DONATE

Staff member

Elena Garreta Bahima

Staff member publications

Pahuja, A, Corredera, IG, Moya-Rull, D, Garreta, E, Montserrat, N, (2024). Engineering physiological environments to advance kidney organoid models from human pluripotent stem cells Current Opinion In Cell Biology 86, 102306

During embryogenesis, the mammalian kidney arises because of reciprocal interactions between the ureteric bud (UB) and the metanephric mesenchyme (MM), driving UB branching and nephron induction. These morphogenetic processes involve a series of cellular rearrangements that are tightly controlled by gene regulatory networks and signaling cascades. Here, we discuss how kidney developmental studies have informed the definition of procedures to obtain kidney organoids from human pluripotent stem cells (hPSCs). Moreover, bioengineering techniques have emerged as potential solutions to externally impose controlled microenvironments for organoid generation from hPSCs. Next, we summarize some of these advances with major focus On recent works merging hPSC-derived kidney organoids (hPSC-kidney organoids) with organ-on-chip to develop robust models for drug discovery and disease modeling applications. We foresee that, in the near future, coupling of different organoid models through bioengineering approaches will help advancing to recreate organ-to-organ crosstalk to increase our understanding on kidney disease progression in the human context and search for new therapeutics.Copyright © 2023 The Authors. Published by Elsevier Ltd.. All rights reserved.

JTD Keywords: Animal, Animals, Bioengineering, Cell differentiation, Embryo development, Embryology, Embryonic structures, Gene regulatory network, Human, Humans, Kidney, Kidney development, Kidney mesenchyme cell, Kidney organoid, Mammal, Mammals, Mesenchyme, Metanephric mesenchyme, Microenvironment, Nephron, Nephrons, Organoid, Organoids, Physiology, Pluripotent stem cell, Pluripotent stem cells, Review, Signal transduction, Ureteric bud


Nauryzgaliyeva, Z, Corredera, IG, Garreta, E, Montserrat, N, (2023). Harnessing mechanobiology for kidney organoid research Frontiers In Cell And Developmental Biology 11, 1273923

Recently, organoids have emerged as revolutionizing tools with the unprecedented potential to recreate organ-specific microanatomy in vitro. Upon their derivation from human pluripotent stem cells (hPSCs), organoids reveal the blueprints of human organogenesis, further allowing the faithful recapitulation of their physiology. Nevertheless, along with the evolution of this field, advanced research exposed the organoids' shortcomings, particularly regarding poor reproducibility rates and overall immatureness. To resolve these challenges, many studies have started to underscore the relevance of mechanical cues as a relevant source to induce and externally control hPSCs differentiation. Indeed, established organoid generation protocols from hPSCs have mainly relyed on the biochemical induction of fundamental signalling pathways present during kidney formation in mammals, whereas mechanical cues have largely been unexplored. This review aims to discuss the pertinence of (bio) physical cues within hPSCs-derived organoid cultures, while deciphering their effect on morphogenesis. Moreover, we will explore state-of-the-art mechanobiology techniques as revolutionizing means for understanding the underlying role of mechanical forces in biological processes in organoid model systems.

JTD Keywords: development, hpscs, mechanobiology, nephrogenesis, Activated ion-channel, Development, Extracellular-matrix viscoelasticity, Forces, Hpscs, Ips cells, Mechanical regulation, Mechanobiology, Nephrogenesis, Nephron progenitors, Organoids, Pluripotent stem-cells, Self-renewal, Substrate mechanics, Tissue


Garreta, E, Moya-Rull, D, Stanifer, ML, Monteil, V, Prado, P, Marco, A, Tarantino, C, Gallo, M, Jonsson, G, Hagelkruys, A, Mirazimi, A, Boulant, S, Penninger, JM, Montserrat, N, (2022). Protocol for SARS-CoV-2 infection of kidney organoids derived from human pluripotent stem cells Star Protocols 3, 101872

This protocol presents the use of SARS-CoV-2 isolates to infect human kidney organoids, enabling exploration of the impact of SARS-CoV-2 infection in a human multicellular in vitro system. We detail steps to generate kidney organoids from human pluripotent stem cells (hPSCs) and emulate a diabetic milieu via organoids exposure to diabetogenic-like cell culture conditions. We further describe preparation and titration steps of SARS-CoV-2 virus stocks, their subsequent use to infect the kidney organoids, and assessment of the infection via immunofluorescence.

JTD Keywords: cell culture, cell differentiation, microbiology, microscopy, organoids, Cell culture, Microbiología, Microscopy, Stem cells


Safi, W, Marco, A, Moya, D, Prado, P, Garreta, E, Montserrat, N, (2022). Assessing kidney development and disease using kidney organoids and CRISPR engineering Frontiers In Cell And Developmental Biology 10, 948395

The differentiation of human pluripotent stem cells (hPSCs) towards organoids is one of the biggest scientific advances in regenerative medicine. Kidney organoids have not only laid the groundwork for various organ-like tissue systems but also provided insights into kidney embryonic development. Thus, several protocols for the differentiation of renal progenitors or mature cell types have been established. Insights into the interplay of developmental pathways in nephrogenesis and determination of different cell fates have enabled the in vitro recapitulation of nephrogenesis. Here we first provide an overview of kidney morphogenesis and patterning in the mouse model in order to dissect signalling pathways that are key to define culture conditions sustaining renal differentiation from hPSCs. Secondly, we also highlight how genome editing approaches have provided insights on the specific role of different genes and molecular pathways during renal differentiation from hPSCs. Based on this knowledge we further review how CRISPR/Cas9 technology has enabled the recapitulation and correction of cellular phenotypes associated with human renal disease. Last, we also revise how the field has positively benefited from emerging technologies as single cell RNA sequencing and discuss current limitations on kidney organoid technology that will take advantage from bioengineering solutions to help standardizing the use of this model systems to study kidney development and disease.Copyright © 2022 Safi, Marco, Moya, Prado, Garreta and Montserrat.

