DONATE

Staff member

Núria Montserrat Pulido

Group Leader / ICREA Research Professor
+34 934 031 391
nmontserratibecbarcelona.eu
CV Summary
Dra. Nuria Montserrat became interested in organ regeneration and stem cells during her master and PhD training that finished in 2006. The same year she got a Postdoctoral fellowship from the Fundaçao para a Ciência e Tecnología (Portugal). In 2007 she moved as a post-doctoral researcher at the Hospital of Santa Creu i Sant Pau in Barcelona.

In 2008 she joined the Center of Regenerative Medicine of Barcelona (CMRB) thanks to the support of a Juan de la Cierva fellowship under the direction of Dr. Izpisúa Belmonte. In 2010 she first co-authored how to reprogram cord blood stem cells for the first time (Nature Protocols, 2010). Then she first-coauthor the first work deriving iPSCs with new factors (Cell Stem Cell, 2013). She also collaborated in projects aimed to characterize the genomic integrity of human iPSCs as well as their differentiation towards different lineages for applications in human disease modeling (Stem Cells, 2011; Nature, 2012; Nature, 2012, Nature Communications, 2014). She has first co-authored how the reactivation of endogenous pathways can be artificially reactivated and promote heart regeneration in mammals (Cell Stem Cell, 2014).

Her expertise in the fields of somatic reprogramming and organ regeneration was recognized with an awarded an ERC Starting Grant by 2014 which allowed her to become Junior group leader at the Institute of Bioengineering of Catalonia (IBEC).

In January 2015 she got a Ramon y Cajal fellowship (first ranked candidate, 100/100 points) and since 2019 she is an ICREA Research Professor and Senior Group Leader at IBEC. During these years her findings in the field of Regenerative Medicine led to the derivation, for the first time, of cardiac grafts from human pluripotent stem cells and decellularized cardiac myocardium (Biomaterials, 2016), and the derivation of renal analogues with 3D bioprinting (Materials Today, 2017) and vascularized kidney organoids from hPSCs (Nature Materials, 2019).

She has recently co-led on the application of kidney organoid technology to model SARS-CoV-2 infections (Cell, 2020) identifying a therapeutic compound that nowadays is under clinical trial in COVID19 patients (The Lancet Respiratory Medicine, 2020; EMBO Molecular Medicine, 2020). Their Cell paper has been highlighted as a Research Highlight in both Nature Reviews Nephrology and in the Nature journal, also attracting remarkable attention (more than 200 national press releases, and 30 radio/television) being awarded as the “Best biomedical research publication 2020” (Constantes y Vitales Prize).
With the aim to further understand on the central role of metabolism in renal fate and damage protection she has recently co-led the first work on the identification of metabolic regulators protecting the renal tubule from acute injury exploiting kidney organoid technology (Cell Metabolism, 2020). In sum, from 2015, her work has attracted almost 8 M€ of direct competitive funding and around 1.2 M€ in personnel related grants, from both Spanish and European institutions.

All these efforts have been recently awarded with the prestigious EMBO Young Investigator Prize. In December 2020 the ERC has recognized all these efforts and she have been awarded with the prestigious ERC Consolidator Grant to study the interplay between mechanobiology and metabolism during human kidney development and disease.

Her commitment to scientific dissemination and communication has allowed her to participate in more than 300 outreach activities to promote our research activities to the general public, with a particular focus to young girls and woman in science. For this reason, she was selected as Commissioner of the first City and Science Biennial of Barcelona in 2019 being re-elected for the second edition (June, 2021).
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


Liu, M, Zhang, C, Gong, XM, Zhang, T, Lian, MM, Chew, EGY, Cardilla, A, Suzuki, K, Wang, HM, Yuan, Y, Li, Y, Naik, MY, Wang, YX, Zhou, BR, Soon, WZ, Aizawa, E, Li, P, Low, JH, Tandiono, M, Montagud, E, Moya-Rull, D, Esteban, CR, Luque, Y, Fang, ML, Khor, CC, Montserrat, N, Campistol, JM, Belmonte, JCI, Foo, JN, Xia, Y, (2024). Kidney organoid models reveal cilium-autophagy metabolic axis as a therapeutic target for PKD both in vitro and in vivo Cell Stem Cell 31, 52-70.e8

Human pluripotent stem cell -derived kidney organoids offer unprecedented opportunities for studying polycystic kidney disease (PKD), which still has no effective cure. Here, we developed both in vitro and in vivo organoid models of PKD that manifested tubular injury and aberrant upregulation of renin-angiotensin aldosterone system. Single -cell analysis revealed that a myriad of metabolic changes occurred during cystogenesis, including defective autophagy. Experimental activation of autophagy via ATG5 overexpression or primary cilia ablation significantly inhibited cystogenesis in PKD kidney organoids. Employing the organoid xenograft model of PKD, which spontaneously developed tubular cysts, we demonstrate that minoxidil, a potent autophagy activator and an FDA -approved drug, effectively attenuated cyst formation in vivo. This in vivo organoid model of PKD will enhance our capability to discover novel disease mechanisms and validate candidate drugs for clinical translation.

JTD Keywords: Adenylate kinase, Adult, Animal cell, Animal experiment, Animal model, Animal tissue, Article, Autophagosome, Autophagy, Autophagy (cellular), Calcium homeostasis, Cilia, Cilium, Cohort analysis, Controlled study, Cyclic amp, Dominant polycystic kidney,pluripotent stem-cells,autosomal-dominant,disease,expression,mutations,growth,generation,epithelium,framewor, Enzyme linked immunosorbent assay, Exon, Female, Food and drug administration, Hepatitis a virus cellular receptor 1, Human, Human cell, Humans, Immunohistochemistry, In vitro study, In vivo study, Kidney, Kidney organoid, Kidney polycystic disease, Male, Minoxidil, Mouse, Nonhuman, Organoid, Organoids, Platelet derived growth factor beta receptor, Polycystic kidney diseases, Protein kinase lkb1, Renin, Sequestosome 1, Single cell analysis, Single cell rna seq, Small nuclear rna, Tunel assay, Upregulation, Western blotting, Whole exome sequencing


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


Iglesias-García, O, Flandes-Iparraguirre, M, Montero, M, Larequi, E, Van Mil, A, Castilho, M, Fernández-Santos, ME, Sánchez, A, Montserrat, N, Fernández-Avilés, F, Sluijter, JPG, Malda, J, Mazo, M, Prósper, F, (2023). Development of an advanced tissue-engineering system through novel 3D printing fabrication methods (52354521444) Tissue Engineering Part a 29, 439-440

Sánchez-Guijo, F, Avendaño-Solá, C, Badimón, L, Bueren, JA, Canals, JM, Delgadillo, J, