Barbier, P., Zejneli, O., Martinho, M., Lasorsa, A., Belle, V., Smet-Nocca, C., et al. (2019). Role of tau as a microtubule-associated protein: structural and functional aspects. Front. Aging Neurosci. 11:204. doi: 10.3389/fnagi.2019.00204

PubMed Abstract | Crossref Full Text | Google Scholar

Bardou, I., Kaercher, R. M., Brothers, H. M., Hopp, S. C., Royer, S., and Wenk, G. L. (2014). Age and duration of inflammatory environment differentially affect the neuroimmune response and catecholaminergic neurons in the midbrain and brainstem. Neurobiol. Aging 35, 1065–1073. doi: 10.1016/j.neurobiolaging.2013.11.006

PubMed Abstract | Crossref Full Text | Google Scholar

Baril, A. A., Picard, C., Labonte, A., Sanchez, E., Duclos, C., Mohammediyan, B., et al. (2024). Longer sleep duration and neuroinflammation in at-risk elderly with a parental history of Alzheimer's disease. Sleep 47:zsae081. doi: 10.1093/sleep/zsae081

PubMed Abstract | Crossref Full Text | Google Scholar

Barsegyan, A., McGaugh, J. L., and Roozendaal, B. (2014). Noradrenergic activation of the basolateral amygdala modulates the consolidation of object-in-context recognition memory. Front. Behav. Neurosci. 8, 1–8. doi: 10.3389/fnbeh.2014.00160

PubMed Abstract | Crossref Full Text | Google Scholar

Barthelemy, N. R., Salvado, G., Schindler, S. E., He, Y. X., Janelidze, S., Collij, L. E., et al. (2024). Highly accurate blood test for Alzheimer's disease is similar or superior to clinical cerebrospinal fluid tests. Nat. Med. 30, 1085–1095. doi: 10.1038/s41591-024-02869-z

PubMed Abstract | Crossref Full Text | Google Scholar

Basser, P. J., Mattiello, J., and LeBihan, D. (1994). MR diffusion tensor spectroscopy and imaging. Biophys. J. 66, 259–267. doi: 10.1016/S0006-3495(94)80775-1

PubMed Abstract | Crossref Full Text | Google Scholar

Beardmore, R., Hou, R., Darekar, A., Holmes, C., and Boche, D. (2021). The locus Coeruleus in aging and Alzheimer's disease: a postmortem and brain imaging review. J. Alzheimers Dis. 83, 5–22. doi: 10.3233/JAD-210191

PubMed Abstract | Crossref Full Text | Google Scholar

Bekar, L. K., Wei, H. S., and Nedergaard, M. (2012). The locus coeruleus-norepinephrine network optimizes coupling of cerebral blood volume with oxygen demand. J. Cereb. Blood Flow Metab. 32, 2135–2145. doi: 10.1038/jcbfm.2012.115

PubMed Abstract | Crossref Full Text | Google Scholar

Benarroch, E. E. (2009). The locus ceruleus norepinephrine system: functional organization and potential clinical significance. Neurology 73, 1699–1704. doi: 10.1212/WNL.0b013e3181c2937c

PubMed Abstract | Crossref Full Text | Google Scholar

Benarroch, E. E. (2018). Locus coeruleus. Cell Tissue Res. 373, 221–232. doi: 10.1007/s00441-017-2649-1

PubMed Abstract | Crossref Full Text | Google Scholar

Berridge, C. W., and Waterhouse, B. D. (2003). The locus coeruleus – noradrenergic system: modulation of behavioral state and state-dependent cognitive processes. Brain Res. Rev. 42, 33–84. doi: 10.1016/s0165-0173(03)00143-7

PubMed Abstract | Crossref Full Text | Google Scholar

Betts, M. J., Cardenas-Blanco, A., Kanowski, M., Jessen, F., and Duzel, E. (2017). In vivo MRI assessment of the human locus coeruleus along its rostrocaudal extent in young and older adults. NeuroImage 163, 150–159. doi: 10.1016/j.neuroimage.2017.09.042

PubMed Abstract | Crossref Full Text | Google Scholar

Betts, M. J., Cardenas-Blanco, A., Kanowski, M., Spottke, A., Teipel, S. J., Kilimann, I., et al. (2019). Locus coeruleus MRI contrast is reduced in Alzheimer's disease dementia and correlates with CSF aβ levels. Alzheimers Dement. (N Y). 11, 281–285. doi: 10.1016/j.dadm.2019.02.001

PubMed Abstract | Crossref Full Text | Google Scholar

Betts, M. J., Kirilina, E., Otaduy, M. C. G., Ivanov, D., Acosta-Cabronero, J., Callaghan, M. F., et al. (2019). Locus coeruleus imaging as a biomarker for noradrenergic dysfunction in neurodegenerative diseases. Brain 142, 2558–2571. doi: 10.1093/brain/awz193

PubMed Abstract | Crossref Full Text | Google Scholar

Biagioni, F., Ferrucci, M., Lazzeri, G., Scioli, M., Frati, A., Puglisi-Allegra, S., et al. (2024). Damage to the locus coeruleus alters the expression of key proteins in limbic neurodegeneration. Int. J. Mol. Sci. 25:3159. doi: 10.3390/ijms25063159

PubMed Abstract | Crossref Full Text | Google Scholar

Bielau, H., Brisch, R., Bernard-Mittelstaedt, J., Dobrowolny, H., Gos, T., Baumann, B., et al. (2012). Immunohistochemical evidence for impaired nitric oxide signaling of the locus coeruleus in bipolar disorder. Brain Res. 1459, 91–99. doi: 10.1016/j.brainres.2012.04.022

PubMed Abstract | Crossref Full Text | Google Scholar

Bondareff, W., Mountjoy, C. Q., Roth, M., Rossor, M. N., Iversen, L. L., Reynolds, G. P., et al. (1987). Neuronal degeneration in locus ceruleus and cortical correlates of Alzheimer disease. Alzheimer Dis. Assoc. Disord. 1, 256–262. doi: 10.1097/00002093-198701040-00005

PubMed Abstract | Crossref Full Text | Google Scholar

Bonisch, H., and Bruss, M. (2006). The norepinephrine transporter in physiology and disease. Handb. Exp. Pharmacol. 175, 485–524. doi: 10.1007/3-540-29784-7_20

PubMed Abstract | Crossref Full Text | Google Scholar

Boxer, A. L., and Sperling, R. (2023). Accelerating Alzheimer's therapeutic development: the past and future of clinical trials. Cell 186, 4757–4772. doi: 10.1016/j.cell.2023.09.023

PubMed Abstract | Crossref Full Text | Google Scholar

Braak, H., and Braak, E. (1991). Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 82, 239–259. doi: 10.1007/BF00308809

PubMed Abstract | Crossref Full Text | Google Scholar

Braak, H., Thal, D. R., and Del Tredici, K. (2011). Stages of the pathologic process in Alzheimer disease: age categories from 1 to 100 years. J. Neuropathol. Exp. Neurol. 70, 960–969. doi: 10.1097/NEN.0b013e318232a379

PubMed Abstract | Crossref Full Text | Google Scholar

Brand, A. L., Lawler, P. E., Bollinger, J. G., Li, Y., Schindler, S. E., Li, M., et al. (2022). The performance of plasma amyloid beta measurements in identifying amyloid plaques in Alzheimer’s disease: a literature review. Alzheimers Res. Therapy. 14, 1–15. doi: 10.1186/s13195-022-01117-1

PubMed Abstract | Crossref Full Text | Google Scholar

Brumberg, J., Tran-Gia, J., Lapa, C., Isaias, I. U., and Samnick, S. (2019). PET imaging of noradrenaline transporters in Parkinson's disease: focus on scan time. Ann. Nucl. Med. 33, 69–77. doi: 10.1007/s12149-018-1305-5

PubMed Abstract | Crossref Full Text | Google Scholar

Bubu, O. M., Brannick, M., Mortimer, J., Umasabor-bubu, O., Sebastião, Y. V., Wen, Y., et al. (2017). Sleep, cognitive impairment, and Alzheimer’s disease: a systematic review and meta-analysis. Sleep 40, 1–18. doi: 10.1093/sleep/zsw032

PubMed Abstract | Crossref Full Text | Google Scholar

Bueicheku, E., Diez, I., Kim, C. M., Becker, J. A., Koops, E. A., Kwong, K., et al. (2024). Spatiotemporal patterns of locus coeruleus integrity predict cortical tau and cognition. Nat. Aging 4, 625–637. doi: 10.1038/s43587-024-00626-y

PubMed Abstract | Crossref Full Text | Google Scholar

Cassidy, C. M., Therriault, J., Pascoal, T. A., Cheung, V., Savard, M., Tuominen, L., et al. (2022). Association of locus coeruleus integrity with Braak stage and neuropsychiatric symptom severity in Alzheimer’s disease. Neuropsychopharmacology 47, 1128–1136. doi: 10.1038/s41386-022-01293-6

PubMed Abstract | Crossref Full Text | Google Scholar

Chandler, D. J., Gao, W.-J., and Waterhouse, B. D. (2014). Heterogeneous organization of the locus coeruleus projections to prefrontal and motor cortices. Proc. Natl. Acad. Sci. USA 111, 6816–6821. doi: 10.1073/pnas.1320827111

PubMed Abstract | Crossref Full Text | Google Scholar

Chandler, D. J., Jensen, P., McCall, J. G., Pickering, A. E., Schwarz, L. A., and Totah, N. K. (2019). Redefining noradrenergic neuromodulation of behavior: impacts of a modular locus coeruleus architecture. J. Neurosci. 39, 8239–8249. doi: 10.1523/JNEUROSCI.1164-19.2019

PubMed Abstract | Crossref Full Text | Google Scholar

Chandley, M. J., Szebeni, K., Szebeni, A., Crawford, J., Stockmeier, C. A., Turecki, G., et al. (2013). Gene expression deficits in pontine locus coeruleus astrocytes in men with major depressive disorder. J. Psychiatry Neurosci. 38, 276–284. doi: 10.1503/jpn.120110

PubMed Abstract | Crossref Full Text | Google Scholar

Chan-Palay, V., and Asan, E. (1989). Quantitation of catecholamine neurons in the locus coeruleus in human brains of normal young and older adults and in depression. J. Comp. Neurol. 287, 357–372. doi: 10.1002/cne.902870307

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, Y., Chen, T., and Hou, R. (2022). Locus coeruleus in the pathogenesis of Alzheimer's disease: a systematic review. Alzheimers Dement. (N Y). 8:e12257. doi: 10.1002/trc2.12257

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, X., Kudo, T., Lapa, C., Buck, A., and Higuchi, T. (2020). Recent advances in radiotracers targeting norepinephrine transporter: structural development and radiolabeling improvements. J. Neural Transm. (Vienna) 127, 851–873. doi: 10.1007/s00702-020-02180-4

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, C. W., Kwok, Y. T., Cheng, Y. T., Huang, Y. S., Kuo, T. B. J., Wu, C. H., et al. (2023). Reduced slow-wave activity and autonomic dysfunction during sleep precede cognitive deficits in Alzheimer’s disease transgenic mice. Sci. Rep. 13, 1–17. doi: 10.1038/s41598-023-38214-6

PubMed Abstract | Crossref Full Text | Google Scholar

Ciampa, C. J., Parent, J. H., Harrison, T. M., Fain, R. M., Betts, M. J., Maass, A., et al. (2022). Associations among locus coeruleus catecholamines, tau pathology, and memory in aging. Neuropsychopharmacology 47, 1106–1113. doi: 10.1038/s41386-022-01269-6

PubMed Abstract | Crossref Full Text | Google Scholar

Clewett, D. V., Lee, T.-h., Greening, S., Ponzio, A., Margalit, E., and Mather, M. (2016). Neurobiology of aging neuromelanin marks the spot: identifying a locus coeruleus biomarker of cognitive reserve in healthy aging. Neurobiol. Aging 37, 117–126. doi: 10.1016/j.neurobiolaging.2015.09.019

PubMed Abstract | Crossref Full Text | Google Scholar

Cohen, R. M., Rezai-Zadeh, K., Weitz, T. M., Rentsendorj, A., Gate, D., Spivak, I., et al. (2013). A transgenic Alzheimer rat with plaques, tau pathology, behavioral impairment, oligomeric abeta, and frank neuronal loss. J. Neurosci. 33, 6245–6256. doi: 10.1523/JNEUROSCI.3672-12.2013

Crossref Full Text | Google Scholar

Dahl, M. J., Bachman, S. L., Dutt, S., Düzel, S., Bodammer, N. C., Lindenberger, U., et al. (2023a). The integrity of dopaminergic and noradrenergic brain regions is associated with different aspects of late-life memory performance. Nature Aging. 3, 1128–1143. doi: 10.1038/s43587-023-00469-z

PubMed Abstract | Crossref Full Text | Google Scholar

Dahl, M. J., Kulesza, A., Werkle-Bergner, M., and Mather, M. (2023b). Declining locus coeruleus–dopaminergic and noradrenergic modulation of long-term memory in aging and Alzheimer’s disease. Neurosci. Biobehav. Rev. 153:105358. doi: 10.1016/j.neubiorev.2023.105358

PubMed Abstract | Crossref Full Text | Google Scholar

Dahl, M. J., Mather, M., Düzel, S., Bodammer, N. C., Lindenberger, U., Kühn, S., et al. (2019). Rostral locus coeruleus integrity is associated with better memory performance in older adults. Nat. Hum. Behav. 3, 1203–1214. doi: 10.1038/s41562-019-0715-2

PubMed Abstract | Crossref Full Text | Google Scholar

Dahl, M. J., Mather, M., Werkle-Bergner, M., Kennedy, B. L., Guzman, S., Hurth, K., et al. (2022). Locus coeruleus integrity is related to tau burden and memory loss in autosomal-dominant Alzheimer's disease. Neurobiol. Aging 112, 39–54. doi: 10.1016/j.neurobiolaging.2021.11.006

PubMed Abstract | Crossref Full Text | Google Scholar

Dahlström, A., and Fuxe, K. (1964). Localization of monoamines in the lower brain stem. Experientia 20, 398–399. doi: 10.1007/BF02147990

PubMed Abstract | Crossref Full Text | Google Scholar

Dai, M., Guo, Z., Chen, J., Liu, H., Li, J., Zhu, M., et al. (2023). Altered functional connectivity of the locus coeruleus in Alzheimer's disease patients with depression symptoms. Exp. Gerontol. 179:112252. doi: 10.1016/j.exger.2023.112252

PubMed Abstract | Crossref Full Text | Google Scholar

David, M. C. B., Del Giovane, M., Liu, K. Y., Gostick, B., Rowe, J. B., Oboh, I., et al. (2022). Cognitive and neuropsychiatric effects of noradrenergic treatment in Alzheimer's disease: systematic review and meta-analysis. J. Neurol. Neurosurg. Psychiatry 93, 1080–1090. doi: 10.1136/jnnp-2022-329136

PubMed Abstract | Crossref Full Text | Google Scholar

de Carvalho, D., Patrone, L. G., Taxini, C. L., Biancardi, V., Vicente, M. C., and Gargaglioni, L. H. (2014). Neurochemical and electrical modulation of the locus coeruleus: contribution to CO2drive to breathe. Front. Physiol. 5:288. doi: 10.3389/fphys.2014.00288

PubMed Abstract | Crossref Full Text | Google Scholar

Ding, Y. S., Lin, K. S., and Logan, J. (2006). PET imaging of norepinephrine transporters. Curr. Pharm. Des. 12, 3831–3845. doi: 10.2174/138161206778559687

PubMed Abstract | Crossref Full Text | Google Scholar

Dordevic, M., Muller-Fotti, A., Muller, P., Schmicker, M., Kaufmann, J., and Muller, N. G. (2017). Optimal cut-off value for locus Coeruleus-to-pons intensity ratio as clinical biomarker for Alzheimer's disease: a pilot study. J. Alzheimers Dis. Rep. 1, 159–167. doi: 10.3233/ADR-170021

PubMed Abstract | Crossref Full Text | Google Scholar

Doublea, K. L., Dedova, V. N., Fedorowb, H., Kettlea, E., Hallidaya, G. M., Garnera, B., et al. (2008). Review the comparative biology of neuromelanin and lipofuscin in the human brain. Cell. Mol. Life Sci. 65, 1669–1682.

Google Scholar

Downs, J. L., Dunn, M. R., Borok, E., Shanabrough, M., Horvath, T. L., Kohama, S. G., et al. (2007). Orexin neuronal changes in the locus coeruleus of the aging rhesus macaque. Neurobiol. Aging 28, 1286–1295. doi: 10.1016/j.neurobiolaging.2006.05.025

PubMed Abstract | Crossref Full Text | Google Scholar

Ehrenberg, A. J., Kelberman, M. A., Liu, K. Y., Dahl, M. J., Weinshenker, D., Falgas, N., et al. (2023). Priorities for research on neuromodulatory subcortical systems in Alzheimer's disease: position paper from the NSS PIA of ISTAART. Alzheimers Dement. 19, 2182–2196. doi: 10.1002/alz.12937

PubMed Abstract | Crossref Full Text | Google Scholar

Ehrenberg, A. J., Nguy, A. K., Theofilas, P., Dunlop, S., Suemoto, C. K., Di Lorenzo Alho, A. T., et al. (2017). Quantifying the accretion of hyperphosphorylated tau in the locus coeruleus and dorsal raphe nucleus: the pathological building blocks of early Alzheimer's disease. Neuropathol. Appl. Neurobiol. 43, 393–408. doi: 10.1111/nan.12387

PubMed Abstract | Crossref Full Text | Google Scholar

Engels-Dominguez, N., Koops, E. A., Prokopiou, P. C., Van Egroo, M., Schneider, C., Riphagen, J. M., et al. (2023). State-of-the-art imaging of neuromodulatory subcortical systems in aging and Alzheimer's disease: challenges and opportunities. Neurosci. Biobehav. Rev. 144:104998. doi: 10.1016/j.neubiorev.2022.104998

PubMed Abstract | Crossref Full Text | Google Scholar

Engels-Dominguez, N., Riphagen, J. M., Van Egroo, M., Koops, E. A., Smegal, L. F., Becker, J. A., et al. (2024). Lower locus Coeruleus integrity signals elevated entorhinal tau and clinical progression in asymptomatic older individuals. Ann. Neurol. 96, 650–661. doi: 10.1002/ana.27022

PubMed Abstract | Crossref Full Text | Google Scholar

Farahani, F., Azizi, H., Janahmadi, M., Seutin, V., and Semnanian, S. (2021). Formalin-induced inflammatory pain increases excitability in locus coeruleus neurons. Brain Res. Bull. 172, 52–60. doi: 10.1016/j.brainresbull.2021.04.002

PubMed Abstract | Crossref Full Text | Google Scholar

Farahani, F., Pachenari, N., Mohammad Ahmadi-Soleimani, S., Azizi, H., and Semnanian, S. (2023). Acute morphine injection persistently affects the electrophysiological characteristics of rat locus coeruleus neurons. Neurosci. Lett. 795:137048. doi: 10.1016/j.neulet.2023.137048

PubMed Abstract | Crossref Full Text | Google Scholar

Feinstein, D. L., Kalinin, S., and Braun, D. (2016). Causes, consequences, and cures for neuroinflammation mediated via the locus coeruleus: noradrenergic signaling system. J. Neurochem. 139, 154–178. doi: 10.1111/jnc.13447

PubMed Abstract | Crossref Full Text | Google Scholar

Fernandes, P., Regala, J., Correia, F., and Gonçalves-Ferreira, A. J. (2012). The human locus coeruleus 3-D stereotactic anatomy. Surg. Radiol. Anat. 34, 879–885. doi: 10.1007/s00276-012-0979-y

PubMed Abstract | Crossref Full Text | Google Scholar

Finley, K. H., and Cobb, S. (1940). The capillary bed of the locus coeruleus. J. Comp. Neurol. 73, 49–58. doi: 10.1002/cne.900730105

Crossref Full Text | Google Scholar

Fitzgerald, P. J. (2021). Diurnal build-up of norepinephrine may underlie sundowning in dementia. Clin. Neurol. Neurosurg. 206:106702. doi: 10.1016/j.clineuro.2021.106702

PubMed Abstract | Crossref Full Text | Google Scholar

Foldes, S. S., Moriarty, J. P., Farseth, P. H., Mittelman, M. S., and Long, K. H. (2018). Medicaid savings from the new York University caregiver intervention for families with dementia. Gerontologist 58, e97–e106. doi: 10.1093/geront/gnx077

PubMed Abstract | Crossref Full Text | Google Scholar

Font, C., Araneda, S., Pujol, J. F., and Bobillier, P. (1982). Biochemical and autoradiographic investigations of the retrograde axonal transport of labeled material following [(3)H] norepinephrine injection in the olfactory bulb. Neurochem. Int. 4, 569–575. doi: 10.1016/0197-0186(82)90047-x

PubMed Abstract | Crossref Full Text | Google Scholar

Food and Drug Administration. (n.d.) Early Alzheimer’s disease: developing drugs for treatment; draft guidance for industry. Available online at: https: //www.fda.gov/media/110903/download

Google Scholar

Foote, S. L., and Berridge, C. W. (2019). New developments and future directions in understanding locus coeruleus – norepinephrine (LC-NE) function. Brain Res. 1709, 81–84. doi: 10.1016/j.brainres.2018.09.033

PubMed Abstract | Crossref Full Text | Google Scholar

Fornai, F., Bassi, L., Torracca, M. T., Alessandri, M. G., Scalori, V., and Corsini, G. U. (1996). Region- and neurotransmitter-dependent species and strain differences in DSP-4-induced monoamine depletion in rodents. Neurodegeneration 5, 241–249. doi: 10.1006/neur.1996.0032

PubMed Abstract | Crossref Full Text | Google Scholar

Fu, Z., Zhao, M., He, Y., Wang, X., Lu, J., Li, S., et al. (2021). Divergent connectivity changes in gray matter structural covariance networks in subjective cognitive decline, amnestic mild cognitive impairment, and Alzheimer's disease. Front. Aging Neurosci. 13:686598. doi: 10.3389/fnagi.2021.686598

PubMed Abstract | Crossref Full Text | Google Scholar

Galgani, A., Lombardo, F., Della Latta, D., Martini, N., Bonuccelli, U., Fornai, F., et al. (2020). Locus Coeruleus magnetic resonance imaging in neurological diseases. Curr. Neurol. Neurosci. Rep. 21:2. doi: 10.1007/s11910-020-01087-7

PubMed Abstract | Crossref Full Text | Google Scholar

Gambardella, S., Ferese, R., Biagioni, F., Busceti, C. L., Campopiano, R., Griguoli, A. M. P., et al. (2017). The monoamine brainstem reticular formation as a paradigm for re-defining various phenotypes of Parkinson's disease owing genetic and anatomical specificity. Front. Cell. Neurosci. 11:102. doi: 10.3389/fncel.2017.00102

PubMed Abstract | Crossref Full Text | Google Scholar

Garcia-Rill, E. (2019). “Arousal and the Alzheimer disease” in Arousal in neurological and psychiatric diseases. ed. E. Garcia-Rill (Academic Press), 131–140.

Google Scholar

Gaspar, P., Berger, B., Febvret, A., Vigny, A., and Henry, J. P. (1989). Catecholamine innervation of the human cerebral cortex as revealed by comparative immunohistochemistry of tyrosine hydroxylase and dopamine-beta-hydroxylase. J. Comp. Neurol. 279, 249–271. doi: 10.1002/cne.902790208

PubMed Abstract | Crossref Full Text | Google Scholar

George, S. A., Knox, D., Curtis, A. L., Aldridge, J. W., Valentino, R. J., and Liberzon, I. (2013). Altered locus coeruleus-norepinephrine function following single prolonged stress. Eur. J. Neurosci. 37, 901–909. doi: 10.1111/ejn.12095

PubMed Abstract | Crossref Full Text | Google Scholar

German, D. C., Walker, B. S., Manaye, K., Smith, W. K., Woodward, D. J., and North, A. J. (1988). The human locus coeruleus: computer reconstruction of cellular distribution. J. Neurosci. 8, 1776–1788. doi: 10.1523/JNEUROSCI.08-05-01776.1988

PubMed Abstract | Crossref Full Text | Google Scholar

Geser, F., Haybaeck, J., and Yilmazer-Hanke, D. (2022). "Dendroarchitectonics": from Santiago Ramon y Cajal to Enrique Ramon-Moliner or vice versa? Neurol. Sci. 43, 5807–5820. doi: 10.1007/s10072-022-06151-3

PubMed Abstract | Crossref Full Text | Google Scholar

Gesi, M., Soldani, P., Giorgi, F. S., Santinami, A., Bonaccorsi, I., and Fornai, F. (2000). The role of the locus coeruleus in the development of Parkinson's disease. Neurosci. Biobehav. Rev. 24, 655–668. doi: 10.1016/S0149-7634(00)00028-2

PubMed Abstract | Crossref Full Text | Google Scholar

Gilsbach, R., Faron-Gorecka, A., Rogoz, Z., Bruss, M., Caron, M. G., Dziedzicka-Wasylewska, M., et al. (2006). Norepinephrine transporter knockout-induced up-regulation of brain alpha2A/C-adrenergic receptors. J. Neurochem. 96, 1111–1120. doi: 10.1111/j.1471-4159.2005.03598.x

PubMed Abstract | Crossref Full Text | Google Scholar

Giorgi, F. S., Galgani, A., Puglisi-Allegra, S., Limanaqi, F., Busceti, C. L., and Fornai, F. (2020). Locus Coeruleus and neurovascular unit: from its role in physiology to its potential role in Alzheimer's disease pathogenesis. J. Neurosci. Res. 98, 2406–2434. doi: 10.1002/jnr.24718

PubMed Abstract | Crossref Full Text | Google Scholar

Giorgi, F. S., Lombardo, F., Galgani, A., Hlavata, H., Della Latta, D., Martini, N., et al. (2022). Locus Coeruleus magnetic resonance imaging in cognitively intact elderly subjects. Brain Imaging Behav. 16, 1077–1087. doi: 10.1007/s11682-021-00562-0

PubMed Abstract | Crossref Full Text | Google Scholar

Giorgi, F. S., Pizzanelli, C., Biagioni, F., Murri, L., and Fornai, F. (2004). The role of norepinephrine in epilepsy: from the bench to the bedside. Neurosci. Biobehav. Rev. 28, 507–524. doi: 10.1016/j.neubiorev.2004.06.008

PubMed Abstract | Crossref Full Text | Google Scholar

Giorgi, F. S., Ryskalin, L., Ruffoli, R., Biagioni, F., Limanaqi, F., Ferrucci, M., et al. (2017). The neuroanatomy of the reticular nucleus locus Coeruleus in Alzheimer's disease. Front. Neuroanat. 11:80. doi: 10.3389/fnana.2017.00080

PubMed Abstract | Crossref Full Text | Google Scholar

Giorgi, F. S., Saccaro, L. F., Galgani, A., Busceti, C. L., Biagioni, F., Frati, A., et al. (2019). The role of locus Coeruleus in neuroinflammation occurring in Alzheimer's disease. Brain Res. Bull. 153, 47–58. doi: 10.1016/j.brainresbull.2019.08.007

PubMed Abstract | Crossref Full Text | Google Scholar

Gompf, H. S., Mathai, C., Fuller, P. M., Wood, D. A., Pedersen, N. P., Saper, C. B., et al. (2010). Locus ceruleus and anterior cingulate cortex sustain wakefulness in a novel environment. J. Neurosci. 30, 14543–14551. doi: 10.1523/JNEUROSCI.3037-10.2010

PubMed Abstract | Crossref Full Text | Google Scholar

Grinberg, L. T., Rueb, U., and Heinsen, H. (2011). Brainstem: neglected locus in neurodegenerative diseases. Front. Neurol. 2:42. doi: 10.3389/fneur.2011.00042

PubMed Abstract | Crossref Full Text | Google Scholar

Grudzien, A., Shaw, P., Weintraub, S., Bigio, E., Mash, D., and Mesulam, M. (2007). Locus coeruleus neurofibrillary degeneration in aging, mild cognitive impairment and early Alzheimer's disease. Neurobiol. Aging 28, 327–335. doi: 10.1016/j.neurobiolaging.2006.02.007

PubMed Abstract | Crossref Full Text | Google Scholar

Gustavsson, A., Norton, N., Fast, T., Frölich, L., Georges, J., Holzapfel, D., et al. (2023). Global estimates on the number of persons across the Alzheimer's disease continuum. Alzheimers Dement. 19, 658–670. doi: 10.1002/alz.12694

PubMed Abstract | Crossref Full Text | Google Scholar

Hämmerer, D., Callaghan, M. F., Hopkins, A., Kosciessa, J., Betts, M., Cardenas-Blanco, A., et al. (2018). Locus coeruleus integrity in old age is selectively related to memories linked with salient negative events. Proc. Natl. Acad. Sci. USA 115, 2228–2233. doi: 10.1073/pnas.1712268115

PubMed Abstract | Crossref Full Text | Google Scholar

Heneka, M. T., Nadrigny, F., Regen, T., Martinez-Hernandez, A., Dumitrescu-Ozimek, L., Terwel, D., et al. (2010). Locus ceruleus controls Alzheimer's disease pathology by modulating microglial functions through norepinephrine. Proc. Natl. Acad. Sci. USA 107, 6058–6063. doi: 10.1073/pnas.0909586107

PubMed Abstract | Crossref Full Text | Google Scholar

Heneka, M. T., Ramanathan, M., Jacobs, A. H., Dumitrescu-Ozimek, L., Bilkei-Gorzo, A., Debeir, T., et al. (2006). Locus ceruleus degeneration promotes Alzheimer pathogenesis in amyloid precursor protein 23 transgenic mice. J. Neurosci. 26, 1343–1354. doi: 10.1523/JNEUROSCI.4236-05.2006

PubMed Abstract | Crossref Full Text | Google Scholar

Herrmann, N., Lanctot, K. L., and Khan, L. R. (2004). The role of norepinephrine in the behavioral and psychological symptoms of dementia. J. Neuropsychiatry Clin. Neurosci. 16, 261–276. doi: 10.1176/jnp.16.3.261

PubMed Abstract | Crossref Full Text | Google Scholar

Himali, J. J., Baril, A.-A., Cavuoto, M. G., Yiallourou, S., Wiedner, C. D., Himali, D., et al. (2023). Association between slow-wave sleep loss and incident dementia. JAMA Neurol. 80, 1326–1333. doi: 10.1001/jamaneurol.2023.3889

PubMed Abstract | Crossref Full Text | Google Scholar

Holth, J. K., Fritschi, S. K., Wang, C., Pedersen, N. P., Cirrito, J. R., Mahan, T. E., et al. (2019). The sleep-wake cycle regulates brain interstitial fluid tau in mice and CSF tau in humans. Science 363, 880–884. doi: 10.1126/science.aav2546

PubMed Abstract | Crossref Full Text | Google Scholar

Hoogendijk, W. J. G., Feenstra, M. G. P., Botterblom, M. H. A., Gilhuis, J., Sommer, I. E. C., Kamphorst, W., et al. (1999). Increased activity of surviving locus ceruleus neurons in Alzheimer's disease. Ann. Neurol. 45, 82–91. doi: 10.1002/1531-8249(199901)45:1<82::AID-ART14>3.0.CO;2-T

PubMed Abstract | Crossref Full Text | Google Scholar

Iannitelli, A. F., and Weinshenker, D. (2023). Riddles in the dark: decoding the relationship between neuromelanin and neurodegeneration in locus coeruleus neurons. Neurosci. Biobehav. Rev. 152:105287. doi: 10.1016/j.neubiorev.2023.105287

PubMed Abstract | Crossref Full Text | Google Scholar

Ito, K., and Enomoto, H. (2016). Retrograde transport of neurotrophic factor signaling: implications in neuronal development and pathogenesis. J. Biochem. 160, 77–85. doi: 10.1093/jb/mvw037

PubMed Abstract | Crossref Full Text | Google Scholar

Jacobs, H., Becker, J. A., Kwong, K., Engels-domínguez, N., Prokopiou, P. C., Papp, K. V., et al. (2022). In vivo and neuropathology data support locus coeruleus integrity as indicator of Alzheimer’s disease pathology and cognitive decline. Sci. Transl. Med. 13, 1–16. doi: 10.1126/scitranslmed.abj2511

Crossref Full Text | Google Scholar

Jacobs, H. I. L., Muller-Ehrenberg, L., Priovoulos, N., and Roebroeck, A. (2018). Curvilinear locus coeruleus functional connectivity trajectories over the adult lifespan: a 7T MRI study. Neurobiol. Aging 69, 167–176. doi: 10.1016/j.neurobiolaging.2018.05.021

PubMed Abstract | Crossref Full Text | Google Scholar

Jacobs, H. I. L., Wiese, S., van de Ven, V., Gronenschild, E. H. B. M., Verhey, F. R. J., and Matthews, P. M. (2015). Relevance of parahippocampal-locus coeruleus connectivity to memory in early dementia. Neurobiol. Aging 36, 618–626. doi: 10.1016/j.neurobiolaging.2014.10.041

PubMed Abstract | Crossref Full Text | Google Scholar

Jacobsohn, L. Über die Kerne des menschlichen Hirnstamms (medulla oblongata, pons und pedunculus cerebri). Berlin: Verlag der Königl. Akademie der Wissenschaften. 1909.

Google Scholar

Jucker, M., and Walker, L. C. (2023). Alzheimer's disease: from immunotherapy to immunoprevention. Cell 186, 4260–4270. doi: 10.1016/j.cell.2023.08.021

PubMed Abstract | Crossref Full Text | Google Scholar

Jutkowitz, E., Pizzi, L. T., Shewmaker, P., Alarid-Escudero, F., Epstein-Lubow, G., Prioli, K. M., et al. (2023). Cost effectiveness of non-drug interventions that reduce nursing home admissions for people living with dementia. Alzheimers Dement. 19, 3867–3893. doi: 10.1002/alz.12964

PubMed Abstract | Crossref Full Text | Google Scholar

Kalinin, S., Feinstein, D. L., Xu, H. L., Huesa, G., Pelligrino, D. A., and Galea, E. (2006). Degeneration of noradrenergic fibres from the locus coeruleus causes tight-junction disorganisation in the rat brain. Eur. J. Neurosci. 24, 3393–3400. doi: 10.1111/j.1460-9568.2006.05223.x

PubMed Abstract | Crossref Full Text | Google Scholar

Kawamoto, Y., Ito, H., Ayaki, T., and Takahashi, R. (2014). Immunohistochemical localization of apoptosome-related proteins in Lewy bodies in Parkinson’s disease and dementia with Lewy bodies. Brain Res. 1571, 39–48. doi: 10.1016/j.brainres.2014.05.007

Crossref Full Text | Google Scholar

Kelberman, M. A., Anderson, C. R., Chlan, E., Rorabaugh, J. M., McCann, K. E., and Weinshenker, D. (2022). Consequences of hyperphosphorylated tau in the locus coeruleus on behavior and cognition in a rat model of Alzheimer's disease. J. Alzheimers Dis. 86, 1037–1059. doi: 10.3233/JAD-215546

PubMed Abstract | Crossref Full Text | Google Scholar

Kelberman, M., Keilholz, S., and Weinshenker, D. (2020). What’s that (blue) spot on my MRI? Multimodal neuroimaging of the locus coeruleus in neurodegenerative disease. Front. Neurosci. 14, 1–17. doi: 10.3389/fnins.2020.583421

PubMed Abstract | Crossref Full Text | Google Scholar

Kelberman, M. A., Rorabaugh, J. M., Anderson, C. R., Marriott, A., DePuy, S. D., Rasmussen, K., et al. (2023). Age-dependent dysregulation of locus coeruleus firing in a transgenic rat model of Alzheimer's disease. Neurobiol. Aging 125, 98–108. doi: 10.1016/j.neurobiolaging.2023.01.016

PubMed Abstract | Crossref Full Text | Google Scholar

Kelly, S. C., He, B., Perez, S. E., Ginsberg, S. D., Mufson, E. J., and Counts, S. E. (2017). Locus coeruleus cellular and molecular pathology during the progression of Alzheimer's disease. Acta Neuropathol. Commun. 5:8. doi: 10.1186/s40478-017-0411-2

PubMed Abstract | Crossref Full Text | Google Scholar

Keren, N. I., Taheri, S., Vazey, E. M., Morgan, P. S., Granholm, A.-c E., Aston-jones, G. S., et al. (2015). Histologic validation of locus coeruleus MRI contrast in post-mortem tissue. Neuro Image 113, 235–245. doi: 10.1016/j.neuroimage.2015.03.020

PubMed Abstract | Crossref Full Text | Google Scholar

Khosravi, M., Peter, J., Wintering, N. A., Serruya, M., Shamchi, S. P., Werner, T. J., et al. (2019). 18F-FDG is a superior Indicator of cognitive performance compared to 18F-Florbetapir in Alzheimer's disease and mild cognitive impairment evaluation: a global quantitative analysis. J. Alzheimers Dis. 70, 1197–1207. doi: 10.3233/JAD-190220

PubMed Abstract | Crossref Full Text | Google Scholar

Kong, Y., Ruan, L., Qian, L., Liu, X., and Le, Y. (2010). Norepinephrine promotes microglia to uptake and degrade amyloid beta peptide through upregulation of mouse formyl peptide receptor 2 and induction of insulin-degrading enzyme. J. Neurosci. 30, 11848–11857. doi: 10.1523/JNEUROSCI.2985-10.2010

PubMed Abstract | Crossref Full Text | Google Scholar

Koops, E. A., Dutta, J., Hanseeuw, B. J., Becker, J. A., Van Egroo, M., Prokopiou, P. C., et al. (2025). Elevated locus coeruleus metabolism provides resilience against cognitive decline in preclinical Alzheimer's disease. Alzheimers Dement. 21:e14385. doi: 10.1002/alz.14385

PubMed Abstract | Crossref Full Text | Google Scholar

Krohn, F., Lancini, E., Ludwig, M., Leiman, M., Guruprasath, G., Haag, L., et al. (2023). Noradrenergic neuromodulation in ageing and disease. Neurosci. Biobehav. Rev. 152:105311. doi: 10.1016/j.neubiorev.2023.105311

PubMed Abstract | Crossref Full Text | Google Scholar

La, A. L., Walsh, C. M., Neylan, T. C., Vossel, K. A., Yaffe, K., Krystal, A. D., et al. (2019). Long-term trazodone use and cognition: a potential therapeutic role for slow-wave sleep enhancers. J. Alzheimers Dis. 67, 911–921. doi: 10.3233/JAD-181145

PubMed Abstract | Crossref Full Text | Google Scholar

Langley, J., Hussain, S., Flores, J. J., Bennett, I. J., and Hu, X. (2020). Characterization of age-related microstructural changes in locus coeruleus and substantia nigra pars compacta. Neurobiol. Aging 87, 89–97. doi: 10.1016/j.neurobiolaging.2019.11.016

PubMed Abstract | Crossref Full Text | Google Scholar

Le, L. H. D., Feidler, A. M., Rodriguez, L. C., Cealie, M., Plunk, E., Li, H., et al. (2025). Noradrenergic signaling controls Alzheimer's disease pathology via activation of microglial beta2 adrenergic receptors. Brain Behav. Immun. 128, 307–322. doi: 10.1016/j.bbi.2025.04.022

PubMed Abstract | Crossref Full Text | Google Scholar

Lee, T. H., Kim, S. H., Katz, B., and Mather, M. (2020). The decline in intrinsic connectivity between the salience network and locus Coeruleus in older adults: implications for distractibility. Front. Aging Neurosci. 12, 1–8. doi: 10.3389/fnagi.2020.00002

PubMed Abstract | Crossref Full Text | Google Scholar

Leiman, M, and Hämmerer, D 2024 The effect of traumatic events on the locus Coeruleus-noradrenergic system (LC-NA)

Google Scholar

Lenglet, C. (2015). “Diffusion tensor imaging” in Brain mapping: An encyclopedic reference, vol. 1 (Academic Press), 245–251.