JTD Keywords: crispr, directed differentiation, epithelial-cells, expression, kidney engineering, kidney organoids, model, mouse, nephrogenesis, nephron number, podocytes, progenitor, Crispr, Kidney engineering, Kidney organoids, Nephrogenesis, Pluripotent stem cells, Pluripotent stem-cells


Garreta, E, Prado, P, Stanifer, ML, Monteil, V, Marco, A, Ullate-Agote, A, Moya-Rull, D, Vilas-Zornoza, A, Tarantino, C, Romero, JP, Jonsson, G, Oria, R, Leopoldi, A, Hagelkruys, A, Gallo, M, González, F, Domingo-Pedrol, P, Gavaldà, A, del Pozo, CH, Ali, OH, Ventura-Aguiar, P, Campistol, JM, Prosper, F, Mirazimi, A, Boulant, S, Penninger, JM, Montserrat, N, (2022). A diabetic milieu increases ACE2 expression and cellular susceptibility to SARS-CoV-2 infections in human kidney organoids and patient cells Cell Metabolism 34, 857-873

It is not well understood why diabetic individuals are more prone to develop severe COVID-19. To this, we here established a human kidney organoid model promoting early hallmarks of diabetic kidney disease development. Upon SARS-CoV-2 infection, diabetic-like kidney organoids exhibited higher viral loads compared with their control counterparts. Genetic deletion of the angiotensin-converting enzyme 2 (ACE2) in kidney organoids under control or diabetic-like conditions prevented viral detection. Moreover, cells isolated from kidney biopsies from diabetic patients exhibited altered mitochondrial respiration and enhanced glycolysis, resulting in higher SARS-CoV-2 infections compared with non-diabetic cells. Conversely, the exposure of patient cells to dichloroacetate (DCA), an inhibitor of aerobic glycolysis, resulted in reduced SARS-CoV-2 infections. Our results provide insights into the identification of diabetic-induced metabolic programming in the kidney as a critical event increasing SARS-CoV-2 infection susceptibility, opening the door to the identification of new interventions in COVID-19 pathogenesis targeting energy metabolism.Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.

JTD Keywords: complications, coronavirus, cultured-cells, disease, distal tubule, mouse, protein, reveals, spike, Ace2, Angiotensin-converting enzyme 2, Angiotensin-converting enzyme-2, Covid-19, Diabetes 2, Human kidney organoids, Sars-cov-2


Batlle, D, Monteil, V, Garreta, E, Hassler, L, Wysocki, J, Chandar, V, Schwartz, RE, Mirazimi, A, Montserrat, N, Bader, M, Penninger, JM, (2022). Evidence in favor of the essentiality of human cell membrane-bound ACE2 and against soluble ACE2 for SARS-CoV-2 infectivity Cell 185, 1837-1839

Garreta, E, Nauryzgaliyeva, Z, Marco, A, Safi, W, Montserrat, N, (2022). Dissecting nephron morphogenesis using kidney organoids from human pluripotent stem cells Current Opinion In Genetics & Development 72, 22-29

During kidney development the emergence of complex multicellular shapes such as the nephron (the functional unit of the kidney) rely on spatiotemporally coordinated developmental programs. These involve gene regulatory networks, signaling pathways and mechanical forces, that work in concert to shape and form the nephron(s). The generation of kidney organoids from human pluripotent stem cells now represent an unprecedented experimental set up to study these processes. Here we discuss the potential applications of kidney organoids to advance our knowledge of how mechanical forces and cell fate specification spatiotemporally interact to orchestrate nephron patterning and morphogenesis in humans. Progress in innovative techniques for quantifying and perturbing these processes in a controlled manner will be crucial. A mechanistic understanding of the multicellular dynamic processes occurring during nephrogenesis will pave the way to unveil new mechanisms of human kidney disease. © 2021

JTD Keywords: differentiation, dynamics, induction, lumen formation, models, mouse, organogenesis, reveals, tubules, Divergent features


Garreta, E, Nauryzgaliyeva, Z, Montserrat, N, (2021). Human induced pluripotent stem cell-derived kidney organoids toward clinical implementations Curr Opin Biomed Eng 20, 100346

The generation of kidney organoids from human pluripotent stem cells (hPSCs) has represented a relevant scientific achievement in the organoid field. Importantly, hPSC-derived kidney organoids contain multiple nephron-like structures that exhibit some renal functional characteristics and have the capacity to respond to nephrotoxic agents. In this review, we first discuss how bioengineering approaches can help overcome current kidney organoid challenges. Next, we focus on recent works exploiting kidney organoids for drug screening and disease modeling applications. Finally, we provide a state of the art on current research toward the potential application of kidney organoids and renal cells derived from hPSCs for future renal replacement therapies.

JTD Keywords: Bioengineering, Converting enzyme-ii, Crispr/cas9 gene editing, Disease, Disease modeling, Extracellular-matrix, Generation, Human pluripotent stem cells, Kidney organoids, Kidney regeneration, Model, Mouse, Reveals, Scaffold, Transplantation


Molins, B, Garreta, E, del Pozo, CH, Adan, A, Montserrat, N, (2021). Modelling retinal disease with a blood-retinal-barrier in vitro system combining human pluripotent stem cells and decellularized retinal tissue Investigative Ophthalmology & Visual Science 62, 2694

Purpose : Reductionist approaches into mechanisms underlying diseases of the outer blood-retinal-barrier (oBRB), such as age-related macular degeneration and diabetic retinopathy (DR) have been hampered by the lack of optimal in vitro models utilizing human cells to provide the 3-D dynamic architecture and allow expression of the in vivo phenotype for both the retinal pigment epithelialium (RPE) and the choroidal endothelium (EC). The main limitations of the current oBRB models also arise from the cell sourcing, the lack of a proper Bruch s membrane (BM) analogue, and lack of choroidal microvasculature with flow. Therefore, we aimed to develop an oBRB-on-a-chip biomimetic system to emulate the cellular interactions that occur in retinal inflammatory disorders. Methods : We have generated a macrofluidic device that allows the simultaneous co-culture of RPE with perfusable EC. Taking advantage of the differentiation potential of human pluripotent stem cells (hPSC), we optimized differentiation protocols to obtain hPSC-RPE and hPSC-EC from hPSC. On the other hand, by combining biomaterial engineering and decellularization protocols we designed a BM analogue that favors the co-culture of hPSC-RPE and hPSC-EC. Results : Differentiated hPSC-RPE showed a phenotype similar to that of mature RPE, while differentiated hPSC-EC showed a mature endothelial phenotype as they showed tubulogenesis properties and expressed endothelial markers. The co-culture of EC with hPSC-RPE cells increased the RPE barrier functional activity, significantly increasing TEER and decreasing the basolateral secretion of VEGF. On the other hand, we developed a decellularization protocol to obtain decellularized BM (dECM-BM) that guarantees DNA removal while preserving collagen and elastin fibers. Moreover, the BM analogue successfully allowed the co-culture of EC and hPSC-RPE cells Finally, we challenged the oBRB model with glucose oscillations and recapitulated the DR microenvironment. Conclusions : Our oBRB biomimetic co-culture system recapitulates the complex cellular interactions of the oBRB, inducing an increased RPE barrier functional activity, and allowing for the emulation of inflammatory microenvironments occurring during retinal disease. Overall, this human oBRB in vitro model represents an optimal platform to study the inflammatory processes underlying retinal pathologies. This is a 2021 ARVO Annual Meeting abstract.

JTD


Garreta, E, Kamm, RD, Lopes, SMCD, Lancaster, MA, Weiss, R, Trepat, X, Hyun, I, Montserrat, N, (2021). Rethinking organoid technology through bioengineering Nature Materials 20, 145-155

In recent years considerable progress has been made in the development of faithful procedures for the differentiation of human pluripotent stem cells (hPSCs). An important step in this direction has also been the derivation of organoids. This technology generally relies on traditional three-dimensional culture techniques that exploit cell-autonomous self-organization responses of hPSCs with minimal control over the external inputs supplied to the system. The convergence of stem cell biology and bioengineering offers the possibility to provide these stimuli in a controlled fashion, resulting in the development of naturally inspired approaches to overcome major limitations of this nascent technology. Based on the current developments, we emphasize the achievements and ongoing challenges of bringing together hPSC organoid differentiation, bioengineering and ethics. This Review underlines the need for providing engineering solutions to gain control of self-organization and functionality of hPSC-derived organoids. We expect that this knowledge will guide the community to generate higher-grade hPSC-derived organoids for further applications in developmental biology, drug screening, disease modelling and personalized medicine. This Review provides an overview of bioengineering technologies that can be harnessed to facilitate the culture, self-organization and functionality of human pluripotent stem cell-derived organoids.

JTD Keywords: Differentiation, Embryonic-tissues, Extracellular-matrix, In-vitro, Kidney organoids, Model, Neural-tube, Pluripotent stem-cells, Reconstitution, Self-organization


Selfa, IL, Gallo, M, Montserrat, N, Garreta, E, (2021). Directed Differentiation of Human Pluripotent Stem Cells for the Generation of High-Order Kidney Organoids Methods In Molecular Biology 2258, 171-192

© 2021, The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature. Our understanding in the inherent properties of human pluripotent stem cells (hPSCs) have made possible the development of differentiation procedures to generate three-dimensional tissue-like cultures, so-called organoids. Here we detail a stepwise methodology to generate kidney organoids from hPSCs. This is achieved through direct differentiation of hPSCs in two-dimensional monolayer culture toward the posterior primitive streak fate, followed by induction of intermediate mesoderm-committed cells, which are further aggregated and cultured in three-dimensions to generate kidney organoids containing segmented nephron-like structures in a process that lasts 20 days. We also provide a concise description on how to assess renal commitment during the time course of kidney organoid generation. This includes the use of flow cytometry and immunocytochemistry analyses for the detection of specific renal differentiation markers.

JTD Keywords: 2d monolayer, 3d organotypic culture, differentiation, flow cytometry, human pluripotent stem cells, immunocytochemistry, intermediate mesoderm, kidney organoid, nephron progenitor cells, nephrons, primitive streak, 2d monolayer, 3d organotypic culture, Differentiation, Flow cytometry, Human pluripotent stem cells, Immunocytochemistry, Intermediate mesoderm, Kidney organoid, Nephron progenitor cells, Nephrons, Primitive streak, Tissue


Kyndiah, A., Leonardi, F., Tarantino, C., Cramer, T., Millan-Solsona, R., Garreta, E., Montserrat, N., Mas-Torrent, M., Gomila, G., (2020). Bioelectronic recordings of cardiomyocytes with accumulation mode electrolyte gated organic field effect transistors Biosensors and Bioelectronics 150, 111844

Organic electronic materials offer an untapped potential for novel tools for low-invasive electrophysiological recording and stimulation devices. Such materials combine semiconducting properties with tailored surface chemistry, elastic mechanical properties and chemical stability in water. In this work, we investigate solution processed Electrolyte Gated Organic Field Effect Transistors (EGOFETs) based on a small molecule semiconductor. We demonstrate that EGOFETs based on a blend of soluble organic semiconductor 2,8-Difluoro-5,11-bis(triethylsilylethynyl)anthradithiophene (diF-TES-ADT) combined with an insulating polymer show excellent sensitivity and long-term recording under electrophysiological applications. Our devices can stably record the extracellular potential of human pluripotent stem cell derived cardiomyocyte cells (hPSCs-CMs) for several weeks. In addition, cytotoxicity tests of pharmaceutical drugs, such as Norepinephrine and Verapamil was achieved with excellent sensitivity. This work demonstrates that organic transistors based on organic blends are excellent bioelectronics transducer for extracellular electrical recording of excitable cells and tissues thus providing a valid alternative to electrochemical transistors.

JTD Keywords: Bioelectronics, Cardiac cells, Organic electronics, Organic field effect transistors, Organic semiconducting blend


Lynch, Cian J., Bernad, Raquel, Martínez-Val, Ana, Shahbazi, Marta N., Nóbrega-Pereira, Sandrina, Calvo, Isabel, Blanco-Aparicio, Carmen, Tarantino, Carolina, Garreta, Elena, Richart-Ginés, Laia, Alcazar, Noelia, Graña-Castro, Osvaldo, Gómez-Lopez, Gonzalo, Aksoy, Irene, Muñoz-Martín, Maribel, Martinez, Sonia, Ortega, Sagrario, Prieto, Susana, Simboeck, Elisabeth, Camasses, Alain, Stephan-Otto Attolini, Camille, Fernandez, Agustin F., Sierra, Marta I., Fraga, Mario F., Pastor, Joaquin, Fisher, Daniel, Montserrat, Nuria, Savatier, Pierre, Muñoz, Javier, Zernicka-Goetz, Magdalena, Serrano, Manuel, (2020). Global hyperactivation of enhancers stabilizes human and mouse naive pluripotency through inhibition of CDK8/19 Mediator kinases Nature Cell Biology 22, (10), 1223-1238

Pluripotent stem cells (PSCs) transition between cell states in vitro, reflecting developmental changes in the early embryo. PSCs can be stabilized in the naive state by blocking extracellular differentiation stimuli, particularly FGF–MEK signalling. Here, we report that multiple features of the naive state in human and mouse PSCs can be recapitulated without affecting FGF–MEK signalling or global DNA methylation. Mechanistically, chemical inhibition of CDK8 and CDK19 (hereafter CDK8/19) kinases removes their ability to repress the Mediator complex at enhancers. CDK8/19 inhibition therefore increases Mediator-driven recruitment of RNA polymerase II (RNA Pol II) to promoters and enhancers. This efficiently stabilizes the naive transcriptional program and confers resistance to enhancer perturbation by BRD4 inhibition. Moreover, naive pluripotency during embryonic development coincides with a reduction in CDK8/19. We conclude that global hyperactivation of enhancers drives naive pluripotency, and this can be achieved in vitro by inhibiting CDK8/19 kinase activity. These principles may apply to other contexts of cellular plasticity.

JTD


Monteil, Vanessa, Kwon, Hyesoo, Prado, Patricia, Hagelkrüys, Astrid, Wimmer, Reiner A., Stahl, Martin, Leopoldi, Alexandra, Garreta, Elena, Hurtado Del Pozo, Carmen, Prosper, Felipe, Romero, Juan Pablo, Wirnsberger, Gerald, Zhang, Haibo, Slutsky, Arthur S., Conder, Ryan, Montserrat, Nuria, Mirazimi, Ali, Penninger, Josef M., (2020). Inhibition of SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2 Cell 181, (4), 905-913.e7

We have previously provided the first genetic evidence that angiotensin converting enzyme 2 (ACE2) is the critical receptor for severe acute respiratory syndrome coronavirus (SARS-CoV), and ACE2 protects the lung from injury, providing a molecular explanation for the severe lung failure and death due to SARS-CoV infections. ACE2 has now also been identified as a key receptor for SARS-CoV-2 infections, and it has been proposed that inhibiting this interaction might be used in treating patients with COVID-19. However, it is not known whether human recombinant soluble ACE2 (hrsACE2) blocks growth of SARS-CoV-2. Here, we show that clinical grade hrsACE2 reduced SARS-CoV-2 recovery from Vero cells by a factor of 1,000-5,000. An equivalent mouse rsACE2 had no effect. We also show that SARS-CoV-2 can directly infect engineered human blood vessel organoids and human kidney organoids, which can be inhibited by hrsACE2. These data demonstrate that hrsACE2 can significantly block early stages of SARS-CoV-2 infections.

JTD Keywords: COVID-19, Angiotensin converting enzyme 2, Blood vessels, Human organoids, Kidney, Severe acute respiratory syndrome coronavirus, Spike glycoproteins, Treatment


Garreta, Elena, Prado, Patricia, Tarantino, Carolina, Oria, Roger, Fanlo, Lucia, Martí, Elisa, Zalvidea, Dobryna, Trepat, Xavier, Roca-Cusachs, Pere, Gavaldà -Navarro, Aleix, Cozzuto, Luca, Campistol, Josep M., Izpisúa Belmonte, Juan Carlos, Hurtado del Pozo, Carmen, Montserrat, Nuria, (2019). Fine tuning the extracellular environment accelerates the derivation of kidney organoids from human pluripotent stem cells Nature Materials 18, 397-405

The generation of organoids is one of the biggest scientific advances in regenerative medicine. Here, by lengthening the time that human pluripotent stem cells (hPSCs) were exposed to a three-dimensional microenvironment, and by applying defined renal inductive signals, we generated kidney organoids that transcriptomically matched second-trimester human fetal kidneys. We validated these results using ex vivo and in vitro assays that model renal development. Furthermore, we developed a transplantation method that utilizes the chick chorioallantoic membrane. This approach created a soft in vivo microenvironment that promoted the growth and differentiation of implanted kidney organoids, as well as providing a vascular component. The stiffness of the in ovo chorioallantoic membrane microenvironment was recapitulated in vitro by fabricating compliant hydrogels. These biomaterials promoted the efficient generation of renal vesicles and nephron structures, demonstrating that a soft environment accelerates the differentiation of hPSC-derived kidney organoids.

JTD


Garreta, Elena, Montserrat, Nuria, Belmonte, Juan Carlos Izpisua, (2018). Kidney organoids for disease modeling Oncotarget 9, (16), 12552-12553

Garreta, Elena, Sanchez, Sonia, Lajara, Jeronimo, Montserrat, Nuria, Belmonte, Juan Carlos Izpisua, (2018). Roadblocks in the path of iPSC to the vlinic Current Transplantation Reports 5, (1), 14-18

PURPOSE OF REVIEW: The goal of this paper is to highlight the major challenges in the translation of human pluripotent stem cells into a clinical setting. RECENT FINDINGS: Innate features from human induced pluripotent stem cells (hiPSCs) positioned these patient-specific cells as an unprecedented cell source for regenerative medicine applications. Immunogenicity of differentiated iPSCs requires more research towards the definition of common criteria for the evaluation of innate and host immune responses as well as in the generation of standardized protocols for iPSC generation and differentiation. The coming years will resolve ongoing clinical trials using both human embryonic stem cells (hESCs) and hiPSCs providing exciting information for the optimization of potential clinical applications of stem cell therapies. SUMMARY: Rapid advances in the field of iPSCs generated high expectations in the field of regenerative medicine. Understanding therapeutic applications of iPSCs certainly needs further investigation on autologous/allogenic iPSC transplantation.

JTD


Latorre, Ernest, Kale, Sohan, Casares, Laura, Gómez-González, Manuel, Uroz, Marina, Valon, Léo, Nair, Roshna V., Garreta, Elena, Montserrat, Nuria, del Campo, Aránzazu, Ladoux, Benoit, Arroyo, Marino, Trepat, Xavier, (2018). Active superelasticity in three-dimensional epithelia of controlled shape Nature 563, (7730), 203-208

Fundamental biological processes are carried out by curved epithelial sheets that enclose a pressurized lumen. How these sheets develop and withstand three-dimensional deformations has remained unclear. Here we combine measurements of epithelial tension and shape with theoretical modelling to show that epithelial sheets are active superelastic materials. We produce arrays of epithelial domes with controlled geometry. Quantification of luminal pressure and epithelial tension reveals a tensional plateau over several-fold areal strains. These extreme strains in the tissue are accommodated by highly heterogeneous strains at a cellular level, in seeming contradiction to the measured tensional uniformity. This phenomenon is reminiscent of superelasticity, a behaviour that is generally attributed to microscopic material instabilities in metal alloys. We show that in epithelial cells this instability is triggered by a stretch-induced dilution of the actin cortex, and is rescued by the intermediate filament network. Our study reveals a type of mechanical behaviour—which we term active superelasticity—that enables epithelial sheets to sustain extreme stretching under constant tension.

JTD


Hurtado del Pozo, Carmen, Garreta, Elena, Izpisúa Belmonte, Juan Carlos, Montserrat, Nuria, (2018). Modeling epigenetic modifications in renal development and disease with organoids and genome editing Disease Models & Mechanisms 11, (11), 035048

Understanding epigenetic mechanisms is crucial to our comprehension of gene regulation in development and disease. In the past decades, different studies have shown the role of epigenetic modifications and modifiers in renal disease, especially during its progression towards chronic and end-stage renal disease. Thus, the identification of genetic variation associated with chronic kidney disease has resulted in better clinical management of patients. Despite the importance of these findings, the translation of genotype–phenotype data into gene-based medicine in chronic kidney disease populations still lacks faithful cellular or animal models that recapitulate the key aspects of the human kidney. The latest advances in the field of stem cells have shown that it is possible to emulate kidney development and function with organoids derived from human pluripotent stem cells. These have successfully recapitulated not only kidney differentiation, but also the specific phenotypical traits related to kidney function. The combination of this methodology with CRISPR/Cas9 genome editing has already helped researchers to model different genetic kidney disorders. Nowadays, CRISPR/Cas9-based approaches also allow epigenetic modifications, and thus represent an unprecedented tool for the screening of genetic variants, epigenetic modifications or even changes in chromatin structure that are altered in renal disease. In this Review, we discuss these technical advances in kidney modeling, and offer an overview of the role of epigenetic regulation in kidney development and disease.

JTD


Garreta, E., González, F., Montserrat, N., (2018). Studying kidney disease using tissue and genome engineering in human pluripotent stem cells Nephron 138, 48-59

Kidney morphogenesis and patterning have been extensively studied in animal models such as the mouse and zebrafish. These seminal studies have been key to define the molecular mechanisms underlying this complex multistep process. Based on this knowledge, the last 3 years have witnessed the development of a cohort of protocols allowing efficient differentiation of human pluripotent stem cells (hPSCs) towards defined kidney progenitor populations using two-dimensional (2D) culture systems or through generating organoids. Kidney organoids are three-dimensional (3D) kidney-like tissues, which are able to partially recapitulate kidney structure and function in vitro. The current possibility to combine state-of-the art tissue engineering with clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated systems 9 (Cas9)-mediated genome engineering provides an unprecedented opportunity for studying kidney disease with hPSCs. Recently, hPSCs with genetic mutations introduced through CRISPR/Cas9-mediated genome engineering have shown to produce kidney organoids able to recapitulate phenotypes of polycystic kidney disease and glomerulopathies. This mini review provides an overview of the most recent advances in differentiation of hPSCs into kidney lineages, and the latest implementation of the CRISPR/Cas9 technology in the organoid setting, as promising platforms to study human kidney development and disease.

JTD Keywords: Clustered regularly interspaced short palindromic repeats/CRISPR-associated systems 9, Disease modeling, Gene editing, Human pluripotent stem cells, Kidney genetics, Tissue engineering


Garreta, Elena, Oria, Roger, Tarantino, Carolina, Pla-Roca, Mateu, Prado, Patricia, Fernández-Avilés, Francisco, Campistol, Josep Maria, Samitier, Josep, Montserrat, Nuria, (2017). Tissue engineering by decellularization and 3D bioprinting Materials Today , 20, (4), 166-178

Discarded human donor organs have been shown to provide decellularized extracellular matrix (dECM) scaffolds suitable for organ engineering. The quest for appropriate cell sources to satisfy the need of multiple cells types in order to fully repopulate human organ-derived dECM scaffolds has opened new venues for the use of human pluripotent stem cells (hPSCs) for recellularization. In addition, three-dimensional (3D) bioprinting techniques are advancing towards the fabrication of biomimetic cell-laden biomaterial constructs. Here, we review recent progress in decellularization/recellularization and 3D bioprinting technologies, aiming to fabricate autologous tissue grafts and organs with an impact in regenerative medicine.

