Skip to main content

MINI REVIEW article

Front. Physiol., 07 December 2022
Sec. Renal Physiology and Pathophysiology
This article is part of the Research Topic Recent Advances on Renoprotection and Kidney Regeneration View all 10 articles

Role of extracellular matrix components and structure in new renal models in vitro

  • 1Renal and Cardiovascular Physiopathology (FISIOPREN), Aragon’s Health Sciences Institute, Zaragoza, Spain
  • 2Tissue Microenvironment Lab (TME Lab), I3A, University of Zaragoza, Zaragoza, Spain
  • 3Carlos Chagas Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
  • 4Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
  • 5Institute for Health Research Aragon (IIS Aragon), Zaragoza, Spain
  • 6School of Medicine, University of Zaragoza, Zaragoza, Spain

The extracellular matrix (ECM), a complex set of fibrillar proteins and proteoglycans, supports the renal parenchyma and provides biomechanical and biochemical cues critical for spatial-temporal patterning of cell development and acquisition of specialized functions. As in vitro models progress towards biomimicry, more attention is paid to reproducing ECM-mediated stimuli. ECM’s role in in vitro models of renal function and disease used to investigate kidney injury and regeneration is discussed. Availability, affordability, and lot-to-lot consistency are the main factors determining the selection of materials to recreate ECM in vitro. While simpler components can be synthesized in vitro, others must be isolated from animal or human tissues, either as single isolated components or as complex mixtures, such as Matrigel or decellularized formulations. Synthetic polymeric materials with dynamic and instructive capacities are also being explored for cell mechanical support to overcome the issues with natural products. ECM components can be used as simple 2D coatings or complex 3D scaffolds combining natural and synthetic materials. The goal is to recreate the biochemical signals provided by glycosaminoglycans and other signaling molecules, together with the stiffness, elasticity, segmentation, and dimensionality of the original kidney tissue, to support the specialized functions of glomerular, tubular, and vascular compartments. ECM mimicking also plays a central role in recent developments aiming to reproduce renal tissue in vitro or even in therapeutical strategies to regenerate renal function. Bioprinting of renal tubules, recellularization of kidney ECM scaffolds, and development of kidney organoids are examples. Future solutions will probably combine these technologies.

Introduction

Extracellular matrix components provide critical cues for renal cell and tissue functions

The extracellular matrix (ECM) is a biological scaffold holding all cellular tissue components together (Supplementary Figure 1). The main components of kidney ECM are collagen I, proteoglycans, and glycosaminoglycans. The basal membrane (BM) surrounding the renal tubules contains collagen IV, laminins, and fibronectin (Theocharis et al., 2016). ECM composition and dimensionality establish biomechanical and biochemical signals essential for kidney’s development (Clause and Barker, 2013; Loganathan et al., 2020), tissue growth, differentiation (Muncie and Weaver, 2018), and function (Frantz et al., 2010; Manninen, 2015; Loganathan et al., 2020).

The stiffness of the ECM influences organ or tissue differentiation and morphogenesis. ECM stiffness is determined by the material’s elasticity, as measured by the Young’s elastic modulus. Conventional plastic cell culture containers (109 Pa) are stiffer than bone (15–20 106 Pa) and the kidney (5–10 103 Pa). The substrate mechanical properties affect cell adhesion, migration, proliferation, and differentiation (Chen et al., 2014; Melica et al., 2019). Topography and dimensionality, which identify ECM forms, features, and distribution, are linked to cell polarization, actin bundle alignment, cell adhesion, orientation, migration, and morphology (Nur-E-Kamal et al., 2006; Kim et al., 2014; Sciancalepore et al., 2016; Hulshof et al., 2018; Bosch-Fortea et al., 2019) and renal progenitors’ fate (Nur-E-Kamal et al., 2006; Kim et al., 2014; Bosch-Fortea et al., 2019; Walma and Yamada, 2020). Microfabrication methods can reproduce ECM mechanical and physical properties and architectural features, but resolution and complexity are still rudimentary (Le Digabel et al., 2010). Engineered ECM can be tailored to meet cell or tissue-specific needs (Beamish et al., 2017).

ECM components, particularly glycosaminoglycans (GAGs), provide biochemical signals that regulate cell functions and the organization of the ECM itself (Weber et al., 2017) (Table 1). Hyaluronan, the most abundant GAG (Iozzo and Schaefer, 2015; Theocharis et al., 2016), heparan sulphate or chondroitin sulphate (Lelongt and Ronco, 2003) are involved in tissue development, by displaying growth factors spatial-temporal distribution during epithelial branching (Nigam and Bush, 2014). ECM-renal cells interact through ECM-binding transmembrane receptors such as integrins (Chen et al., 2004; Clause and Barker, 2013; Handorf et al., 2015; Bülow and Boor, 2019) or polycystins (Nickel et al., 2002) that translate ECM biomechanical features into intracellular signals (Hagelaars et al., 2022). In diabetic nephropathy (Kolset et al., 2012) or autosomal dominant polycystic kidney disease (ADPKD) (Zhang et al., 2020), ECM disruptions result in alterations in kidney function. Changes in ECM synthesis and turnover of laminin, heparan sulphate, and chondroitin sulphate proteoglycans contribute to disease pathogenesis (Zhang et al., 2020). Unresolved renal parenchyma damage causes scarring by abnormal deposition of ECM. Chronic damage or excessive scarring leads to fibrosis, a hallmark of chronic kidney disease (Clause and Barker, 2013; Bülow and Boor, 2019). Any in vitro model aiming to accurately represent kidney function, damage, and regeneration should incorporate the ECM compartment. We review ECM’s role in in vitro kidney models. Established models are briefly discussed to better understand the advantages of new methodological developments.

TABLE 1
www.frontiersin.org

TABLE 1. Role of ECM in in vitro models of renal function and disease.

Extracellular matrix sources for in vitro models

In vivo, stromal cells (fibroblasts) produce ECM, and renal epithelial cells contribute themselves to BM synthesis, which can be exploited in in vitro models (Satyam et al., 2020). Simple ECM proteins like laminins are commercially available as recombinant proteins with proven utility for in vitro kidney models (Karamessinis et al., 2002; Chung et al., 2008; Zhang et al., 2009; Sebinger et al., 2013; Homan et al., 2019; Adelfio et al., 2020). However, most in vitro research uses ECM extracts from animal tissues because it is difficult to make complex macromolecular GAGs and large proteoglycans (Petkau-Milroy and Brunsveld, 2013; Aisenbrey and Murphy, 2020; Xing et al., 2020). Stroma-rich tissues like bone or cartilage can yield large quantities of pure ECM components. Commercial sources for human and animal collagen in various isoforms, hyaluronic acid, and fibronectin are available.

The biochemical complexity present in the original tissue is required to induce or maintain a specific phenotype. Here it is best to use complex, unfractionated tissue extracts containing a complex mix of glycosaminoglycans and other signaling molecules. Several commercial products, the best known being Matrigel (Kleinman and Martin, 2005; Passaniti et al., 2021), are readily available in different formulations (e.g., reduced growth factors). There are many examples of in vitro renal models employing such extracts (Zhang et al., 2009; Lam et al., 2014; Takasato et al., 2015; Figliuzzi et al., 2017; King et al., 2017; Hiraki et al., 2018; Howden et al., 2019; Otero et al., 2020) (Supplementary Table S1). Matrigel complexity (contains laminin, collagen IV, entactin, heparan sulfate proteoglycan and bound growth factors) yields better results than gelatin, collagen I, poly-L-lysine, and laminin alone (Hughes et al., 2010; Gao et al., 2011; Passaniti et al., 2021).

Matrigel-like products are expensive, batch-variable, ethically questionable (made from tumors grown in animals) and cannot be employed in human cell-therapy downstream applications. To solve recent availability and ethics-related issues, JellaGel, made from jellyfish Collagen 0 isolates, has recently become available. However, this formulation does not fully mimic the kidney ECM’s specific proteomic signature. ECM extracts from decellularized human kidneys can capture this specificity (Figliuzzi et al., 2017; Hiraki et al., 2018).

In recent years, artificial ECMs have been designed to replace natural ECM for renal epithelium scaffolding to reduce batch variability and degradation. This alternative allows more control over biochemical and mechanical properties and functionalization with instructive biomolecular tags to enhance cell attachment, proliferation, and differentiation (Aisenbrey and Murphy, 2020). Synthetic ECMs can be formed as hydrogels (Minuth et al., 2004; Chung et al., 2008; Astashkina et al., 2012) or hollow fibers (Dankers et al., 2011; Jansen et al., 2015; Shen et al., 2015; Chevtchik et al., 2016; Jansen et al., 2019; Xie et al., 2020; Myram et al., 2021) (Supplementary Table S1). The goal is to obtain a material whose composition can be tailored to control physiochemical matrix properties such as elasticity (Love et al., 2019), density, and stiffness, while ensuring low degradation under specific conditions (Petkau-Milroy and Brunsveld, 2013; Cruz-Acuña et al., 2019).

Conventional models of kidney function and disease

Two-dimensional renal cell culture on extracellular matrix-Coated surfaces

In the simplest culture configuration, renal cells grew directly on plastic surfaces as two-dimensional (2D) epithelial monolayers (Figure 1A). Adsorbing (coating) ECM components on plastic surfaces enhances renal cell adhesion, proliferation, and differentiation (Narayanan et al., 2013; van Gaal et al., 2021). Matrigel’s complex set of biochemical signals is used when cell differentiation is the goal (Narayanan et al., 2013). ECM coatings are also used to functionalize synthetic scaffolds (Chung et al., 2008; Ni et al., 2011). Relevant examples of 2D in vitro kidney models are provided in Table 1. However, lack of complex cell interactions can lead to undesired effects, such as epithelial-to-mesenchymal transition (EMT) (Forino et al., 2006).