Google Scholar

Leslie, F., Loughlin, S., Sternberg, D., McGaugh, J., Young, L., and Zornetzer, S. (1985). Noradrenergic changes and memory loss in aged mice. Brain Res. 359, 292–299. doi: 10.1016/0006-8993(85)91439-8

PubMed Abstract | Crossref Full Text | Google Scholar

Lew, C. H., Petersen, C., Neylan, T. C., and Grinberg, L. T. (2021). Tau-driven degeneration of sleep- and wake-regulating neurons in Alzheimer's disease. Sleep Med. Rev. 60:101541. doi: 10.1016/j.smrv.2021.101541

PubMed Abstract | Crossref Full Text | Google Scholar

Lewis, D. A., and Morrison, J. H. (1989). Noradrenergic innervation of monkey prefrontal cortex: a dopamine-beta-hydroxylase immunohistochemical study. J. Comp. Neurol. 282, 317–330. doi: 10.1002/cne.902820302

PubMed Abstract | Crossref Full Text | Google Scholar

Li, Y., Schindler, S. E., Bollinger, J. G., Ovod, V., Mawuenyega, K. G., Weiner, M. W., et al. (2022). Validation of plasma amyloid-β 42/40 for detecting Alzheimer disease amyloid plaques. Neurology 98, E688–e699. doi: 10.1212/WNL.0000000000013211

PubMed Abstract | Crossref Full Text | Google Scholar

Liebe, T., Kaufmann, J., Li, M., Skalej, M., Wagner, G., and Walter, M. (2020). In vivo anatomical mapping of human locus coeruleus functional connectivity at 3 T MRI. Hum. Brain Mapp. 41, 2136–2151. doi: 10.1002/hbm.24935

PubMed Abstract | Crossref Full Text | Google Scholar

Lin, C. P., Frigerio, I., Bol, J., Bouwman, M. M. A., Wesseling, A. J., Dahl, M. J., et al. (2024). Microstructural integrity of the locus coeruleus and its tracts reflect noradrenergic degeneration in Alzheimer's disease and Parkinson's disease. Transl. Neurodegener. 13:9. doi: 10.1186/s40035-024-00400-5

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, K. Y., Acosta-Cabronero, J., Cardenas-Blanco, A., Loane, C., Berry, A. J., Betts, M. J., et al. (2019). In vivo visualization of age-related differences in the locus coeruleus. Neurobiol. Aging 74, 101–111. doi: 10.1016/j.neurobiolaging.2018.10.014

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, K. Y., Acosta-Cabronero, J., Hong, Y. T., Yi, Y. J., Hammerer, D., Howard, R., et al. (2021). FDG-PET assessment of the locus coeruleus in Alzheimer's disease. Neuroimage Rep. 1:100002. doi: 10.1016/j.ynirp.2020.100002

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, K. Y., Betts, M. J., Hammerer, D., Duzel, E., Mather, M., Roiser, J. P., et al. (2025). Locus coeruleus signal intensity and emotion regulation in agitation in Alzheimer's disease. Brain Commun. 7:fcae457. doi: 10.1093/braincomms/fcae457

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, K. Y., Kievit, R. A., Tsvetanov, K. A., Betts, M. J., Duzel, E., Rowe, J. B., et al. (2020). Noradrenergic-dependent functions are associated with age-related locus coeruleus signal intensity differences. Nat. Commun. 11:1712. doi: 10.1038/s41467-020-15410-w

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, K. Y., Stringer, A. E., Reeves, S. J., and Howard, R. J. (2018). The neurochemistry of agitation in Alzheimer's disease: a systematic review. Ageing Res. Rev. 43, 99–107. doi: 10.1016/j.arr.2018.03.003

PubMed Abstract | Crossref Full Text | Google Scholar

Lucey, B. P., Liu, H., Toedebusch, C. D., Freund, D., Redrick, T., Chahin, S. L., et al. (2023). Suvorexant acutely decreases tau phosphorylation and Abeta in the human CNS. Ann. Neurol. 94, 27–40. doi: 10.1002/ana.26641

Crossref Full Text | Google Scholar

Ludwig, M., Yi, Y. J., Lusebrink, F., Callaghan, M. F., Betts, M. J., Yakupov, R., et al. (2024). Functional locus coeruleus imaging to investigate an ageing noradrenergic system. Commun. Biol. 7:777. doi: 10.1038/s42003-024-06446-5

PubMed Abstract | Crossref Full Text | Google Scholar

Maeda, T. (2000). The locus coeruleus: history. J. Chem. Neuroanat. 18, 57–64. doi: 10.1016/S0891-0618(99)00051-4

PubMed Abstract | Crossref Full Text | Google Scholar

Manaye, K. F., McIntire, D. D., Mann, D. M. A., and German, D. C. (1995). Locus coeruleus cell loss in the aging human brain: a non-random process. J. Comp. Neurol. 358, 79–87. doi: 10.1002/cne.903580105

PubMed Abstract | Crossref Full Text | Google Scholar

Mann, D. M. A. (1983). The locus coeruleus and its possible role in ageing and degenerative disease of the human central nervous system. Mech. Ageing Dev. 23, 73–94. doi: 10.1016/0047-6374(83)90100-8

PubMed Abstract | Crossref Full Text | Google Scholar

Mann, D. M., Yates, P. O., and Hawkes, J. (1982). The noradrenergic system in Alzheimer and multi-infarct dementias. J. Neurol. Neurosurg. Psychiatry 45, 113–119. doi: 10.1136/jnnp.45.2.113

PubMed Abstract | Crossref Full Text | Google Scholar

Marien, M. R., Colpaert, F. C., and Rosenquist, A. C. (2004). Noradrenergic mechanisms in neurodegenerative diseases: a theory. Brain Res. Rev. 45, 38–78. doi: 10.1016/j.brainresrev.2004.02.002

PubMed Abstract | Crossref Full Text | Google Scholar

Marquie, M., Normandin, M. D., Vanderburg, C. R., Costantino, I. M., Bien, E. A., Rycyna, L. G., et al. (2015). Validating novel tau positron emission tomography tracer [F-18]-AV-1451 (T807) on postmortem brain tissue. Ann. Neurol. 78, 787–800. doi: 10.1002/ana.24517

PubMed Abstract | Crossref Full Text | Google Scholar

Mather, M. (2018). “The locus coeruleus-norepinephrine system role in cognition and how it changes with aging” in The cognitive neurosciences. eds. D. M. Poppel and M. Gazzaniga (Cambridge, MA: MIT Press), 1689–1699.

Google Scholar

Mather, M., and Harley, C. W. (2016). The locus Coeruleus: essential for maintaining cognitive function and the aging brain. Trends Cogn. Sci. 20, 214–226. doi: 10.1016/j.tics.2016.01.001

PubMed Abstract | Crossref Full Text | Google Scholar

Matschke, L. A., Bertoune, M., Roeper, J., Snutch, T. P., Oertel, W. H., Rinné, S., et al. (2015). A concerted action of L- and T-type Ca2+ channels regulates locus coeruleus pacemaking. Mol. Cell. Neurosci. 68, 293–302. doi: 10.1016/j.mcn.2015.08.012

PubMed Abstract | Crossref Full Text | Google Scholar

Mayer, G., Frohnhofen, H., Jokisch, M., Hermann, D. M., and Gronewold, J. (2024). Associations of sleep disorders with all-cause MCI/dementia and different types of dementia - clinical evidence, potential pathomechanisms and treatment options: a narrative review. Front. Neurosci. 18. doi: 10.3389/fnins.2024.1372326

PubMed Abstract | Crossref Full Text | Google Scholar

Mijalkov, M., Storm, L., Zufiria-Gerboles, B., Vereb, D., Xu, Z., Canal-Garcia, A., et al. (2025). Computational memory capacity predicts aging and cognitive decline. Nat. Commun. 16:2748. doi: 10.1038/s41467-025-57995-0

PubMed Abstract | Crossref Full Text | Google Scholar

Miller, M. A., Kolb, P. E., Leverenz, J. B., Peskind, E. R., and Raskind, M. A. (1999). Preservation of noradrenergic neurons in the locus ceruleus that coexpress galanin mRNA in Alzheimer's disease. J. Neurochem. 73, 2028–2036. doi: 10.1046/j.1471-4159.1999.02028.x

PubMed Abstract | Crossref Full Text | Google Scholar

Moriguchi, S., Kimura, Y., Ichise, M., Arakawa, R., Takano, H., Seki, C., et al. (2017). PET quantification of the norepinephrine transporter in human brain with (S,S)-(18)F-FMeNER-D (2). J. Nucl. Med. 58, 1140–1145. doi: 10.2967/jnumed.116.178913

PubMed Abstract | Crossref Full Text | Google Scholar

Mouton, P. R., Pakkenberg, B., Gundersen, H. J., and Price, D. L. (1994). Absolute number and size of pigmented locus coeruleus neurons in young and aged individuals. J. Chem. Neuroanat. 7, 185–190. doi: 10.1016/0891-0618(94)90028-0

PubMed Abstract | Crossref Full Text | Google Scholar

Mravec, B., Lejavova, K., and Cubinkova, V. (2014). Locus (coeruleus) minoris resistentiae in pathogenesis of Alzheimer's disease. Curr. Alzheimer Res. 11, 992–1001. doi: 10.2174/1567205011666141107130505

PubMed Abstract | Crossref Full Text | Google Scholar

Neylan, T. C., and Walsh, C. M. (2024). Wake, NREM, and REM sleep measures predict incident dementia. Sleep 47:zsad329. doi: 10.1093/sleep/zsad329

PubMed Abstract | Crossref Full Text | Google Scholar

Nick, H., Fenik, P., Zhu, Y., and Veasey, S. (2022). Hypocretin/orexin influences chronic sleep disruption injury in the hippocampus. Front. Aging Neurosci. 14:1025402. doi: 10.3389/fnagi.2022.1025402