JTD


Garreta, Elena, Marco, Andrés, Eguizábal, Cristina, Tarantino, Carolina, Samitier, Mireia, Badiola, Maider, Gutiérrez, Joaquín, Samitier, Josep, Montserrat, Nuria, (2017). Pluripotent stem cells and skeletal muscle differentiation: Challenges and immediate applications The Plasticity of Skeletal Muscle: From Molecular Mechanism to Clinical Applications (ed. Sakuma, Kunihiro), Springer Singapore (Singapore, Singapore) 2018, 1-35

Recent advances in the generation of skeletal muscle derivatives from pluripotent stem cells (PSCs) provide innovative tools for muscle development, disease modeling, and cell replacement therapies. Here, we revise major relevant findings that have contributed to these advances in the field, by the revision of how early findings using mouse embryonic stem cells (ESCs) set the bases for the derivation of skeletal muscle cells from human pluripotent stem cells (hPSCs) and patient-derived human-induced pluripotent stem cells (hiPSCs) to the use of genome editing platforms allowing for disease modeling in the petri dish.

JTD Keywords: Pluripotent stem cells, Differentiation, Genome editing, Disease modeling


Garreta, Elena, Marco, Andres, Izpisua Belmonte, Juan Carlos, Montserrat, Nuria, (2016). Genome editing in human pluripotent stem cells: a systematic approach unrevealing pancreas development and disease Stem Cell Investigation , 4, (11), 1-4

Although mouse models have represented a major tool for understanding and predicting molecular mechanisms responsible for several human genetic diseases, still species-specific differences between mouse and humans in their biochemical and physiological characteristics represent a major hurdle when translating promising findings into the human setting (1). For instance, in several types of maturity onset diabetes of the young (MODY; autosomal dominant), mice with heterozygous mutations do not develop diabetes (2). In this regard, the derivation of human embryonic stem cells (hESCs) in 1998 represented an unprecedented opportunity for human disease modelling, and a promising source for cell replacement therapies (3). Later on, the possibility to generate patient-derived induced pluripotent stem cells (iPSCs) has opened new venues for the potential translation of stem-cell related studies into the clinic (4).

JTD


Garreta, E., de Oñate, L., Fernández-Santos, M. E., Oria, R., Tarantino, C., Climent, A. M., Marco, A., Samitier, M., Martínez, Elena, Valls-Margarit, M., Matesanz, R., Taylor, D. A., Fernández-Avilés, F., Izpisua Belmonte, J. C., Montserrat, N., (2016). Myocardial commitment from human pluripotent stem cells: Rapid production of human heart grafts Biomaterials 98, 64-78

Genome editing on human pluripotent stem cells (hPSCs) together with the development of protocols for organ decellularization opens the door to the generation of autologous bioartificial hearts. Here we sought to generate for the first time a fluorescent reporter human embryonic stem cell (hESC) line by means of Transcription activator-like effector nucleases (TALENs) to efficiently produce cardiomyocyte-like cells (CLCs) from hPSCs and repopulate decellularized human heart ventricles for heart engineering. In our hands, targeting myosin heavy chain locus (MYH6) with mCherry fluorescent reporter by TALEN technology in hESCs did not alter major pluripotent-related features, and allowed for the definition of a robust protocol for CLCs production also from human induced pluripotent stem cells (hiPSCs) in 14 days. hPSCs-derived CLCs (hPSCs-CLCs) were next used to recellularize acellular cardiac scaffolds. Electrophysiological responses encountered when hPSCs-CLCs were cultured on ventricular decellularized extracellular matrix (vdECM) correlated with significant increases in the levels of expression of different ion channels determinant for calcium homeostasis and heart contractile function. Overall, the approach described here allows for the rapid generation of human cardiac grafts from hPSCs, in a total of 24 days, providing a suitable platform for cardiac engineering and disease modeling in the human setting.

JTD Keywords: Cardiac function, Extracellular matrix, Gene targeting, Pluripotent stem cells


Montserrat, N., Garreta, E., Izpisua Belmonte, J. C., (2016). Regenerative strategies for kidney engineering FEBS Journal , 283, (18), 3303-3324

The kidney is the most important organ for water homeostasis and waste excretion. It performs several important physiological functions for homeostasis: it filters the metabolic waste out of circulation, regulates body fluid balances, and acts as an immune regulator and modulator of cardiovascular physiology. The development of in vitro renal disease models with pluripotent stem cells (both human embryonic stem cells and induced pluripotent stem cells) and the generation of robust protocols for in vitro derivation of renal-specific-like cells from patient induced pluripotent stem cells have just emerged. Here we review major findings in the field of kidney regeneration with a major focus on the development of stepwise protocols for kidney cell production from human pluripotent stem cells and the latest advances in kidney bioengineering (i.e. decellularized kidney scaffolds and bioprinting). The possibility of generating renal-like three-dimensional structures to be recellularized with renal-derived induced pluripotent stem cells may offer new avenues to develop functional kidney grafts on-demand.

JTD Keywords: Induced pluripotent stem cells, Kidney disease, Kidney engineering, Pluripotent stem cells, Renal differentiation


de Oñate, L., Garreta, E., Tarantino, C., Martínez, Elena, Capilla, E., Navarro, I., Gutiérrez, J., Samitier, J., Campistol, J.M., Muñoz-Cánovas, P., Montserrat, N., (2015). Research on skeletal muscle diseases using pluripotent stem cells Muscle Cell and Tissue (ed. Sakuma, K.), InTech (Rijeka, Croatia) , 333-357

The generation of induced pluripotent stem cells (iPSCs), especially the generation of patient-derived pluripotent stem cells (PSCs) suitable for disease modelling in vitro, opens the door for the potential translation of stem-cell related studies into the clinic. Successful replacement, or augmentation, of the function of damaged cells by patient-derived differentiated stem cells would provide a novel cell-based therapy for skeletal muscle-related diseases. Since iPSCs resemble human embryonic stem cells (hESCs) in their ability to generate cells of the three germ layers, patient-specific iPSCs offer definitive solutions for the ethical and histo-incompatibility issues related to hESCs. Indeed human iPSC (hiPSC)-based autologous transplantation is heralded as the future of regenerative medicine. Interestingly, during the last years intense research has been published on disease-specific hiPSCs derivation and differentiation into relevant tissues/organs providing a unique scenario for modelling disease progression, to screen patient-specific drugs and enabling immunosupression-free cell replacement therapies. Here, we revise the most relevant findings in skeletal muscle differentiation using mouse and human PSCs. Finally and in an effort to bring iPSC technology to the daily routine of the laboratory, we provide two different protocols for the generation of patient-derived iPSCs.