FIGURE 1
www.frontiersin.org

FIGURE 1. Simple models in vitro of renal epithelia employ ECM components of basal membrane (BM) and extracellular matrix (ECM) Coatings are simply ECM materials, usually collagen, adsorbed to the plastic (A) or permeable membrane (B). Permeable membranes and scaffold-based models improve epithelial polarity by offering two fluid compartments. Tubulogenesis is stimulated by growing renal epithelial cells onto (C) or within (D) hydrogels. New technologies [microfluidics (E), bioprinting (F), ECM decellularization (G) and organoids development (H)] enable complex models that introduce cell heterogeneity, vascular and interstitial compartments, and biomechanical stimuli. Compared to simpler, conventional models, these models have disadvantages related to their complexity. However, complex models allow for more faithful modeling of kidney function and disease. A licensed version of BioRender was used to prepare this figure.

Epithelial 2D monolayers are frequently grown on permeable supports, like the Transwell system, to promote cell polarization (Gao et al., 2011; Ni et al., 2011; Shamir and Ewald, 2014; Schophuizen et al., 2015; Hulshof et al., 2018) (Figure 1B). Porous membranes can be coated with ECM (Ni et al., 2011; Shamir and Ewald, 2014) or used as scaffolds for thin hydrogels to improve mechanical properties (Shamir and Ewald, 2014) (Table 1). This configuration also facilitates co-culture with other kidney-relevant components (King et al., 2017).

When grown on top of hydrogels, kidney primary cells (Zhang et al., 2011) and most renal cell lines form tubular structures (tubulogenesis; termed 2.5D architecture; Figure 1C). Formation of tubules requires adding Matrigel to the hydrogel and/or to the medium, stressing the need for specific ECM chemical signals (Shamir and Ewald, 2014; Bosch-Fortea et al., 2019). Tubulogenesis studies on the MDCK cell line have been instrumental for understanding molecular the processes involved in epithelial differentiation and polarization (Bosch-Fortea et al., 2019). Hagelaars et al. have recently used this model to show cellular differences in how matrix stiffness affects integrin-mediated cell-ECM attachment and cell polarization (Hagelaars et al., 2022). Better differentiation can happen when the tubule is fully embedded in the ECM (3D architecture, discussed below), however, direct exposure to culture medium in 2.5D structures facilitates imaging and biochemical treatments and assays.

Three-dimensional renal cell culture in hydrogel and scaffolds

Primary or continuous renal cell lines grown within collagen I or Matrigel hydrogels spontaneously form tubule-like structures (Taub et al., 1990; Sakurai et al., 1997; Zegers et al., 2003; Joraku et al., 2009; Schlüter and Margolis, 2009) (Figure 1D). Dissociated cells are mixed with biocompatible hydrogels in liquid form and allowed to polymerize (Miya et al., 2011; DesRochers et al., 2013). Alternatively, cells can be sandwiched between two ECM layers (Montesano et al., 1991a; Montesano et al., 1991b; Kher et al., 2011; Secker et al., 2018; Dixon et al., 2020) In the presence of appropriate factors, cells form hollow cysts (Zegers et al., 2003; Schlüter and Margolis, 2009) and continue to differentiate by elongation (Joraku et al., 2009) and tubule branching. Different nephron segments, including the glomerulus, have been modeled in 3D hydrogels (Joraku et al., 2009; Lü et al., 2012; Hirashima et al., 2017).

3D-culture allows co-culturing of different renal cell lineages, introducing complexity in kidney models (Montesano et al., 1991a; Montesano et al., 1991b; Secker et al., 2018). Wang et al. showed that using a sandwich 3D co-culture model is possible to recreate in vitro the tubule-interstitial-vascular unit, with more tubular cell polarity and enhanced functional gene expression (Wang et al., 2020). Gelatin-fibrin hydrogels outperformed conventional Transwell co-culture in modeling the proximal tubule-vascular unit in vitro (Lin et al., 2019). Tubulointerstitial fibrosis, a common feature in chronic kidney disease, has been modeled in 3D co-cultures (Moll et al., 2013).

Collagen I and Matrigel-like ECM extracts are often used, alone, mixed, or combined with other ECM components like collagen IV, fibronectin, or laminins (Weber et al., 2017). Functionality depends on the 3D hydrogel’s composition, protein concentration, and stiffness (Shamir and Ewald, 2014; Hirashima et al., 2017; Hiraki et al., 2018). Hyaluronic acid is an interesting alternative to tissue extracts because it provides good mechanical and biochemical stimuli. 3D organoids containing proximal tubule structures were generated from mouse kidney explants by prolonged (6 weeks) culture in hyaluronic acid hydrogels resembling in vivo environment (Astashkina et al., 2012). This model has been successfully employed in the preclinical evaluation of nanoparticle nephrotoxicity (Astashkina et al., 2014). Hyaluronic acid modulated ureteric bud branching and promoted mesenchymal-to-epithelial transition (Rosines et al., 2007). The polarity of tubular structures obtained by growing human renal cells (HK-2, ciPTEC, and primary proximal tubule cells) in glycosaminoglycan-based hydrogels was found to depend on sulphated GAGs (Weber et al., 2017). Matrix stiffness strongly affects tubulogenesis in MDCK cells (Hirashima et al., 2017). Such studies (Table 1) exemplify how morphogenesis and function can be modulated by adjusting hydrogel degradability, growth factor signaling, and mechanics.

ECM-derived hydrogels have low resistance to mechanical stress, partly a consequence of active cell remodeling, limiting their use. A potential solution is to exploit the mechanical properties of natural (silk, alginate) or synthetic (PEG, PCL, PLA) polymers to complement natural ECM components (Chung et al., 2008; Enemchukwu et al., 2016; Gupta et al., 2019) (Supplementary Table S2). Plastic materials are easily deposited in 2D or 3D structures by electrospinning or printing techniques (Dankers et al., 2011; Sobreiro-Almeida et al., 2020; van Genderen et al., 2021). Murine PKD1 knockout renal tubular cells seeded in silk-scaffolds filled with Matrigel and collagen hydrogels has been shown to reproduce morphological and functional abnormalities present in Autosomal Dominant Polycystic Kidney disease (ADPKD) (Subramanian et al., 2010; Subramanian et al., 2012).

Recently, 3D models have gained momentum with microfabrication techniques. The organ-on-a-chip technology aims to mimic in vivo tissue architecture by providing independent but connected compartments (Rayner et al., 2018) (Figure 1E). Models based on microfabricated devices recapitulate intercellular and cell-ECM interactions at the microscale. Microfluidics integration adds flow-mediated shear stress, a critical mechanical stimulus for the renal tubule (Jang et al., 2013). Mimetas Organoplate allows for a high throughput culture and analysis of 3D tissue units under fluidic stimulus (Schutgens et al., 2019) and it has proven useful in modeling nephrotoxicity (Vormann et al., 2021) and acute kidney injury (Vormann et al., 2022). Commercial organ-on-chip solutions are listed in Supplementary Table S2.

3D models of tubulogenesis have been instrumental in defining chemical and mechanical stimuli involved in ECM-cell interactions and their role in kidney development and function. Lumen access, high-resolution imaging, biochemical studies, and manipulation for functional or nephrotoxicity assays are, however, limited in their architecture.

New strategies in in vitro modeling of kidney function and disease

Bioprinted scaffolds

Bioprinting has recently emerged as a tool for building complex tissue structures. Biocompatible polymers (bioinks) are layered to create 3D structures (Figure 1F). Cells are seeded on these 3D scaffolds or directly mixed in the bioink. Bioprinting offers unprecedented flexibility and versatility to recreate in vivo environments at the microscale through stereotaxic control of bioink deposition (Fransen et al., 2021).

Bioinks are chosen for their rheological properties and printing device compatibility. As technology evolves, the use of bioinks derived from native tissue like collagen and decellularized ECM is favored (Garreta et al., 2017; Ali et al., 2019; Dzobo et al., 2019; Sobreiro-Almeida et al., 2021). This enhances kidney-specific gene expression by providing tissue-specific biochemical cues (Singh et al., 2020). However, because ECM bioinks exhibit poor mechanical stability, they are often combined with other polymers like methacrylate (Ali et al., 2019).

Using bioprinting, perfused renal tubules have been successfully made. Lewis’s group used fugitive ink to cast tubular conduits within hydrogels, which they populated with proximal tubule cells or endothelial cells and perfused in a closed circuit for days. These tubular-vascular units expressed differentiated phenotypes, and their response to pathogenic insults mimicked those observed in native human tissues (Homan et al., 2016; Lin et al., 2019; Aceves et al., 2022). Tröndle et al. recently reported a modified 3D sandwich model in which renal cells were bioprinted as clusters at a controlled topography on a collagen and Matrigel substrate gel. Cell clusters formed lumen-containing spheroids, which coalesced into tubular structures that could be connected to fluidic systems (Tröndle et al., 2021). Both strategies use fibrin polymers to improve hydrogel biomechanics. A third strategy used a proprietary bioprinting technology (the Organovo 3D printing platform) to sequentially print epithelial tubule, fibroblast-containing ECM, and endothelial vessels (King et al., 2017). A glomerular functional unit was successfully recreated by printing hollow tubules from a functional hybrid bioink (alginate plus decellularized ECM) (Singh et al., 2020).

Bioprinting’s flexibility and automatization capabilities make it a promising method for in vitro modeling of the kidney’s basic functional unit. An immediate challenge is to make it affordable for the general laboratory.

Decellularized kidney as a tissue-specific scaffold

Regenerative medicine has long sought to fabricate a functional kidney using a donor’s decellularized ECM scaffold repopulated with host cells to mitigate the shortage of organs available for transplant (Sullivan et al., 2012). The technique involves perfusing whole kidneys with detergent solutions to remove cells and preserve ECM microscopic architecture and tissue-specific ECM components like collagens and laminins, as well as basement membranes (Song et al., 2013) (Figure 1G). Conservation of signaling molecules, namely glycosaminoglycans, requires proper detergent composition and perfusion rates and timing (Caralt et al., 2015; Poornejad et al., 2016; He et al., 2017; Kajbafzadeh et al., 2019; Zhou et al., 2020; Shahraki et al., 2022). Decellularization can be successfully applied to stored frozen tissues (Chani et al., 2017). Decellularized scaffolds have been successfully repopulated with pluripotent, progenitor, epithelial, or endothelial cells. When implanted in animal models, this bioengineered tissue integrates with host structures and shows some kidney functions (Bonandrini et al., 2014; Caralt et al., 2015; Figliuzzi et al., 2017; Ciampi et al., 2019; Han et al., 2019; Zhang et al., 2019) or helps to revert EMT and fibrosis (Hu et al., 2020).