PubMed Abstract | Crossref Full Text | Google Scholar

Oh, J., Eser, R. A., Ehrenberg, A. J., Morales, D., Petersen, C., Kudlacek, J., et al. (2019). Profound degeneration of wake-promoting neurons in Alzheimer's disease. Alzheimers Dement. 15, 1253–1263. doi: 10.1016/j.jalz.2019.06.3916

PubMed Abstract | Crossref Full Text | Google Scholar

Oh, J. Y., Walsh, C. M., Ranasinghe, K., Mladinov, M., Pereira, F. L., Petersen, C., et al. (2022). Subcortical neuronal correlates of sleep in neurodegenerative diseases. JAMA Neurol. 79, 498–508. doi: 10.1001/jamaneurol.2022.0429

PubMed Abstract | Crossref Full Text | Google Scholar

Omoluabi, T., Hasan, Z., Piche, J. E., Flynn, A. R. S., Dore, J. J. E., Walling, S. G., et al. (2025). Locus coeruleus vulnerability to tau hyperphosphorylation in a rat model. Aging Cell 24:e14405. doi: 10.1111/acel.14405

PubMed Abstract | Crossref Full Text | Google Scholar

Orlando, I. F., Shine, J. M., Robbins, T. W., Rowe, J. B., and O'Callaghan, C. (2023). Noradrenergic and cholinergic systems take centre stage in neuropsychiatric diseases of ageing. Neurosci. Biobehav. Rev. 149:105167. doi: 10.1016/j.neubiorev.2023.105167

Crossref Full Text | Google Scholar

Pamphlett, R. (2014). Uptake of environmental toxicants by the locus ceruleus: a potential trigger for neurodegenerative, demyelinating and psychiatric disorders. Med. Hypotheses 82, 97–104. doi: 10.1016/j.mehy.2013.11.016

PubMed Abstract | Crossref Full Text | Google Scholar

Pamphlett, R., and Kum Jew, S. (2014). Uptake of inorganic mercury by human locus ceruleus and corticomotor neurons: implications for amyotrophic lateral sclerosis. Acta Neuropathol. Commun. 1:1. doi: 10.1186/2051-5960-1-13

PubMed Abstract | Crossref Full Text | Google Scholar

Pamphlett, R., and Kum Jew, S. (2015). Different populations of human locus ceruleus neurons contain heavy metals or hyperphosphorylated tau: implications for amyloid-β and tau pathology in Alzheimer's disease. J. Alzheimers Dis. 45, 437–447. doi: 10.3233/JAD-142445

PubMed Abstract | Crossref Full Text | Google Scholar

Pamphlett, R., Mak, R., Lee, J., Buckland, M. E., Harding, J., Jew, S. K., et al. (2020). Concentrations of toxic metals and essential trace elements vary among individual neurons in the human locus ceruleus. PLoS One 15, 1–18. doi: 10.1371/journal.pone.0233300

Crossref Full Text | Google Scholar

Parhizkar, S., and Holtzman, D. M. (2025). The night's watch: exploring how sleep protects against neurodegeneration. Neuron 113, 817–837. doi: 10.1016/j.neuron.2025.02.004

PubMed Abstract | Crossref Full Text | Google Scholar

Patt, S., and Gerhard, L. (1993). A Golgi study of human locus coeruleus in normal brains and in Parkinson's disease. Neuropathol. Appl. Neurobiol. 19, 519–523. doi: 10.1111/j.1365-2990.1993.tb00480.x

PubMed Abstract | Crossref Full Text | Google Scholar

Perry, E. K., Tomlinson, B. E., Blessed, G., Perry, R. H., Cross, A. J., and Crow, T. J. (1981). Neuropathological and biochemical observations on the noradrenergic system in Alzheimer's disease. J. Neurol. Sci. 51, 279–287. doi: 10.1016/0022-510X(81)90106-4

PubMed Abstract | Crossref Full Text | Google Scholar

Pietrzak, R. H., Gallezot, J. D., Ding, Y. S., Henry, S., Potenza, M. N., Southwick, S. M., et al. (2013). Association of posttraumatic stress disorder with reduced in vivo norepinephrine transporter availability in the locus coeruleus. JAMA Psychiatry 70, 1199–1205. doi: 10.1001/jamapsychiatry.2013.399

PubMed Abstract | Crossref Full Text | Google Scholar

Pless, A., Ware, D., Saggu, S., Rehman, H., Morgan, J., and Wang, Q. (2023). Understanding neuropsychiatric symptoms in Alzheimer's disease: challenges and advances in diagnosis and treatment. Front. Neurosci. 17:1263771. doi: 10.3389/fnins.2023.1263771

PubMed Abstract | Crossref Full Text | Google Scholar

Poe, G. R., Foote, S., Eschenko, O., Johansen, J. P., Bouret, S., Aston-Jones, G., et al. (2020). Locus coeruleus: a new look at the blue spot. Nat. Rev. Neurosci. 21, 644–659. doi: 10.1038/s41583-020-0360-9

PubMed Abstract | Crossref Full Text | Google Scholar

Polak, P. E., Kalinin, S., and Feinstein, D. L. (2011). Locus coeruleus damage and noradrenaline reductions in multiple sclerosis and experimental autoimmune encephalomyelitis. Brain 134, 665–677. doi: 10.1093/brain/awq362

PubMed Abstract | Crossref Full Text | Google Scholar

Quan, P., Mao, T., Zhang, X., Wang, R., Lei, H., Wang, J., et al. (2024). Locus coeruleus microstructural integrity is associated with vigilance vulnerability to sleep deprivation. Hum. Brain Mapp. 45:e70013. doi: 10.1002/hbm.70013

PubMed Abstract | Crossref Full Text | Google Scholar

Quattrini, G., Pini, L., Boscolo Galazzo, I., Jelescu, I. O., Jovicich, J., Manenti, R., et al. (2024). Microstructural alterations in the locus coeruleus-entorhinal cortex pathway in Alzheimer's disease and frontotemporal dementia. Alzheimers Dement. (Amst). 16:e12513. doi: 10.1002/dad2.12513

PubMed Abstract | Crossref Full Text | Google Scholar

Rafii, M. S., Sperling, R. A., Donohue, M. C., Zhou, J., Roberts, C., Irizarry, M. C., et al. (2023). The AHEAD 3-45 study: design of a prevention trial for Alzheimer's disease. Alzheimers Dement. 19, 1227–1233. doi: 10.1002/alz.12748

PubMed Abstract | Crossref Full Text | Google Scholar

Rajda, C., Bencsik, K., Füvesi, J., Seres, E., Vécsei, L., and Bergquist, J. (2006). The norepinephrine level is decreased in the lymphocytes of long-term interferon-beta-treated multiple sclerosis patients. Multiple Scler. 12, 265–270. doi: 10.1191/135248506ms1269oa

PubMed Abstract | Crossref Full Text | Google Scholar

Ramon-Moliner, E., and Nauta, W. J. (1966). The isodendritic core of the brain stem. J. Comp. Neurol. 126, 311–335. doi: 10.1002/cne.901260301

PubMed Abstract | Crossref Full Text | Google Scholar

Reil, J. C. (1809). Untersuchungen über den Bau des grossen Gehirns im Menschen. Arch. Physiol. (Halle) 9, 485–524. doi: 10.1515/9783112667361

PubMed Abstract | Crossref Full Text | Google Scholar

Remy, S., Naveilhan, P., Brachet, P., and Neveu, I. (2001). Differential regulation of GDNF, Neurturin, and their receptors in primary cultures of rat glial cells. J. Neurosci. Res. 64, 242–251. doi: 10.1002/jnr.1072

PubMed Abstract | Crossref Full Text | Google Scholar

Rodriguez-Martin, T., Cuchillo-Ibanez, I., Noble, W., Nyenya, F., Anderton, B. H., and Hanger, D. P. (2013). Tau phosphorylation affects its axonal transport and degradation. Neurobiol. Aging 34, 2146–2157. doi: 10.1016/j.neurobiolaging.2013.03.015

PubMed Abstract | Crossref Full Text | Google Scholar

Rohn, T. T., Rissman, R. A., Davis, M. C., Kim, Y. E., Cotman, C. W., and Head, E. (2002). Caspase-9 activation and caspase cleavage of tau in the Alzheimer's disease brain. Neurobiol. Dis. 11, 341–354. doi: 10.1006/nbdi.2002.0549

PubMed Abstract | Crossref Full Text | Google Scholar

Rorabaugh, J. M., Chalermpalanupap, T., Botz-Zapp, C. A., Fu, V. M., Lembeck, N. A., Cohen, R. M., et al. (2017). Chemogenetic locus coeruleus activation restores reversal learning in a rat model of Alzheimer's disease. Brain 140, 3023–3038. doi: 10.1093/brain/awx232

PubMed Abstract | Crossref Full Text | Google Scholar

Ross, S. B., and Stenfors, C. (2015). DSP4, a selective neurotoxin for the locus Coeruleus noradrenergic system. A review of its mode of action. Neurotox. Res. 27, 15–30. doi: 10.1007/s12640-014-9482-z

PubMed Abstract | Crossref Full Text | Google Scholar

Samuels, E., and Szabadi, E. (2008). Functional neuroanatomy of the noradrenergic locus coeruleus: its roles in the regulation of arousal and autonomic function part I: principles of functional organisation. Curr. Neuropharmacol. 6, 235–253. doi: 10.2174/157015908785777229

PubMed Abstract | Crossref Full Text | Google Scholar

Sanchez-Padilla, J., Guzman, J. N., Ilijic, E., Kondapalli, J., Galtieri, D. J., Yang, B., et al. (2014). Mitochondrial oxidant stress in locus coeruleus is regulated by activity and nitric oxide synthase. Nat. Neurosci. 17, 832–840. doi: 10.1038/nn.3717

PubMed Abstract | Crossref Full Text | Google Scholar

Sara, S. J. (2009). The locus coeruleus and noradrenergic modulation of cognition. Nat. Rev. Neurosci. 10, 211–223. doi: 10.1038/nrn2573