JTD Keywords: Pluripotent stem cells, Myogenic differentiation, Disease modelling, Patient-specific induced pluripotent stem cells, Muscular dystrophy


Garreta, Elena, Melo, Esther, Navajas, Daniel, Farré, Ramon, (2014). Low oxygen tension enhances the generation of lung progenitor cells from mouse embryonic and induced pluripotent stem cells Physiological Reports , 2, (7), e12075

Abstract Whole-organ decellularization technology has emerged as a new alternative for the fabrication of bioartificial lungs. Embryonic stem cells (ESC) and induced pluripotent stem cells (iPSC) are potentially useful for recellularization since they can be directed to express phenotypic marker genes of lung epithelial cells. Normal pulmonary development takes place in a low oxygen environment ranging from 1 to 5%. By contrast, in vitro ESC and iPSC differentiation protocols are usually carried out at room-air oxygen tension. Here, we sought to determine the role played by oxygen tension on the derivation of Nkx2.1+ lung/thyroid progenitor cells from mouse ESC and iPSC. A step-wise differentiation protocol was used to generate Nkx2.1+ lung/thyroid progenitors under 20% and 5% oxygen tension. On day 12, gene expression analysis revealed that Nkx2.1 and Foxa2 (endodermal and early lung epithelial cell marker) were significantly upregulated at 5% oxygen tension in ESC and iPSC differentiated cultures compared to 20% oxygen conditions. In addition, quantification of Foxa2+Nkx2.1+Pax8- cells corresponding to the lung field, with exclusion of the potential thyroid fate identified by Pax8 expression, confirmed that the low physiologic oxygen tension exerted a significant positive effect on early pulmonary differentiation of ESC and iPSC. In conclusion, we found that 5% oxygen tension enhanced the derivation of lung progenitors from mouse ESC and iPSC compared to 20% room-air oxygen tension.

JTD


Melo, E., Garreta, E., Luque, T., Cortiella, J., Nichols, J., Navajas, D., Farré, R., (2014). Effects of the decellularization method on the local stiffness of acellular lungs Tissue Engineering Part C: Methods , 20, (5), 412-422

Lung bioengineering, a novel approach to obtain organs potentially available for transplantation, is based on decellularizing donor lungs and seeding natural scaffolds with stem cells. Various physicochemical protocols have been used to decellularize lungs, and their performance has been evaluated in terms of efficient decellularization and matrix preservation. No data are available, however, on the effect of different decellularization procedures on the local stiffness of the acellular lung. This information is important since stem cells directly sense the rigidity of the local site they are engrafting to during recellularization, and it has been shown that substrate stiffness modulates cell fate into different phenotypes. The aim of this study was to assess the effects of the decellularization procedure on the inhomogeneous local stiffness of the acellular lung on five different sites: alveolar septa, alveolar junctions, pleura, and vessels' tunica intima and tunica adventitia. Local matrix stiffness was measured by computing Young's modulus with atomic force microscopy after decellularizing the lungs of 36 healthy rats (Sprague-Dawley, male, 250–300 g) with four different protocols with/without perfusion through the lung circulatory system and using two different detergents (sodium dodecyl sulfate [SDS] and 3-[(3-cholamidopropyl) dimethylammonio]-1-propanesulfonate [CHAPS]). The local stiffness of the acellular lung matrix significantly depended on the site within the matrix (p<0.001), ranging from ∼15 kPa at the alveolar septum to ∼60 kPa at the tunica intima. Acellular lung stiffness (p=0.003) depended significantly, albeit modestly, on the decellularization process. Whereas perfusion did not induce any significant differences in stiffness, the use of CHAPS resulted in a ∼35% reduction compared with SDS, the influence of the detergent being more important in the tunica intima. In conclusion, lung matrix stiffness is considerably inhomogeneous, and conventional decellularization procedures do not result in substantially different local stiffness in the acellular lung.

JTD


Melo, E., Cárdenes, N., Garreta, E., Luque, T., Rojas, M., Navajas, D., Farré, R., (2014). Inhomogeneity of local stiffness in the extracellular matrix scaffold of fibrotic mouse lungs Journal of the Mechanical Behavior of Biomedical Materials , 37, 186-195

Lung disease models are useful to study how cell engraftment, proliferation and differentiation are modulated in lung bioengineering. The aim of this work was to characterize the local stiffness of decellularized lungs in aged and fibrotic mice. Mice (2- and 24-month old; 14 of each) with lung fibrosis (N=20) and healthy controls (N=8) were euthanized after 11 days of intratracheal bleomycin (fibrosis) or saline (controls) infusion. The lungs were excised, decellularized by a conventional detergent-based (sodium-dodecyl sulfate) procedure and slices of the acellular lungs were prepared to measure the local stiffness by means of atomic force microscopy. The local stiffness of the different sites in acellular fibrotic lungs was very inhomogeneous within the lung and increased according to the degree of the structural fibrotic lesion. Local stiffness of the acellular lungs did not show statistically significant differences caused by age. The group of mice most affected by fibrosis exhibited local stiffness that were ~2-fold higher than in the control mice: from 27.2±1.64 to 64.8±7.1. kPa in the alveolar septa, from 56.6±4.6 to 99.9±11.7. kPa in the visceral pleura, from 41.1±8.0 to 105.2±13.6. kPa in the tunica adventitia, and from 79.3±7.2 to 146.6±28.8. kPa in the tunica intima. Since acellular lungs from mice with bleomycin-induced fibrosis present considerable micromechanical inhomogeneity, this model can be a useful tool to better investigate how different degrees of extracellular matrix lesion modulate cell fate in the process of organ bioengineering from decellularized lungs.