Regenerating a fully functional organ from a decellularized scaffold is a formidable challenge because of the kidney’s high structural and functional complexity. Nevertheless, studies on kidney decellularization have provided valuable information on cell-ECM interactions, supporting GAGs’ critical role (Louzao-Martinez et al., 2019; Ullah et al., 2020). Moreover, decellularized scaffolds are useful for in vitro method development. Decellularized kidney sections serve as scaffolds for growing renal cells in nephrotoxicity models (Fedecostante et al., 2018). These scaffolds allow for the investigation of cell-ECM interactions in specific organ or tissue microdomains. For example, the fate of pluripotent or progenitor cells in a recellularized scaffold can be followed to learn about specific cell differentiation determinants (Du et al., 2016; Bombelli et al., 2018; Zhang et al., 2019; Bombelli et al., 2020; Ullah et al., 2020).

An acid hydrolysate of decellularized kidney scaffolds, termed dKECM, can be used as a source of tissue-specific ECM materials for surface coating and hydrogel fabrication (Hiraki et al., 2018; Zhou et al., 2020; Shen et al., 2021; Lee et al., 2022). Combining dKECM with other natural or synthetic compounds can enhance their rheological or biophysical properties (Lih et al., 2019; Sobreiro-Almeida et al., 2019; Sobreiro-Almeida et al., 2020; Geng et al., 2021; Ko et al., 2021; Sobreiro-Almeida et al., 2021). Accordingly, dKECM is becoming a favorite bioink in bioprinting applications (Ali et al., 2019; Han et al., 2019). The undesired effects observed when growing human glomerular endothelial cells within hydrogels made of porcine dKECM (Su et al., 2018) illustrates the remarkable specificity of biochemical signals delivered by ECM.

Role of ECM in kidney organoids development

Two strategies are currently used to develop 3D renal structures from progenitor or pluripotent cells by exploiting kidney development programs. Tubuloids are generated from primary cells and kidney organoids from pluripotent stem cells. Both situations require ECM components. Tubuloid culture is a refined version of 3D culture in Matrigel hydrogels where specific biochemical factors are added to stimulate progenitor cell proliferation and differentiation (Schutgens et al., 2019; Wiraja et al., 2021). Human tubuloid culture allows for long-term propagation of donor-specific primary kidney epithelium without requiring immortalization or genetic modification. A recent study comparing the polarization of tubuloid-derived cells and MDCK cells in response to substrate stiffness demonstrated tubuloid-derived cells appear to have different requirements and use different polarization mechanisms (Hagelaars et al., 2022). Unlike tubuloids, immortalized, well-established cell lines have been selected to grow on plastic substrates. By skipping the phase of culture on a stiff substrate, tubuloids might retain more of their physiological responses to ECM. This makes them a simple and affordable alternative cell source for in vitro models.

In the organoid technique (Figure 1H), Matrigel (Xia et al., 2013; Kang and Han, 2014; Takasato et al., 2015; Takasato and Little, 2017; Howden et al., 2019; Low et al., 2019) or Geltrex (Lam et al., 2014; Morizane et al., 2015; Morizane and Bonventre, 2017) coatings or hydrogels are used in feeder-free culture of stem cells or at several differentiation steps. For instance, Taguchi et al. used a 50% Matrigel culture medium to stimulate branching morphogenesis in ureteric buds and to induce interactions with nephron progenitors (Taguchi and Nishinakamura, 2017). Freedman et al. induced epiblast spheroids differentiation by sandwiching hPSC between two layers of diluted Matrigel (Freedman et al., 2015). Under the appropriate concentration and timing of specific biochemical inducers, complex self-organized 3D structures develop.

Organoids contain kidney parenchyma and stroma components, and the synthesis of ECM has been observed (Lam et al., 2014; Takasato et al., 2015; Howden et al., 2019). Given the complexity and animal origin of Matrigel and similar products, there have been efforts to replace it with recombinant ECM proteins, such as laminins (Howden et al., 2019; Mae et al., 2020) or vitronectin (van den Berg et al., 2018), or synthetic products like Synthemax (Toyohara et al., 2015). Recently, Geunes et al. cultured kidney organoids in thiol-ene cross-linked alginate hydrogels and showed a reduction in the onset of aberrant ECM expression and off-target cell populations (Geuens et al., 2021). By engineering gel mechanics and dynamics, ECM deposition and organoid maturation could be tuned, highlighting the role of engineered matrices in stirring organoid commitment (Ruiter et al., 2022).

Incomplete maturation and lack of vascularization are unsolved issues in organoid development where a proper selection and use of ECM components could help. Garreta et al. demonstrated that ECM biophysical properties modulate hPSC proliferation and differentiation (Garreta et al., 2019). Soft hydrogels with stiffness in the physiological range better mimic the early stages of embryonic development. Vascular compartment expression improved when organoids were grown in hydrogels made from decellularized human kidney extracellular matrix (Kim et al., 2022). Bioprinting cellular bioinks allows for precise and reproducible manipulation of organoid size and more differentiated cells (Howden et al., 2019; Lawlor et al., 2021). Perfusion of organoids in microfluidic devices induces higher expression of vascular and podocyte compartments (Homan et al., 2016; Lee et al., 2021). High levels of structural and functional complexity in bioprinted, perfused organoids model more faithfully renal function and disease, as shown recently for APKD (Howden et al., 2021; Hiratsuka et al., 2022).

Relevance of ECM-based in vitro models of renal disease for studies of renoprotection and kidney regeneration

Increasing rates of chronic kidney disease (CKD) represent a major burden for social and healthcare systems worldwide. Fighting underlying causes (diabetes, obesity, cardiovascular disease, etc.) is key. But it is equally important to prevent, slow down, or reverse CKD progression, which very often results from maladaptive responses to acute kidney injury.

Shortcomings of traditional preclinical models (animal experimentation and conventional cell culture) have fueled the development of sophisticated in vitro kidney models that take advantage of recent technological advances (Morizane et al., 2015). Only through such complex models is it possible, for instance, to recreate the delicate glomerular filtration barrier, allowing for the investigation of the varied glomerulopathies (Lü et al., 2012; Du et al., 2016; Xie et al., 2020). Sophisticated models, such as organoids, are already being used successfully to study genetic (e.g., ADPKD (Subramanian et al., 2010; Freedman et al., 2015; Dixon et al., 2020; Zhang et al., 2020; Howden et al., 2021; Myram et al., 2021; Hiratsuka et al., 2022)) or metabolic tubulopathies (Fabry’s disease (Kim et al., 2022)). Investigating the mechanisms of drug-related nephrotoxicity and discovering ways to prevent it is frequently the goal behind model design or validation (Astashkina et al., 2012; DesRochers et al., 2013; King et al., 2017; Fedecostante et al., 2018; Vormann et al., 2021; Tröndle et al., 2022). Research on common mechanisms underlying CKD progression, irrespective of its cause, such as epithelial-to-mesenchymal transition (Forino et al., 2006) or fibrosis (Moll et al., 2013; Hu et al., 2020; Li et al., 2022), requires the presence of all participants in such complex processes. Studying tubulogenesis or cell-repair mechanisms in complex in vitro models helps identification of signals needed for kidney regeneration (Miya et al., 2011). These processes, which involve multiple actors from distinct compartments, cannot be studied adequately with conventional in vitro models. Some bioengineering strategies are originally aimed at fabricating tissue-like structures for regenerative techniques, based on the concept of regenerating an entire organ from a decellularized scaffold (Song et al., 2013; Du et al., 2016; Figliuzzi et al., 2017; Ciampi et al., 2019) or by stimulating regeneration in vivo via cell or tissue implants (Lih et al., 2019; Ko et al., 2021; van den Berg et al., 2018; Garreta et al., 2019; Kim et al., 2022).

We have summarized the essential roles of ECM in in vitro kidney function and disease modeling. ECM provides the biochemical and mechanical stimuli required for promoting and maintaining cell differentiation. A proper 3D architecture also permits cell-cell interactions and facilitates the presence of all necessary compartments, including fluid convection, to faithfully mimic in vivo kidney function. More efforts are needed to make the technical skills required simpler and to lower the costs associated with using such models, which would increase their adoption in kidney translational research.

Author contributions

All authors: conceptual design, data adquisition and interpretation, critical revision, final approval of the version to be published.

Funding

Publication of this review has been made possible through funds provided by the Government of Spain (RTI2018-0099946-B-100 to I.G.) and a “2017 Research Fellowship” from Sociedad Española de Nefrología.

Acknowledgments

We would like to thank all the excellent technical assistance provided by the people at our institutions that make possible working with complex and sophisticated cell culture models.

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.

Publisher’s note

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

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fphys.2022.1048738/full#supplementary-material

References

Aceves J. O., Heja S., Kobayashi K., Robinson S. S., Miyoshi T., Matsumoto T., et al. (2022). 3D proximal tubule-on-chip model derived from kidney organoids with improved drug uptake. Sci. Rep. 12 (1), 14997. doi:10.1038/s41598-022-19293-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Adelfio M., Szymkowiak S., Kaplan D. L. (2020). Matrigel-free laminin-entactin matrix to induce human renal proximal tubule structure formation in vitro. ACS Biomater. Sci. Eng. 6 (12), 6618–6625. doi:10.1021/acsbiomaterials.0c01385

PubMed Abstract | CrossRef Full Text | Google Scholar

Aisenbrey E. A., Murphy W. L. (2020). Synthetic alternatives to matrigel. Nat. Rev. Mat. 5 (7), 539–551. doi:10.1038/s41578-020-0199-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Ali M., Pr A. K., Yoo J. J., Zahran F., Atala A., Lee S. J. (2019). A photo-crosslinkable kidney ECM-derived bioink accelerates renal tissue formation. Adv. Healthc. Mat. 8 (7), e1800992. doi:10.1002/adhm.201800992

PubMed Abstract | CrossRef Full Text | Google Scholar

Astashkina A. I., Jones C. F., Thiagarajan G., Kurtzeborn K., Ghandehari H., Brooks B. D., et al. (2014). Nanoparticle toxicity assessment using an invitro 3-D kidney organoid culture model. Biomaterials 35 (24), 6323–6331. doi:10.1016/j.biomaterials.2014.04.060

PubMed Abstract | CrossRef Full Text | Google Scholar

Astashkina A. I., Mann B. K., Prestwich G. D., Grainger D. W. (2012). A 3-D organoid kidney culture model engineered for high-throughput nephrotoxicity assays. Biomaterials 33 (18), 4700–4711. doi:10.1016/j.biomaterials.2012.02.063

PubMed Abstract | CrossRef Full Text | Google Scholar

Beamish J. A., Chen E., Putnam A. J. (2017). Engineered extracellular matrices with controlled mechanics modulate renal proximal tubular cell epithelialization. PLoS One 12 (7), e0181085. doi:10.1371/journal.pone.0181085

PubMed Abstract | CrossRef Full Text | Google Scholar

Bombelli S., Meregalli C., Grasselli C., Bolognesi M. M., Bruno A., Eriani S., et al. (2020). PKHhigh/CD133+/CD24- renal stem-like cells isolated from human nephrospheres exhibit in vitro multipotency. Cells 9 (8), E1805. doi:10.3390/cells9081805

PubMed Abstract | CrossRef Full Text | Google Scholar

Bombelli S., Meregalli C., Scalia C., Bovo G., Torsello B., De Marco S., et al. (2018). Nephrosphere-derived cells are induced to multilineage differentiation when cultured on human decellularized kidney scaffolds. Am. J. Pathol. 188 (1), 184–195. doi:10.1016/j.ajpath.2017.09.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Bonandrini B., Figliuzzi M., Papadimou E., Morigi M., Perico N., Casiraghi F., et al. (2014). Recellularization of well-preserved acellular kidney scaffold using embryonic stem cells. Tissue Eng. Part A 20 (9–10), 1486–1498. doi:10.1089/ten.TEA.2013.0269

PubMed Abstract | CrossRef Full Text | Google Scholar

Bosch-Fortea M., Rodriguez-Fraticelli A. E., Herranz G., Hachimi M., Barea M. D., Young J., et al. (2019). Micropattern-based platform as a physiologically relevant model to study epithelial morphogenesis and nephrotoxicity. Biomaterials 218, 119339. doi:10.1016/j.biomaterials.2019.119339

PubMed Abstract | CrossRef Full Text | Google Scholar

Bülow R. D., Boor P. (2019). Extracellular matrix in kidney fibrosis: More than just a scaffold. J. Histochem. Cytochem. 67 (9), 643–661. doi:10.1369/0022155419849388

PubMed Abstract | CrossRef Full Text | Google Scholar

Caralt M., Uzarski J. S., Iacob S., Obergfell K. P., Berg N., Bijonowski B. M., et al. (2015). Optimization and critical evaluation of decellularization strategies to develop renal extracellular matrix scaffolds as biological templates for organ engineering and transplantation. Am. J. Transpl. 15 (1), 64–75. doi:10.1111/ajt.12999

PubMed Abstract | CrossRef Full Text | Google Scholar

Chani B., Puri V., Sobti R. C., Jha V., Puri S. (2017). Decellularized scaffold of cryopreserved rat kidney retains its recellularization potential. PLoS One 12 (3), 01730400–e173120. doi:10.1371/journal.pone.0173040

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen D., Roberts R., Pohl M., Nigam S., Kreidberg J., Wang Z., et al. (2004). Differential expression of collagen- and laminin-binding integrins mediates ureteric bud and inner medullary collecting duct cell tubulogenesis. Am. J. Physiol. Ren. Physiol. 287 (4 56-4), 602–611. doi:10.1152/ajprenal.00015.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen W. C., Lin H. H., Tang M. J. (2014). Regulation of proximal tubular cell differentiation and proliferation in primary culture by matrix stiffness and ECM components. Am. J. Physiol. Ren. Physiol. 307 (6), F695–F707. doi:10.1152/ajprenal.00684.2013

PubMed Abstract | CrossRef Full Text | Google Scholar

Chevtchik N. V., Fedecostante M., Jansen J., Mihajlovic M., Wilmer M., Rüth M., et al. (2016). Upscaling of a living membrane for bioartificial kidney device. Eur. J. Pharmacol. 790, 28–35. doi:10.1016/j.ejphar.2016.07.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Chung I. M., Enemchukwu N. O., Khaja S. D., Murthy N., Mantalaris A., García A. J. (2008). Bioadhesive hydrogel microenvironments to modulate epithelial morphogenesis. Biomaterials 29 (17), 2637–2645. doi:10.1016/j.biomaterials.2008.03.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Ciampi O., Bonandrini B., Derosas M., Conti S., Rizzo P., Benedetti V., et al. (2019). Engineering the vasculature of decellularized rat kidney scaffolds using human induced pluripotent stem cell-derived endothelial cells. Sci. Rep. 9 (1), 8001. doi:10.1038/s41598-019-44393-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Clause K. C., Barker T. H. (2013). Extracellular matrix signaling in morphogenesis and repair. Curr. Opin. Biotechnol. 24 (5), 830–833. doi:10.1016/j.copbio.2013.04.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Cruz-Acuña R., Mulero-Russe A., Clark A. Y., Zent R., García A. J. (2019). Identification of matrix physicochemical properties required for renal epithelial cell tubulogenesis by using synthetic hydrogels. J. Cell Sci. 132 (20), jcs226639. doi:10.1242/jcs.226639

PubMed Abstract | CrossRef Full Text | Google Scholar

Dankers P. Y. W., Boomker J. M., Huizinga-van der Vlag A., Wisse E., Appel W. P. J., Smedts F. M. M., et al. (2011). Bioengineering of living renal membranes consisting of hierarchical, bioactive supramolecular meshes and human tubular cells. Biomaterials 32 (3), 723–733. doi:10.1016/j.biomaterials.2010.09.020

PubMed Abstract | CrossRef Full Text | Google Scholar

DesRochers T. M., Suter L., Roth A., Kaplan D. L. (2013). Bioengineered 3D human kidney tissue, a platform for the determination of nephrotoxicity. PLoS One 8 (3), e59219. doi:10.1371/journal.pone.0059219

PubMed Abstract | CrossRef Full Text | Google Scholar

Dixon E. E., Maxim D. S., Halperin Kuhns V. L., Lane-Harris A. C., Outeda P., Ewald A. J., et al. (2020). GDNF drives rapid tubule morphogenesis in a novel 3D in vitro model for ADPKD. J. Cell Sci. 133 (14), jcs249557. doi:10.1242/jcs.249557

PubMed Abstract | CrossRef Full Text | Google Scholar

Du C., Narayanan K., Leong M. F., Ibrahim M. S., Chua Y. P., Khoo V. M. H., et al. (2016). Functional kidney bioengineering with pluripotent stem-cell-derived renal progenitor cells and decellularized kidney scaffolds. Adv. Healthc. Mat. 5 (16), 2080–2091. doi:10.1002/adhm.201600120

PubMed Abstract | CrossRef Full Text | Google Scholar

Dzobo K., Motaung K. S. C. M., Adesida A. (2019). Recent trends in decellularized extracellular matrix bioinks for 3D printing: An updated review. Int. J. Mol. Sci. 20 (18), E4628. doi:10.3390/ijms20184628

PubMed Abstract | CrossRef Full Text | Google Scholar

Enemchukwu N. O., Cruz-Acuña R., Bongiorno T., Johnson C. T., García J. R., Sulchek T., et al. (2016). Synthetic matrices reveal contributions of ECM biophysical and biochemical properties to epithelial morphogenesis. J. Cell Biol. 212 (1), 113–124. doi:10.1083/jcb.201506055

PubMed Abstract | CrossRef Full Text | Google Scholar

Fedecostante M., Westphal K. G. C., Buono M. F., Romero N. S., Wilmer M. J., Kerkering J., et al. (2018). Recellularized native kidney scaffolds as a novel tool in nephrotoxicity screening. Drug Metab. Dispos. 46 (9), 1338–1350. doi:10.1124/dmd.118.080721

PubMed Abstract | CrossRef Full Text | Google Scholar

Figliuzzi M., Bonandrini B., Remuzzi A. (2017). Decellularized kidney matrix as functional material for whole organ tissue engineering. J. Appl. Biomater. Funct. Mat. 15 (4), e326–e333. doi:10.5301/jabfm.5000393

PubMed Abstract | CrossRef Full Text | Google Scholar

Forino M., Torregrossa R., Ceol M., Murer L., Della Vella M., Del Prete D., et al. (2006). TGFbeta1 induces epithelial-mesenchymal transition, but not myofibroblast transdifferentiation of human kidney tubular epithelial cells in primary culture. Int. J. Exp. Pathol. 87 (3), 197–208. doi:10.1111/j.1365-2613.2006.00479.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Fransen M. F. J., Addario G., Bouten C. V. C., Halary F., Moroni L., Mota C. (2021). Bioprinting of kidney in vitro models: Cells, biomaterials, and manufacturing techniques. Essays Biochem. 65 (3), 587–602. doi:10.1042/EBC20200158

PubMed Abstract | CrossRef Full Text | Google Scholar

Frantz C., Stewart K. M., Weaver V. M. (2010). The extracellular matrix at a glance. J. Cell Sci. 123 (24), 4195–4200. doi:10.1242/jcs.023820

PubMed Abstract | CrossRef Full Text | Google Scholar

Freedman B. S., Brooks C. R., Lam A. Q., Fu H., Morizane R., Agrawal V., et al. (2015). Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids. Nat. Commun. 6, 8715. doi:10.1038/ncomms9715

PubMed Abstract | CrossRef Full Text | Google Scholar

Gao X., Tanaka Y., Sugii Y., Mawatari K., Kitamori T. (2011). Basic structure and cell culture condition of a bioartificial renal tubule on chip towards a cell-based separation microdevice. Anal. Sci. 27 (9), 907–912. doi:10.2116/analsci.27.907

PubMed Abstract | CrossRef Full Text | Google Scholar

Garreta E., Oria R., Tarantino C., Pla-Roca M., Prado P., Fernández-Avilés F., et al. (2017). Tissue engineering by decellularization and 3D bioprinting. Mat. TodayKidlingt. 20 (4), 166–178. doi:10.1016/j.mattod.2016.12.005

CrossRef Full Text | Google Scholar

Garreta E., Prado P., Tarantino C., Oria R., Fanlo L., Martí E., et al. (2019). Fine tuning the extracellular environment accelerates the derivation of kidney organoids from human pluripotent stem cells. Nat. Mat. 18 (4), 397–405. doi:10.1038/s41563-019-0287-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Geng G., Xiao Y., Shang Y., Zhang Y., Zhu F., Tang L., et al. (2021). Naphthalenephenylalanine-phenylalanine-glycine-arginine-glycine-aspartic promotes self-assembly of nephron progenitor cells in decellularized scaffolds to construct bioengineered kidneys. Biomater. Adv. 134, 112590. doi:10.1016/j.msec.2021.112590

PubMed Abstract | CrossRef Full Text | Google Scholar

Geuens T., Ruiter F. A. A., Schumacher A., Morgan F. L. C., Rademakers T., Wiersma L. E., et al. (2021). Thiol-ene cross-linked alginate hydrogel encapsulation modulates the extracellular matrix of kidney organoids by reducing abnormal type 1a1 collagen deposition. Biomaterials 275, 120976. doi:10.1016/j.biomaterials.2021.120976

PubMed Abstract | CrossRef Full Text | Google Scholar

Gupta A. K., Coburn J. M., Davis-Knowlton J., Kimmerling E., Kaplan D. L., Oxburgh L. (2019). Scaffolding kidney organoids on silk. J. Tissue Eng. Regen. Med. 13 (5), 812–822. doi:10.1002/term.2830

PubMed Abstract | CrossRef Full Text | Google Scholar

Hagelaars M. J., Yousef Yengej F. A., Verhaar M. C., Rookmaaker M. B., Loerakker S., Bouten C. V. C. (2022). Substrate stiffness determines the establishment of apical-basal polarization in renal epithelial cells but not in tubuloid-derived cells. Front. Bioeng. Biotechnol. 10, 820930. doi:10.3389/fbioe.2022.820930

PubMed Abstract | CrossRef Full Text | Google Scholar

Han W., Singh N. K., Kim J. J., Kim H., Kim B. S., Park J. Y., et al. (2019). Directed differential behaviors of multipotent adult stem cells from decellularized tissue/organ extracellular matrix bioinks. Biomaterials 224, 119496. doi:10.1016/j.biomaterials.2019.119496

PubMed Abstract | CrossRef Full Text | Google Scholar

Handorf A. M., Zhou Y., Halanski M. A., Li W. J. (2015). Tissue stiffness dictates development, homeostasis, and disease progression. Organogenesis, 11 (1), 1–15. doi:10.1080/15476278.2015.1019687

PubMed Abstract | CrossRef Full Text | Google Scholar

He M., Callanan A., Lagaras K., Steele J. A. M., Stevens M. M. (2017). Optimization of SDS exposure on preservation of ECM characteristics in whole organ decellularization of rat kidneys. J. Biomed. Mat. Res. B Appl. Biomater. 105 (6), 1352–1360. doi:10.1002/jbm.b.33668

PubMed Abstract | CrossRef Full Text | Google Scholar

Hiraki H. L., Nagao R. J., Himmelfarb J., Zheng Y. (2018). Fabricating a kidney cortex extracellular matrix-derived hydrogel. J. Vis. Exp. 140, e58314. doi:10.3791/58314

PubMed Abstract | CrossRef Full Text | Google Scholar

Hirashima T., Hoshuyama M., Adachi T. (2017). In vitro tubulogenesis of Madin–Darby canine kidney (MDCK) spheroids occurs depending on constituent cell number and scaffold gel concentration. J. Theor. Biol. 435, 110–115. doi:10.1016/j.jtbi.2017.09.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Hiratsuka K., Miyoshi T., Kroll K. T., Gupta N. R., Valerius M. T., Ferrante T., et al. (2022). Organoid-on-a-chip model of human ARPKD reveals mechanosensing pathomechanisms for drug discovery. Sci. Adv. 8 (38), eabq0866. doi:10.1126/sciadv.abq0866

PubMed Abstract | CrossRef Full Text | Google Scholar

Homan K. A., Gupta N., Kroll K. T., Kolesky D. B., Skylar-Scott M., Miyoshi T., et al. (2019). Flow-enhanced vascularization and maturation of kidney organoids in vitro. Nat. Methods 16 (3), 255–262. doi:10.1038/s41592-019-0325-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Homan K. A., Kolesky D. B., Skylar-Scott M. A., Herrmann J., Obuobi H., Moisan A., et al. (2016). Bioprinting of 3D convoluted renal proximal tubules on perfusable chips. Sci. Rep. 6, 34845. doi:10.1038/srep34845

PubMed Abstract | CrossRef Full Text | Google Scholar

Howden S. E., Vanslambrouck J. M., Wilson S. B., Tan K. S., Little M. H. (2019). Reporter-based fate mapping in human kidney organoids confirms nephron lineage relationships and reveals synchronous nephron formation. EMBO Rep. 20 (4), 474833–e47513. doi:10.15252/embr.201847483

PubMed Abstract | CrossRef Full Text | Google Scholar

Howden S. E., Wilson S. B., Groenewegen E., Starks L., Forbes T. A., Tan K. S., et al. (2021). Plasticity of distal nephron epithelia from human kidney organoids enables the induction of ureteric tip and stalk. Cell Stem Cell 28 (4), 671–684.e6. e6. doi:10.1016/j.stem.2020.12.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu D., Zhang D., Liu B., Liu Y., Zhou Y., Yu Y., et al. (2020). Human ucMSCs seeded in a decellularized kidney scaffold attenuate renal fibrosis by reducing epithelial-mesenchymal transition via the TGF-β/Smad signaling pathway. Pediatr. Res. 88 (2), 192–201. doi:10.1038/s41390-019-0736-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Hughes C. S., Postovit L. M., Lajoie G. A. (2010). Matrigel: A complex protein mixture required for optimal growth of cell culture. Proteomics 10 (9), 1886–1890. doi:10.1002/pmic.200900758

PubMed Abstract | CrossRef Full Text | Google Scholar

Hulshof F., Schophuizen C., Mihajlovic M., van Blitterswijk C., Masereeuw R., de Boer J., et al. (2018). New insights into the effects of biomaterial chemistry and topography on the morphology of kidney epithelial cells. J. Tissue Eng. Regen. Med. 12 (2), e817–e827. doi:10.1002/term.2387

PubMed Abstract | CrossRef Full Text | Google Scholar

Iozzo R. V., Schaefer L. (2015). Matrix biology. Proteoglycan Funct. A Compr. Nomencl. proteoglycans 42, 11–55.

Google Scholar

Jang K. J., Mehr A. P., Hamilton G. A., McPartlin L. A., Chung S., Suh K. Y., et al. (2013). Human kidney proximal tubule-on-a-chip for drug transport and nephrotoxicity assessment. Integr. Biol. 5 (9), 1119–1129. doi:10.1039/c3ib40049b

PubMed Abstract | CrossRef Full Text | Google Scholar

Jansen J., De Napoli I. E., Fedecostante M., Schophuizen C. M. S., Chevtchik N. V., Wilmer M. J., et al. (2015). Human proximal tubule epithelial cells cultured on hollow fibers: Living membranes that actively transport organic cations. Sci. Rep. 5 (1), 16702. doi:10.1038/srep16702

PubMed Abstract | CrossRef Full Text | Google Scholar

Jansen K., Castilho M., Aarts S., Kaminski M. M., Lienkamp S. S., Pichler R., et al. (2019). Fabrication of kidney proximal tubule grafts using biofunctionalized electrospun polymer scaffolds. Macromol. Biosci. 19 (2), e1800412–e1800419. doi:10.1002/mabi.201800412

PubMed Abstract | CrossRef Full Text | Google Scholar

Joraku A., Stern K. A., Atala A., Yoo J. J. (2009). In vitro generation of three-dimensional renal structures. Methods 47 (2), 129–133. doi:10.1016/j.ymeth.2008.09.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Kajbafzadeh A. M., Khorramirouz R., Nabavizadeh B., Ladi Seyedian S. S., Akbarzadeh A., Heidari R., et al. (2019). Whole organ sheep kidney tissue engineering and in vivo transplantation: Effects of perfusion-based decellularization on vascular integrity. Mat. Sci. Eng. C Mat. Biol. Appl. 98, 392–400. doi:10.1016/j.msec.2019.01.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Kang M., Han Y. M. (2014). Differentiation of human pluripotent stem cells into nephron progenitor cells in a serum and feeder free system. PLoS One 9 (4), e94888. doi:10.1371/journal.pone.0094888

PubMed Abstract | CrossRef Full Text | Google Scholar

Karamessinis P. M., Tzinia A. K., Kitsiou P. V., Stetler-Stevenson W. G., Michael A. F., Fan W-W., et al. (2002). Proximal tubular epithelial cell integrins respond to high glucose by altered cell-matrix interactions and differentially regulate matrixin expression. Lab. Invest. 82 (8), 1081–1093. doi:10.1097/01.lab.0000022224.86237.31

PubMed Abstract | CrossRef Full Text | Google Scholar

Kher R., Sha E. C., Escobar M. R., Andreoli E. M., Wang P., Xu W. M., et al. (2011). Ectopic expression of cadherin 8 is sufficient to cause cyst formation in a novel 3D collagen matrix renal tubule culture. Am. J. Physiol. Cell Physiol. 301 (1), C99–C105. doi:10.1152/ajpcell.00151.2010

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim J. W., Nam S. A., Yi J., Kim J. Y., Lee J. Y., Park S. Y., et al. (2022). Kidney decellularized extracellular matrix enhanced the vascularization and maturation of human kidney organoids. Adv. Sci. 9 (15), e2103526. doi:10.1002/advs.202103526

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim M-H., Sawada Y., Taya M., Kino-Oka M. (2014). Influence of surface topography on the human epithelial cell response to micropatterned substrates with convex and concave architectures. J. Biol. Eng. 8 (1), 13. doi:10.1186/1754-1611-8-13