PubMed Abstract | Crossref Full Text | Google Scholar

Satoh, A., and Iijima, K. M. (2019). Roles of tau pathology in the locus coeruleus (LC) in age-associated pathophysiology and Alzheimer's disease pathogenesis: potential strategies to protect the LC against aging. Brain Res. 1702, 17–28. doi: 10.1016/j.brainres.2017.12.027

PubMed Abstract | Crossref Full Text | Google Scholar

Scarmeas, N., Brandt, J., Blacker, D., Albert, M., Hadjigeorgiou, G., Dubois, B., et al. (2007). Disruptive behavior as a predictor in Alzheimer disease. Arch. Neurol. 64, 1755–1761. doi: 10.1001/archneur.64.12.1755

PubMed Abstract | Crossref Full Text | Google Scholar

Schwarz, L. A., and Luo, L. (2015). Organization of the locus coeruleus-norepinephrine system. Curr. Biol. 25, R1051–R1056. doi: 10.1016/j.cub.2015.09.039

PubMed Abstract | Crossref Full Text | Google Scholar

Schwarz, L. A., Miyamichi, K., Gao, X. J., Beier, K. T., Weissbourd, B., Deloach, K. E., et al. (2015). Viral-genetic tracing of the input-output organization of a central noradrenaline circuit. Nature 524, 88–92. doi: 10.1038/nature14600

PubMed Abstract | Crossref Full Text | Google Scholar

Serra, L., D'Amelio, M., Di Domenico, C., Dipasquale, O., Marra, C., Mercuri, N. B., et al. (2018). In vivo mapping of brainstem nuclei functional connectivity disruption in Alzheimer's disease. Neurobiol. Aging 72, 72–82. doi: 10.1016/j.neurobiolaging.2018.08.012

PubMed Abstract | Crossref Full Text | Google Scholar

Sharma, Y., Xu, T., Graf, W. M., Fobbs, A., Sherwood, C. C., Hof, P. R., et al. (2010). Comparative anatomy of the locus coeruleus in humans and nonhuman primates. J. Comp. Neurol. 518, 963–971. doi: 10.1002/cne.22249

PubMed Abstract | Crossref Full Text | Google Scholar

Sobral, A. F., Costa, I., Teixeira, V., Silva, R., and Barbosa, D. J. (2025). Molecular Motors in Blood-Brain Barrier Maintenance by astrocytes. Brain Sci. 15:279. doi: 10.3390/brainsci15030279

PubMed Abstract | Crossref Full Text | Google Scholar

Stouffer, K. M., Grande, X., Duzel, E., Johansson, M., Creese, B., Witter, M. P., et al. (2024). Amidst an amygdala renaissance in Alzheimer's disease. Brain 147, 816–829. doi: 10.1093/brain/awad411

PubMed Abstract | Crossref Full Text | Google Scholar

Suárez-Pereira, I., Llorca-Torralba, M., Bravo, L., Camarena-Delgado, C., Soriano-Mas, C., and Berrocoso, E. (2022). The role of the locus Coeruleus in pain and associated stress-related disorders. Biol. Psychiatry 91, 786–797. doi: 10.1016/j.biopsych.2021.11.023

PubMed Abstract | Crossref Full Text | Google Scholar

Sulzer, D., Cassidy, C., Horga, G., Kang, U. J., Fahn, S., Casella, L., et al. (2018). Neuromelanin detection by magnetic resonance imaging (MRI) and its promise as a biomarker for Parkinson's disease. NPJ Parkinsons Dis. 4:11. doi: 10.1038/s41531-018-0047-3

PubMed Abstract | Crossref Full Text | Google Scholar

Sun, H., Paixao, L., Oliva, J. T., Goparaju, B., Carvalho, D. Z., van Leeuwen, K. G., et al. (2019). Brain age from the electroencephalogram of sleep. Neurobiol. Aging 74, 112–120. doi: 10.1016/j.neurobiolaging.2018.10.016

PubMed Abstract | Crossref Full Text | Google Scholar

Sun, W., Tang, Y., Qiao, Y., Ge, X., Mather, M., Ringman, J. M., et al. (2020). A probabilistic atlas of locus coeruleus pathways to transentorhinal cortex for connectome imaging in Alzheimer's disease. NeuroImage 223:117301. doi: 10.1016/j.neuroimage.2020.117301

PubMed Abstract | Crossref Full Text | Google Scholar

Svendsen, U. G., Ibsen, H., Rasmussen, S., Leth, A., Nielsen, M. D., Dige-Petersen, H., et al. (1986). Effects of combined therapy with amiloride and hydrochlorothiazide on plasma and total body potassium, blood pressure, and the renin-angiotensin-aldosterone system in hypertensive patients. Eur. J. Clin. Pharmacol. 30, 151–156. doi: 10.1007/BF00614293

PubMed Abstract | Crossref Full Text | Google Scholar

Swanson, LW. Neuroanatomical terminology: A lexicon of classical origins and historical foundations. Oxford and New York: Oxford University Press. 2015.

Google Scholar

Szabadi, E. (2013). Functional neuroanatomy of the central noradrenergic system. J. Psychopharmacol. 27, 659–693. doi: 10.1177/0269881113490326

PubMed Abstract | Crossref Full Text | Google Scholar

Szot, P., Miguelez, C., White, S. S., Franklin, A., Sikkema, C., Wilkinson, C. W., et al. (2010). A comprehensive analysis of the effect of DSP4 on the locus coeruleus noradrenergic system in the rat. Neuroscience 166, 279–291. doi: 10.1016/j.neuroscience.2009.12.027

PubMed Abstract | Crossref Full Text | Google Scholar

Takahashi, J., Shibata, T., Sasaki, M., Kudo, M., Yanezawa, H., Obara, S., et al. (2015). Detection of changes in the locus coeruleus in patients with mild cognitive impairment and Alzheimer’s disease: high-resolution fast spin-echo T1-weighted imaging. Geriatr. Gerontol. Int. 15, 334–340. doi: 10.1111/ggi.12280

PubMed Abstract | Crossref Full Text | Google Scholar

Tang, Y., Cao, M., Li, Y., Lin, Y., Wu, X., Chen, M., et al. (2023). Altered structural covariance of locus coeruleus in individuals with significant memory concern and patients with mild cognitive impairment. Cereb. Cortex 33, 8523–8533. doi: 10.1093/cercor/bhad137

PubMed Abstract | Crossref Full Text | Google Scholar

Taylor, B. K., and Westlund, K. N. (2017). The noradrenergic locus coeruleus as a chronic pain generator. J. Neurosci. Res. 95, 1336–1346. doi: 10.1002/jnr.23956

PubMed Abstract | Crossref Full Text | Google Scholar

Theofilas, P., Dunlop, S., Heinsen, H., and Grinberg, L. T. (2015). Turning on the light within: subcortical nuclei of the Isodentritic Core and their role in Alzheimer's disease pathogenesis. J. Alzheimers Dis. 46, 17–34. doi: 10.3233/JAD-142682

PubMed Abstract | Crossref Full Text | Google Scholar

Theofilas, P., Ehrenberg, A. J., Dunlop, S., Di Lorenzo Alho, A. T., Nguy, A., Leite, R. E. P., et al. (2017). Locus coeruleus volume and cell population changes during Alzheimer's disease progression: a stereological study in human postmortem brains with potential implication for early-stage biomarker discovery. Alzheimers Dement. 13, 236–246. doi: 10.1016/j.jalz.2016.06.2362

PubMed Abstract | Crossref Full Text | Google Scholar

Tomlinson, B., Irving, D., and Blessed, G. (1981). Cell loss in the locus coeruleus in senile dementia of Alzheimer type. J. Neurol. Sci. 49, 419–428. doi: 10.1016/0022-510X(81)90031-9

PubMed Abstract | Crossref Full Text | Google Scholar

Tona, K. D., Keuken, M. C., de Rover, M., Lakke, E., Forstmann, B. U., Nieuwenhuis, S., et al. (2017). In vivo visualization of the locus coeruleus in humans: quantifying the test-retest reliability. Brain Struct. Funct. 222, 4203–4217. doi: 10.1007/s00429-017-1464-5

PubMed Abstract | Crossref Full Text | Google Scholar

Tubbs, R. S., Loukas, M., Shoja, M. M., Mortazavi, M. M., and Cohen-Gadol, A. A. (2011). Felix Vicq d'Azyr (1746-1794): early founder of neuroanatomy and royal French physician. Childs Nerv. Syst. 27, 1031–1034. doi: 10.1007/s00381-011-1424-y

Crossref Full Text | Google Scholar

Um, Y. H., Wang, S. M., Kang, D. W., Kim, S., Lee, C. U., Kim, D., et al. (2024). Impact of apolipoprotein E4 on the locus Coeruleus functional connectivity in preclinical Alzheimer's disease. J. Alzheimers Dis. 99, 705–714. doi: 10.3233/JAD-240065

PubMed Abstract | Crossref Full Text | Google Scholar

Van Egroo, M., Koshmanova, E., Vandewalle, G., and Jacobs, H. I. L. (2022). Importance of the locus coeruleus-norepinephrine system in sleep-wake regulation: implications for aging and Alzheimer's disease. Sleep Med. Rev. 62:101592. doi: 10.1016/j.smrv.2022.101592

PubMed Abstract | Crossref Full Text | Google Scholar

Van Egroo, M., Riphagen, J. M., Ashton, N. J., Janelidze, S., Sperling, R. A., Johnson, K. A., et al. (2023). Ultra-high field imaging, plasma markers and autopsy data uncover a specific rostral locus coeruleus vulnerability to hyperphosphorylated tau. Mol. Psychiatry 28, 2412–2422. doi: 10.1038/s41380-023-02041-y

PubMed Abstract | Crossref Full Text | Google Scholar

Van Egroo, M., van Hooren, R. W. E., and Jacobs, H. I. L. (2021). Associations between locus coeruleus integrity and nocturnal awakenings in the context of Alzheimer's disease plasma biomarkers: a 7T MRI study. Alzheimers Res. Ther. 13:159. doi: 10.1186/s13195-021-00902-8

PubMed Abstract | Crossref Full Text | Google Scholar

Vereb, D., Mijalkov, M., Canal-Garcia, A., Chang, Y. W., Gomez-Ruiz, E., Gerboles, B. Z., et al. (2023). Age-related differences in the functional topography of the locus coeruleus and their implications for cognitive and affective functions. eLife 12:12. doi: 10.7554/eLife.87188

PubMed Abstract | Crossref Full Text | Google Scholar

Verkhratsky, A., Butt, A., Li, B., Illes, P., Zorec, R., Semyanov, A., et al. (2023). Astrocytes in human central nervous system diseases: a frontier for new therapies. Signal Transduct. Target. Ther. 8:396. doi: 10.1038/s41392-023-01628-9

PubMed Abstract | Crossref Full Text | Google Scholar

Vicq d’Azyr, F. (1786). Traité d’anatomie et de physiologie—avec des planches colorës representant au naturel les divers organs de l’homme et des animaux. Paris: Francois Ambroise Didot l’Aine.