JTD Keywords: Ageing, Atomic force microscopy, Decellularization, Lung fibrosis, Tissue engineering, Atomic force microscopy, Biological organs, Peptides, Sodium dodecyl sulfate, Sodium sulfate, Tissue engineering, Ageing, Decellularization, Extracellular matrices, Healthy controls, Inhomogeneities, Lung fibrosis, Micro-mechanical, Statistically significant difference, Mammals, bleomycin, adventitia, animal experiment, animal model, article, atomic force microscopy, bleomycin-induced pulmonary fibrosis, cell fate, controlled study, extracellular matrix, female, intima, lung alveolus, lung fibrosis, lung mechanics, mechanical probe, microenvironment, mouse, nonhuman, pleura, priority journal, rigidity, tissue engineering


Nonaka, P. N., Campillo, N., Uriarte, J. J., Garreta, E., Melo, E., de Oliveira, L. V. F., Navajas, D., Farré, R., (2014). Effects of freezing/thawing on the mechanical properties of decellularized lungs Journal of Biomedical Materials Research - Part A , 102, (2), 413-419

Lung bioengineering based on decellularized organ scaffolds is a potential alternative for transplantation. Freezing/thawing, a usual procedure in organ decellularization and storage could modify the mechanical properties of the lung scaffold and reduce the performance of the bioengineered lung when subjected to the physiological inflation-deflation breathing cycles. The aim of this study was to determine the effects of repeated freezing/thawing on the mechanical properties of decellularized lungs in the physiological pressure-volume regime associated with normal ventilation. Fifteen mice lungs (C57BL/6) were decellularized using a conventional protocol not involving organ freezing and based on sodium dodecyl sulfate detergent. Subsequently, the mechanical properties of the acellular lungs were measured before and after subjecting them to three consecutive cycles of freezing/thawing. The resistance (RL) and elastance (EL) of the decellularized lungs were computed by linear regression fitting of the recorded signals (tracheal pressure, flow, and volume) during mechanical ventilation. RL was not significantly modified by freezing-thawing: from 0.88 ± 0.37 to 0.90 ± 0.38 cmH2O·s·mL-1 (mean ± SE). EL slightly increased from 64.4 ± 11.1 to 73.0 ± 16.3 cmH2O·mL-1 after the three freeze-thaw cycles (p = 0.0013). In conclusion, the freezing/thawing process that is commonly used for both organ decellularization and storage induces only minor changes in the ventilation mechanical properties of the organ scaffold.

JTD Keywords: Elastance, Freezing/thawing, Lung bioengineering, Lung decellularization, Mechanical ventilation, Organ scaffold


Luque, T., Melo, E., Garreta, E., Cortiella, J., Nichols, J., Farré, R., Navajas, D., (2013). Local micromechanical properties of decellularized lung scaffolds measured with atomic force microscopy Acta Biomaterialia 9, (6), 6852-6859

Bioartificial lungs re-engineered from decellularized organ scaffolds are a promising alternative to lung transplantation. Critical features for improving scaffold repopulation depend on the mechanical properties of the cell microenvironment. However, the mechanics of the lung extracellular matrix (ECM) is poorly defined. The local mechanical properties of the ECM were measured in different regions of decellularized rat lung scaffolds with atomic force microscopy. Lungs excised from rats (n = 11) were decellularized with sodium dodecyl sulfate (SDS) and cut into ∼7 μm thick slices. The complex elastic modulus (G*) of lung ECM was measured over a frequency band ranging from 0.1 to 11.45 Hz. Measurements were taken in alveolar wall segments, alveolar wall junctions and pleural regions. The storage modulus (G′, real part of G*) of alveolar ECM was ∼6 kPa, showing small changes between wall segments and junctions. Pleural regions were threefold stiffer than alveolar walls. G′ of alveolar walls and pleura increased with frequency as a weak power law with exponent 0.05. The loss modulus (G″, imaginary part of G*) was 10-fold lower and showed a frequency dependence similar to that of G′ at low frequencies (0.1-1 Hz), but increased more markedly at higher frequencies. Local differences in mechanical properties and topology of the parenchymal site could be relevant mechanical cues for regulating the spatial distribution, differentiation and function of lung cells.

JTD


Tsapikouni, T., Garreta, E., Melo, E., Navajas, D., Farré, R., (2012). A bioreactor for subjecting cultured cells to fast-rate intermittent hypoxia Respiratory Physiology & Neurobiology , 182, (1), 47-52

High frequency intermittent hypoxia is one of the most relevant injurious stimuli experienced by patients with obstructive sleep apnea (OSA). Given that the conventional setting for culturing cells under intermittent hypoxia conditions is limited by long equilibration times, we designed a simple bioreactor capable of effectively subjecting cultured cells to controlled high-frequency hypoxic/normoxic stimuli. The bioreactor's operation is based on exposing cells to a medium that is bubbled with the appropriate mixture of gases into two separate containers, and from there it is directed to the cell culture dish with the aid of two bidirectional peristaltic pumps. The device was tested on human alveolar epithelial cells (A549) and mouse melanoma cells (B16-F10), subjecting them to patterns of intermittent hypoxia (20s at 5% O 2 and 50s at 20% O 2), which realistically mimic OSA of up to severe intensity as defined by the apnea hypopnea index. The proposed bioreactor can be easily and inexpensively assembled and is of practical use for investigating the effects of high-rate changes in oxygen concentration in the cell culture medium.

JTD Keywords: Hypoxia-reoxygenation, Obstructive sleep apnea, Oxygen partial pressure