PubMed Abstract | CrossRef Full Text | Google Scholar

King S. M., Higgins J. W., Nino C. R., Smith T. R., Paffenroth E. H., Fairbairn C. E., et al. (2017). 3D proximal tubule tissues recapitulate key aspects of renal physiology to enable nephrotoxicity testing. Front. Physiol. 8 (MAR), 123. doi:10.3389/fphys.2017.00123

PubMed Abstract | CrossRef Full Text | Google Scholar

Kleinman H. K., Martin G. R. (2005). Matrigel: Basement membrane matrix with biological activity. Semin. Cancer Biol. 15 (5), 378–386. doi:10.1016/j.semcancer.2005.05.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Ko K. W., Park S. Y., Lee E. H., Yoo Y. I., Kim D. S., Kim J. Y., et al. (2021). Integrated bioactive scaffold with polydeoxyribonucleotide and stem-cell-derived extracellular vesicles for kidney regeneration. ACS Nano 15 (4), 7575–7585. doi:10.1021/acsnano.1c01098

PubMed Abstract | CrossRef Full Text | Google Scholar

Kolset S. O., Reinholt F. P., Jenssen T. (2012). Diabetic nephropathy and extracellular matrix. J. Histochem. Cytochem. 60 (12), 976–986. doi:10.1369/0022155412465073

PubMed Abstract | CrossRef Full Text | Google Scholar

Lam A. Q., Freedman B. S., Morizane R., Lerou P. H., Valerius M. T., Bonventre J. V. (2014). Rapid and efficient differentiation of human pluripotent stem cells into intermediate mesoderm that forms tubules expressing kidney proximal tubular markers. J. Am. Soc. Nephrol. 25 (6), 1211–1225. doi:10.1681/ASN.2013080831

PubMed Abstract | CrossRef Full Text | Google Scholar

Lawlor K. T., Vanslambrouck J. M., Higgins J. W., Chambon A., Bishard K., Arndt D., et al. (2021). Cellular extrusion bioprinting improves kidney organoid reproducibility and conformation. Nat. Mat. 20 (2), 260–271. doi:10.1038/s41563-020-00853-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Le Digabel J., Ghibaudo M., Trichet L., Richert A., Ladoux B. (2010). Microfabricated substrates as a tool to study cell mechanotransduction. Med. Biol. Eng. Comput. 48 (10), 965–976. doi:10.1007/s11517-010-0619-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee E. H., Chun S. Y., Yoon B. H., Kim H. T., Chung J-W., Lee J. N., et al. (2022). Application of porcine kidney-derived extracellular matrix as coating, hydrogel, and scaffold material for renal proximal tubular epithelial cell. Biomed. Res. Int. 2022, 2220641. doi:10.1155/2022/2220641

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee H. N., Choi Y. Y., Kim J. W., Lee Y. S., Choi J. W., Kang T., et al. (2021). Effect of biochemical and biomechanical factors on vascularization of kidney organoid-on-a-chip. Nano Converg. 8 (1), 35. doi:10.1186/s40580-021-00285-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Lelongt B., Ronco P. (2003). Role of extracellular matrix in kidney development and repair. Pediatr. Nephrol. 18, 731–742. doi:10.1007/s00467-003-1153-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Li L., Fu H., Liu Y. (2022). The fibrogenic niche in kidney fibrosis: Components and mechanisms. Nat. Rev. Nephrol. 18 (9), 545–557. doi:10.1038/s41581-022-00590-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Lih E., Park W., Park K. W., Chun S. Y., Kim H., Joung Y. K., et al. (2019). A bioinspired scaffold with anti-inflammatory magnesium hydroxide and decellularized extracellular matrix for renal tissue regeneration. ACS Cent. Sci. 5 (3), 458–467. doi:10.1021/acscentsci.8b00812

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin N. Y. C., Homan K. A., Robinson S. S., Kolesky D. B., Duarte N., Moisan A., et al. (2019). Renal reabsorption in 3D vascularized proximal tubule models. Proc. Natl. Acad. Sci. U. S. A. 116 (12), 5399–5404. doi:10.1073/pnas.1815208116

PubMed Abstract | CrossRef Full Text | Google Scholar

Loganathan R., Little C. D., Rongish B. J. (2020). Extracellular matrix dynamics in tubulogenesis. Cell. Signal. 72, 109619. doi:10.1016/j.cellsig.2020.109619

PubMed Abstract | CrossRef Full Text | Google Scholar

Louzao-Martinez L., van Dijk C. G. M., Xu Y. J., Korn A., Bekker N. J., Brouwhuis R., et al. (2019). A proteome comparison between human fetal and mature renal extracellular matrix identifies EMILIN1 as a regulator of renal epithelial cell adhesion. Matrix Biol. Plus 4, 100011. doi:10.1016/j.mbplus.2019.100011

PubMed Abstract | CrossRef Full Text | Google Scholar

Love H. D., Ao M., Jorgensen S., Swearingen L., Ferrell N., Evans R., et al. (2019). Substrate elasticity governs differentiation of renal tubule cells in prolonged culture. Tissue Eng. Part A 25 (13–14), 1013–1022. doi:10.1089/ten.TEA.2018.0182

PubMed Abstract | CrossRef Full Text | Google Scholar

Low J. H., Li P., Chew E. G. Y., Zhou B., Suzuki K., Zhang T., et al. (2019). Generation of human PSC-derived kidney organoids with patterned nephron segments and a de novo vascular network. Cell Stem Cell 25 (3), 373–387. e9. doi:10.1016/j.stem.2019.06.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Lü S. H., Lin Q., Liu Y. N., Gao Q., Hao T., Wang Y., et al. (2012). Self-assembly of renal cells into engineered renal tissues in collagen/Matrigel scaffold in vitro. J. Tissue Eng. Regen. Med. 6 (10), 786–792. doi:10.1002/term.484

PubMed Abstract | CrossRef Full Text | Google Scholar

Mae S-I., Ryosaka M., Sakamoto S., Matsuse K., Nozaki A., Igami M., et al. (2020). Expansion of human iPSC-derived ureteric bud organoids with repeated branching potential. Cell Rep. 32 (4), 107963. doi:10.1016/j.celrep.2020.107963

PubMed Abstract | CrossRef Full Text | Google Scholar

Manninen A. (2015). Epithelial polarity - generating and integrating signals from the ECM with integrins. Exp. Cell Res. 334 (2), 337–349. doi:10.1016/j.yexcr.2015.01.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Melica M. E., La Regina G. L., Parri M., Peired A. J., Romagnani P., Lasagni L. (2019). Substrate stiffness modulates renal progenitor cell properties via a ROCK-mediated mechanotransduction mechanism. Cells 8 (12), E1561. doi:10.3390/cells8121561

PubMed Abstract | CrossRef Full Text | Google Scholar

Minuth W. W., Sorokin L., Schumacher K. (2004). Generation of renal tubules at the interface of an artificial interstitium. Cell. Physiol. biochem. 14 (4–6), 387–394. doi:10.1159/000080348

PubMed Abstract | CrossRef Full Text | Google Scholar

Miya M., Maeshima A., Mishima K., Sakurai N., Ikeuchi H., Kuroiwa T., et al. (2011). Enhancement of in vitro human tubulogenesis by endothelial cell-derived factors: Implications for in vivo tubular regeneration after injury. Am. J. Physiol. Ren. Physiol. 301 (2), 387–395. doi:10.1152/ajprenal.00619.2010

PubMed Abstract | CrossRef Full Text | Google Scholar

Moll S., Ebeling M., Weibel F., Farina A., Araujo Del Rosario A., Hoflack J. C., et al. (2013). Epithelial cells as active player in fibrosis: Findings from an in vitro model. PLoS One 8 (2), e56575. doi:10.1371/journal.pone.0056575

PubMed Abstract | CrossRef Full Text | Google Scholar

Montesano R., Matsumoto K., Nakamura T., Orci L. (1991). Identification of a fibroblast-derived epithelial morphogen as hepatocyte growth factor. Cell 67 (5), 901–908. doi:10.1016/0092-8674(91)90363-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Montesano R., Schaller G., Orci L. (1991). Induction of epithelial tubular morphogenesis in vitro by fibroblast-derived soluble factors. Cell 66 (4), 697–711. doi:10.1016/0092-8674(91)90115-f

PubMed Abstract | CrossRef Full Text | Google Scholar

Morizane R., Bonventre J. V. (2017). Generation of nephron progenitor cells and kidney organoids from human pluripotent stem cells. Nat. Protoc. 12 (1), 195–207. doi:10.1038/nprot.2016.170

PubMed Abstract | CrossRef Full Text | Google Scholar

Morizane R., Lam A. Q., Freedman B. S., Kishi S., Valerius M. T., Bonventre J. V. (2015). Nephron organoids derived from human pluripotent stem cells model kidney development and injury. Nat. Biotechnol. 33 (11), 1193–1200. doi:10.1038/nbt.3392

PubMed Abstract | CrossRef Full Text | Google Scholar

Muncie J. M., Weaver V. M. (2018). The physical and biochemical properties of the extracellular matrix regulate cell fate. Curr. Top. Dev. Biol. 130, 1–37. doi:10.1016/bs.ctdb.2018.02.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Myram S., Venzac B., Lapin B., Battistella A., Cayrac F., Cinquin B., et al. (2021). A multitubular kidney-on-chip to decipher pathophysiological mechanisms in renal cystic diseases. Front. Bioeng. Biotechnol. 9, 624553. doi:10.3389/fbioe.2021.624553

PubMed Abstract | CrossRef Full Text | Google Scholar

Narayanan K., Schumacher K. M., Tasnim F., Kandasamy K., Schumacher A., Ni M., et al. (2013). Human embryonic stem cells differentiate into functional renal proximal tubular-like cells. Kidney Int. 83 (4), 593–603. doi:10.1038/ki.2012.442

PubMed Abstract | CrossRef Full Text | Google Scholar

Ni M., Teo J. C. M., Ibrahim M. S., Zhang K., Tasnim F., Chow P-Y., et al. (2011). Characterization of membrane materials and membrane coatings for bioreactor units of bioartificial kidneys. Biomaterials 32 (6), 1465–1476. doi:10.1016/j.biomaterials.2010.10.061