Google Scholar

Vijayashankar, N., and Brody, H. (1979). A quantitative study of the pigmented neurons in the nuclei locus coeruleus and subcoeruleus in man as related to aging. J. Neuropathol. Exp. Neurol. 38, 490–497. doi: 10.1097/00005072-197909000-00004

PubMed Abstract | Crossref Full Text | Google Scholar

Von Euler, U. S. (1946). Sympathin in adrenergic nerve fibres. J. Physiol. 105:26

PubMed Abstract | Google Scholar

Wai, M. S. M., Liang, Y., Shi, C., Cho, E. Y. P., Kung, H. F., and Yew, D. T. (2009). Co-localization of hyperphosphorylated tau and caspases in the brainstem of Alzheimer's disease patients. Biogerontology 10, 457–469. doi: 10.1007/s10522-008-9189-8

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, C., and Holtzman, D. M. (2020). Bidirectional relationship between sleep and Alzheimer’s disease: role of amyloid, tau, and other factors. Neuropsychopharmacology 45, 104–120. doi: 10.1038/s41386-019-0478-5

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, X., and Michaelis, E. K. (2010). Selective neuronal vulnerability to oxidative stress in the brain. Front. Aging Neurosci. 2, 1–13. doi: 10.3389/fnagi.2010.00012

PubMed Abstract | Crossref Full Text | Google Scholar

Watamura, N., Foiani, M. S., Bez, S., Bourdenx, M., Santambrogio, A., Frodsham, C., et al. (2025). In vivo hyperphosphorylation of tau is associated with synaptic loss and behavioral abnormalities in the absence of tau seeds. Nat. Neurosci. 28, 293–307. doi: 10.1038/s41593-024-01829-7

PubMed Abstract | Crossref Full Text | Google Scholar

Waterhouse, B. D., Devilbiss, D., Fleischer, D., Sessler, F. M., and Simpson, K. L. (1977). New perspectives on the functional organization and postsynaptic influences of the locus ceruleus efferent projection system. Adv. Pharmacol. 42, 749–754.

Google Scholar

Weimer, D. L., and Sager, M. A. (2009). Early identification and treatment of Alzheimer's disease: social and fiscal outcomes. Alzheimers Dement. 5, 215–226. doi: 10.1016/j.jalz.2009.01.028

PubMed Abstract | Crossref Full Text | Google Scholar

Weinshenker, D. (2018). Long road to ruin: noradrenergic dysfunction in neurodegenerative disease. Trends Neurosci. 41, 211–223. doi: 10.1016/j.tins.2018.01.010

PubMed Abstract | Crossref Full Text | Google Scholar

Wenzel, J. W., and Karl, (1812). De penitiori structura cerebri hominis et brutorum. Tübingen: Apud Cottam, 354.

Google Scholar

Williams, J. T., North, R. A., Shefner, S. A., Nishi, S., and Egan, T. M. (1984). Membrane properties of rat locus coeruleus neurones. Neuroscience 13, 137–156. doi: 10.1016/0306-4522(84)90265-3

PubMed Abstract | Crossref Full Text | Google Scholar

Wilson, R. S., Nag, S., Boyle, P. A., Hizel, L. P., Yu, L., Buchman, A. S., et al. (2013). Neural reserve, neuronal density in the locus ceruleus, and cognitive decline. Neurology 80, 1202–1208. doi: 10.1212/WNL.0b013e3182897103

PubMed Abstract | Crossref Full Text | Google Scholar

World Health Organization. (n.d.) Available online at: https://www.who.int/news-room/fact-sheets/detail/dementia.

Google Scholar

Wree, A., Braak, H., Schleicher, A., and Zilles, K. (1980). Biomathematical analysis of the neuronal loss in the aging human brain of both sexes, demonstrated in pigment preparations of the pars cerebellaris loci coerulei. Anat. Embryol. 160, 105–119. doi: 10.1007/BF00315653

PubMed Abstract | Crossref Full Text | Google Scholar

Wu, J., Toporek, A., Lin, Q., Goldstein, F. C., Loring, D. W., Kelberman, M. A., et al. (2025). Probing locus coeruleus functional network in healthy aging and its association with Alzheimer's disease biomarkers using pupillometry. Alzheimers Res. Ther. 17:53. doi: 10.1186/s13195-025-01701-1

PubMed Abstract | Crossref Full Text | Google Scholar

Yang, Y., Kim, W. S., Michaelian, J. C., Lewis, S. J. G., Phillips, C. L., D'Rozario, A. L., et al. (2024). Predicting neurodegeneration from sleep related biofluid changes. Neurobiol. Dis. 190:106369. doi: 10.1016/j.nbd.2023.106369

PubMed Abstract | Crossref Full Text | Google Scholar

Zarow, C., Lyness, S. A., Mortimer, J. A., and Chui, H. C. (2003). Neuronal loss is greater in the locus coeruleus than nucleus basalis and substantia nigra in Alzheimer and Parkinson diseases. Arch. Neurol. 60, 337–341. doi: 10.1001/archneur.60.3.337

PubMed Abstract | Crossref Full Text | Google Scholar

Zavecz, Z., Shah, V. D., Murillo, O. G., Vallat, R., Mander, B. A., Winer, J. R., et al. (2023). NREM sleep as a novel protective cognitive reserve factor in the face of Alzheimer's disease pathology. BMC Med. 21, 1–12. doi: 10.1186/s12916-023-02811-z

PubMed Abstract | Crossref Full Text | Google Scholar

Zecca, L., Bellei, C., Costi, P., Albertini, A., Monzani, E., Casella, L., et al. (2008). New melanic pigments in the human brain that accumulate in aging and block environmental toxic metals. Proc. Natl. Acad. Sci. USA 105, 17567–17572. doi: 10.1073/pnas.0808768105

PubMed Abstract | Crossref Full Text | Google Scholar

Zhan, Y., Raza, M. U., Yuan, L., and Zhu, M. Y. (2019). Critical role of Oxidatively damaged DNA in selective noradrenergic vulnerability. Neuroscience 422, 184–201. doi: 10.1016/j.neuroscience.2019.09.036

PubMed Abstract | Crossref Full Text | Google Scholar

Zhou, M., and Tang, S. (2022). Effect of a dual orexin receptor antagonist on Alzheimer's disease: sleep disorders and cognition. Front. Med. (Lausanne) 9:984227. doi: 10.3389/fmed.2022.984227

PubMed Abstract | Crossref Full Text | Google Scholar

Zhu, Y., Fenik, P., Zhan, G., Xin, R., and Veasey, S. C. (2015). Degeneration in arousal neurons in chronic sleep disruption modeling sleep apnea. Front. Neurol. 6:109. doi: 10.3389/fneur.2015.00109

PubMed Abstract | Crossref Full Text | Google Scholar

Zorec, R., Vardjan, N., and Verkhratsky, A. (2017). “Chapter 1 – Locus Coeruleus noradrenergic neurons and Astroglia in health and disease” in Noradrenergic signaling and astroglia. eds. N. Vardjan and R. Zorec (Academic Press), 1–24.

Google Scholar

Zoukos, Y., Leonard, J. P., Thomaides, T., Thompson, A. J., and Cuzner, M. L. (1992). β-Adrenergic receptor density and function of peripheral blood mononuclear cells are increased in multiple sclerosis: a regulatory role for cortisol and interleukin-1. Ann. Neurol. 31, 657–662. doi: 10.1002/ana.410310614

PubMed Abstract | Crossref Full Text | Google Scholar

Zucca, F. A., Segura-Aguilar, J., Ferrari, E., Muñoz, P., Paris, I., Sulzer, D., et al. (2017). Interactions of iron, dopamine and neuromelanin pathways in brain aging and Parkinson's disease. Prog. Neurobiol. 155, 96–119. doi: 10.1016/j.pneurobio.2015.09.012

PubMed Abstract | Crossref Full Text | Google Scholar

Zweig, R. M., Ross, C. A., Hedreen, J. C., Peyser, C., Cardillo, J. E., Folstein, S. E., et al. (1992). Locus coeruleus involvement in Huntington's disease. Arch. Neurol. 49, 152–156. doi: 10.1001/archneur.1992.00530260052019

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: locus coeruleus, vulnerability, neurodegenerative disease, noradrenaline, biomarker, Alzheimer’s disease

Citation: Farahani F, Anaclet C, Azizi H and Gompf HS (2025) The locus coeruleus, a blue spot for early diagnosis and prognosis of Alzheimer’s disease. Front. Aging Neurosci. 17:1632236. doi: 10.3389/fnagi.2025.1632236

Received: 20 May 2025; Revised: 03 November 2025; Accepted: 12 November 2025;
Published: 01 December 2025.

Edited by:

Stephen D. Ginsberg, Nathan S. Kline Institute for Psychiatric Research, United States

Reviewed by:

James C. Vickers, University of Tasmania, Australia
Elouise Alexandra Koops, Massachusetts General Hospital and Harvard Medical School, United States

Copyright © 2025 Farahani, Anaclet, Azizi and Gompf. 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: Heinrich S. Gompf, aGVpbnJpY2guZ29tcGZAZ21haWwuY29t

ORCID: Heinrich S. Gompf, https://orcid.org/0000-0002-8749-5776

Disclaimer: 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.