PubMed Abstract | CrossRef Full Text | Google Scholar

Nickel C., Benzing T., Sellin L., Gerke P., Karihaloo A., Liu Z-X., et al. (2002). The polycystin-1 C-terminal fragment triggers branching morphogenesis and migration of tubular kidney epithelial cells. J. Clin. Invest. 109 (4), 481–489. doi:10.1172/JCI12867

PubMed Abstract | CrossRef Full Text | Google Scholar

Nigam S. K., Bush K. T. (2014). Growth factor-heparan sulfate “switches” regulating stages of branching morphogenesis. Pediatr. Nephrol. 29 (4), 727–735. doi:10.1007/s00467-013-2725-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Nur-E-Kamal A., Ahmed I., Kamal J., Schindler M., Meiners S. (2006). Three-dimensional nanofibrillar surfaces promote self-renewal in mouse embryonic stem cells. Stem Cells 24 (2), 426–433. doi:10.1634/stemcells.2005-0170

PubMed Abstract | CrossRef Full Text | Google Scholar

Otero J., Navajas D., Alcaraz J. (2020). Characterization of the elastic properties of extracellular matrix models by atomic force microscopy. Methods Cell Biol. 156, 59–83. doi:10.1016/bs.mcb.2019.11.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Passaniti A., Kleinman H. K., Martin G. R. (2021). Matrigel: History/background, uses, and future applications. J. Cell Commun. Signal. doi:10.1007/s12079-021-00643-1

CrossRef Full Text | Google Scholar

Petkau-Milroy K., Brunsveld L. (2013). Supramolecular chemical biology; bioactive synthetic self-assemblies. Org. Biomol. Chem. 11 (2), 219–232. doi:10.1039/c2ob26790j

PubMed Abstract | CrossRef Full Text | Google Scholar

Poornejad N., Schaumann L. B., Buckmiller E. M., Momtahan N., Gassman J. R., Ma H. H., et al. (2016). The impact of decellularization agents on renal tissue extracellular matrix. J. Biomater. Appl. 31 (4), 521–533. doi:10.1177/0885328216656099

PubMed Abstract | CrossRef Full Text | Google Scholar

Rayner S. G., Phong K. T., Xue J., Lih D., Shankland S. J., Kelly E. J., et al. (2018). Reconstructing the human renal vascular–tubular unit in vitro. Adv. Healthc. Mat. 7 (23), e1801120. doi:10.1002/adhm.201801120

PubMed Abstract | CrossRef Full Text | Google Scholar

Rosines E., Schmidt H. J., Nigam S. K. (2007). The effect of hyaluronic acid size and concentration on branching morphogenesis and tubule differentiation in developing kidney culture systems: Potential applications to engineering of renal tissues. Biomaterials 28 (32), 4806–4817. doi:10.1016/j.biomaterials.2007.07.034

PubMed Abstract | CrossRef Full Text | Google Scholar

Ruiter F. A. A., Morgan F. L. C., Roumans N., Schumacher A., Slaats G. G., Moroni L., et al. (2022). Soft, dynamic hydrogel confinement improves kidney organoid lumen morphology and reduces epithelial-mesenchymal transition in culture. Adv. Sci. 9 (20), e2200543. doi:10.1002/advs.202200543

PubMed Abstract | CrossRef Full Text | Google Scholar

Sakurai H., Barros E. J., Tsukamoto T., Barasch J., Nigam S. K. (1997). An in vitro tubulogenesis system using cell lines derived from the embryonic kidney shows dependence on multiple soluble growth factors. Proc. Natl. Acad. Sci. U. S. A. 94 (12), 6279–6284. doi:10.1073/pnas.94.12.6279

PubMed Abstract | CrossRef Full Text | Google Scholar

Satyam A., Tsokos M. G., Tresback J. S., Zeugolis D. I., Tsokos G. C. (2020). Cell derived extracellular matrix-rich biomimetic substrate supports podocyte proliferation, differentiation and maintenance of native phenotype. Adv. Funct. Mat. 30 (44), 1908752. doi:10.1002/adfm.201908752

PubMed Abstract | CrossRef Full Text | Google Scholar

Schlüter M. A., Margolis B. (2009). Apical lumen formation in renal epithelia. J. Am. Soc. Nephrol. 20 (7), 1444–1452. doi:10.1681/ASN.2008090949

PubMed Abstract | CrossRef Full Text | Google Scholar

Schophuizen C. M. S., De Napoli I. E., Jansen J., Teixeira S., Wilmer M. J., Hoenderop J. G. J., et al. (2015). Development of a living membrane comprising a functional human renal proximal tubule cell monolayer on polyethersulfone polymeric membrane. Acta Biomater. 14, 22–32. doi:10.1016/j.actbio.2014.12.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Schutgens F., Rookmaaker M. B., Margaritis T., Rios A., Ammerlaan C., Jansen J., et al. (2019). Tubuloids derived from human adult kidney and urine for personalized disease modeling. Nat. Biotechnol. 37 (3), 303–313. doi:10.1038/s41587-019-0048-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Sciancalepore A. G., Portone A., Moffa M., Persano L., De Luca M., Paiano A., et al. (2016). Micropatterning control of tubular commitment in human adult renal stem cells. Biomaterials 94, 57–69. doi:10.1016/j.biomaterials.2016.03.042

PubMed Abstract | CrossRef Full Text | Google Scholar

Sebinger D. D. R., Ofenbauer A., Gruber P., Malik S., Werner C. (2013). ECM modulated early kidney development in embryonic organ culture. Biomaterials 34 (28), 6670–6682. doi:10.1016/j.biomaterials.2013.05.031

PubMed Abstract | CrossRef Full Text | Google Scholar

Secker P. F., Luks L., Schlichenmaier N., Dietrich D. R. (2018). RPTEC/TERT1 cells form highly differentiated tubules when cultured in a 3D matrix. ALTEX 35 (2), 223–234. doi:10.14573/altex.1710181

PubMed Abstract | CrossRef Full Text | Google Scholar

Shahraki S., Bideskan A. E., Aslzare M., Tavakkoli M., Bahrami A. R., Hosseinian S., et al. (2022). Decellularization with triton X-100 provides a suitable model for human kidney bioengineering using human mesenchymal stem cells. Life Sci. 295, 120167. doi:10.1016/j.lfs.2021.120167

PubMed Abstract | CrossRef Full Text | Google Scholar

Shamir E. R., Ewald A. J. (2014). Three-dimensional organotypic culture: Experimental models of mammalian biology and disease. Nat. Rev. Mol. Cell Biol. 15 (10), 647–664. doi:10.1038/nrm3873

PubMed Abstract | CrossRef Full Text | Google Scholar

Shen C., Zhang G., Wang Q., Meng Q. (2015). Fabrication of collagen gel hollow fibers by covalent cross-linking for construction of bioengineering renal tubules. ACS Appl. Mat. Interfaces 7 (35), 19789–19797. doi:10.1021/acsami.5b05809

PubMed Abstract | CrossRef Full Text | Google Scholar

Shen L., Song X., Xu Y., Tian R., Wang Y., Li P., et al. (2021). Patterned vascularization in a directional ice-templated scaffold of decellularized matrix. Eng. Life Sci. 21 (10), 683–692. doi:10.1002/elsc.202100034

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh N. K., Han W., Nam S. A., Kim J. W., Kim J. Y., Kim Y. K., et al. (2020201911973). Three-dimensional cell-printing of advanced renal tubular tissue analogue. Biomaterials 232, 119734. doi:10.1016/j.biomaterials.2019.119734

PubMed Abstract | CrossRef Full Text | Google Scholar

Sobreiro-Almeida R., Fonseca D. R., Neves N. M. (2019). Extracellular matrix electrospun membranes for mimicking natural renal filtration barriers. Mat. Sci. Eng. C Mat. Biol. Appl. 103, 109866. doi:10.1016/j.msec.2019.109866

PubMed Abstract | CrossRef Full Text | Google Scholar

Sobreiro-Almeida R., Gómez-Florit M., Quinteira R., Reis R. L., Gomes M. E., Neves N. M. (2021). Decellularized kidney extracellular matrix bioinks recapitulate renal 3D microenvironment in vitro. Biofabrication 13 (4), 045006. doi:10.1088/1758-5090/ac0fca

PubMed Abstract | CrossRef Full Text | Google Scholar

Sobreiro-Almeida R., Melica M. E., Lasagni L., Romagnani P., Neves N. M. (2020). Co-cultures of renal progenitors and endothelial cells on kidney decellularized matrices replicate the renal tubular environment in vitro. Acta Physiol. 230 (1), e13491. doi:10.1111/apha.13491

PubMed Abstract | CrossRef Full Text | Google Scholar

Song J. J., Guyette J. P., Gilpin S. E., Gonzalez G., Vacanti J. P., Ott H. C. (2013). Regeneration and experimental orthotopic transplantation of a bioengineered kidney. Nat. Med. 19 (5), 646–651. doi:10.1038/nm.3154

PubMed Abstract | CrossRef Full Text | Google Scholar

Su J., Satchell S. C., Shah R. N., Wertheim J. A. (2018). Kidney decellularized extracellular matrix hydrogels: Rheological characterization and human glomerular endothelial cell response to encapsulation. J. Biomed. Mat. Res. A 106 (9), 2448–2462. doi:10.1002/jbm.a.36439

PubMed Abstract | CrossRef Full Text | Google Scholar

Subramanian B., Ko W. C., Yadav V., DesRochers T. M., Perrone R. D., Zhou J., et al. (2012). The regulation of cystogenesis in a tissue engineered kidney disease system by abnormal matrix interactions. Biomaterials 33 (33), 8383–8394. doi:10.1016/j.biomaterials.2012.08.020

PubMed Abstract | CrossRef Full Text | Google Scholar

Subramanian B., Rudym D., Cannizzaro C., Perrone R., Zhou J., Kaplan D. L. (2010). Tissue-engineered three-dimensional in vitro models for normal and diseased kidney. Tissue Eng. Part A 16 (9), 2821–2831. doi:10.1089/ten.tea.2009.0595

PubMed Abstract | CrossRef Full Text | Google Scholar

Sullivan D. C., Mirmalek-Sani S. H., Deegan D. B., Baptista P. M., Aboushwareb T., Atala A., et al. (2012). Decellularization methods of porcine kidneys for whole organ engineering using a high-throughput system. Biomaterials 33 (31), 7756–7764. doi:10.1016/j.biomaterials.2012.07.023

PubMed Abstract | CrossRef Full Text | Google Scholar

Taguchi A., Nishinakamura R. (2017). Higher-order kidney organogenesis from pluripotent stem cells. Cell Stem Cell 21 (6), 730–746. e6. doi:10.1016/j.stem.2017.10.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Takasato M., Er P. X., Chiu H. S., Maier B., Baillie G. J., Ferguson C., et al. (2015). Kidney organoids from human iPS cells contain multiple lineages and model human nephrogenesis. Nature 526 (7574), 564–568. doi:10.1038/nature15695

PubMed Abstract | CrossRef Full Text | Google Scholar

Takasato M., Little M. H. (2017). Making a kidney organoid using the directed differentiation of human pluripotent stem cells. Methods Mol. Biol. 1597, 195–206. doi:10.1007/978-1-4939-6949-4_14

PubMed Abstract | CrossRef Full Text | Google Scholar

Taub M., Wang Y., Szczesny T. M., Kleinman H. K. (1990). Epidermal growth factor or transforming growth factor α is required for kidney tubulogenesis in matrigel cultures in serum-free medium. Proc. Natl. Acad. Sci. U. S. A. 87 (10), 4002–4006. doi:10.1073/pnas.87.10.4002

PubMed Abstract | CrossRef Full Text | Google Scholar

Theocharis A. D., Skandalis S. S., Gialeli C., Karamanos N. K. (2016). Extracellular matrix structure. Adv. Drug Deliv. Rev. 97, 4–27. doi:10.1016/j.addr.2015.11.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Toyohara T., Mae S-I., Sueta S-I., Inoue T., Yamagishi Y., Kawamoto T., et al. (2015). Cell therapy using human induced pluripotent stem cell-derived renal progenitors ameliorates acute kidney injury in mice. Stem Cells Transl. Med. 4 (9), 980–992. doi:10.5966/sctm.2014-0219

PubMed Abstract | CrossRef Full Text | Google Scholar

Tröndle K., Miotto G., Rizzo L., Pichler R., Koch F., Koltay P., et al. (2022). Deep learning-assisted nephrotoxicity testing with bioprinted renal spheroids. Int. J. Bioprint. 8 (2), 528. doi:10.18063/ijb.v8i2.528

PubMed Abstract | CrossRef Full Text | Google Scholar

Tröndle K., Rizzo L., Pichler R., Koch F., Itani A., Zengerle R., et al. (2021). Scalable fabrication of renal spheroids and nephron-like tubules by bioprinting and controlled self-assembly of epithelial cells. Biofabrication 13 (3), 035019. doi:10.1088/1758-5090/abe185

PubMed Abstract | CrossRef Full Text | Google Scholar

Ullah I., Busch J. F., Rabien A., Ergün B., Stamm C., Knosalla C., et al. (2020). Adult tissue extracellular matrix determines tissue specification of human iPSC-derived embryonic stage mesodermal precursor cells. Adv. Sci. 7 (5), 1901198. doi:10.1002/advs.201901198

PubMed Abstract | CrossRef Full Text | Google Scholar

van den Berg C. W., Ritsma L., Avramut M. C., Wiersma L. E., van den Berg B. M., Leuning D. G., et al. (2018). Renal subcapsular transplantation of PSC-derived kidney organoids induces neo-vasculogenesis and significant glomerular and tubular maturation in vivo. Stem Cell Rep. 10 (3), 751–765. doi:10.1016/j.stemcr.2018.01.041

PubMed Abstract | CrossRef Full Text | Google Scholar

van Gaal R. C., Vrehen A. F., van Sprang J. F., Fransen P-P. K. H., van Turnhout M. C., Dankers P. Y. W. (2021). Biomaterial screening of protein coatings and peptide additives: Towards a simple synthetic mimic of a complex natural coating for a bio-artificial kidney. Biomater. Sci. 9 (6), 2209–2220. doi:10.1039/d0bm01930e

PubMed Abstract | CrossRef Full Text | Google Scholar

van Genderen A. M., Jansen K., Kristen M., van Duijn J., Li Y., Schuurmans C. C. L., et al. (2021). Topographic guidance in melt-electrowritten tubular scaffolds enhances engineered kidney tubule performance. Front. Bioeng. Biotechnol. 8, 617364. doi:10.3389/fbioe.2020.617364

PubMed Abstract | CrossRef Full Text | Google Scholar

Vormann M. K., Tool L. M., Ohbuchi M., Gijzen L., van Vught R., Hankemeier T., et al. (2022). Modelling and prevention of acute kidney injury through ischemia and reperfusion in a combined human renal proximal tubule/blood vessel-on-a-chip. Kidney360 3 (2), 217–231. doi:10.34067/KID.0003622021

PubMed Abstract | CrossRef Full Text | Google Scholar

Vormann M. K., Vriend J., Lanz H. L., Gijzen L., van den Heuvel A., Hutter S., et al. (2021). Implementation of a human renal proximal tubule on a chip for nephrotoxicity and drug interaction studies. J. Pharm. Sci. 110 (4), 1601–1614. doi:10.1016/j.xphs.2021.01.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Walma D. A. C., Yamada K. M. (2020). The extracellular matrix in development. Development 147 (10), dev175596–9. doi:10.1242/dev.175596

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang X., Guo C., Chen Y., Tozzi L., Szymkowiak S., Li C., et al. (2020). Developing a self-organized tubulogenesis model of human renal proximal tubular epithelial cells in vitro. J. Biomed. Mat. Res. A 108 (3), 795–804. doi:10.1002/jbm.a.36858

PubMed Abstract | CrossRef Full Text | Google Scholar

Weber H. M., Tsurkan M. V., Magno V., Freudenberg U., Werner C. (2017). Heparin-based hydrogels induce human renal tubulogenesis in vitro. Acta Biomater. 57, 59–69. doi:10.1016/j.actbio.2017.05.035

PubMed Abstract | CrossRef Full Text | Google Scholar

Wiraja C., Mori Y., Ichimura T., Hwang J., Xu C., Bonventre J. V. (2021). Nephrotoxicity assessment with human kidney tubuloids using spherical nucleic acid-based mRNA nanoflares. Nano Lett. 21 (13), 5850–5858. doi:10.1021/acs.nanolett.1c01840

PubMed Abstract | CrossRef Full Text | Google Scholar

Xia Y., Nivet E., Sancho-Martinez I., Gallegos T., Suzuki K., Okamura D., et al. (2013). Directed differentiation of human pluripotent cells to ureteric bud kidney progenitor-like cells. Nat. Cell Biol. 15 (12), 1507–1515. doi:10.1038/ncb2872

PubMed Abstract | CrossRef Full Text | Google Scholar

Xie R., Korolj A., Liu C., Song X., Lu R. X. Z., Zhang B., et al. (2020). h-FIBER: Microfluidic topographical hollow fiber for studies of glomerular filtration barrier. ACS Cent. Sci. 6 (6), 903–912. doi:10.1021/acscentsci.9b01097

PubMed Abstract | CrossRef Full Text | Google Scholar

Xing H., Lee H., Luo L., Kyriakides T. R. (2020). Extracellular matrix-derived biomaterials in engineering cell function. Biotechnol. Adv. 42, 107421. doi:10.1016/j.biotechadv.2019.107421

PubMed Abstract | CrossRef Full Text | Google Scholar

Zegers M. M. P., O’Brien L. E., Yu W., Datta A., Mostov K. E. (2003). Epithelial polarity and tubulogenesis in vitro. Trends Cell Biol. 13 (4), 169–176. doi:10.1016/s0962-8924(03)00036-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang H., Lau S. F-T., Heng B. F., Teo P. Y., Alahakoon P. K. D. T., Ni M., et al. (2011). Generation of easily accessible human kidney tubules on two-dimensional surfaces in vitro. J. Cell. Mol. Med. 15 (6), 1287–1298. doi:10.1111/j.1582-4934.2010.01113.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang H., Tasnim F., Ying J. Y., Zink D. (2009). The impact of extracellular matrix coatings on the performance of human renal cells applied in bioartificial kidneys. Biomaterials 30 (15), 2899–2911. doi:10.1016/j.biomaterials.2009.01.046

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang J., Li K., Kong F., Sun C., Zhang D., Yu X., et al. (2019). Induced intermediate mesoderm combined with decellularized kidney scaffolds for functional engineering kidney. Tissue Eng. Regen. Med. 16 (5), 501–512. doi:10.1007/s13770-019-00197-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang Y., Reif G., Wallace D. P. (2020). Extracellular matrix, integrins, and focal adhesion signaling in polycystic kidney disease. Cell. Signal. 72, 109646. doi:10.1016/j.cellsig.2020.109646

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou C., Zhou L., Liu J., Xu L., Xu Z., Chen Z., et al. (2020). Kidney extracellular matrix hydrogel enhances therapeutic potential of adipose-derived mesenchymal stem cells for renal ischemia reperfusion injury. Acta Biomater. 115, 250–263. doi:10.1016/j.actbio.2020.07.056

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: extracellular matrix, matrigel, kidney, bioprinting, scaffolds, microfluidics, organoids

Citation: Lacueva-Aparicio A, Lindoso RS, Mihăilă SM and Giménez I (2022) Role of extracellular matrix components and structure in new renal models in vitro. Front. Physiol. 13:1048738. doi: 10.3389/fphys.2022.1048738

Received: 19 September 2022; Accepted: 31 October 2022;
Published: 07 December 2022.

Edited by:

Nuria M. Pastor-Soler, University of Southern California, United States

Reviewed by:

Biagio Saitta, University of Southern California, United States
Mariela Mendez, Henry Ford Hospital, United States

Copyright © 2022 Lacueva-Aparicio, Lindoso, Mihăilă and Giménez. 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: Ignacio Giménez, igimenez@unizar.es

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.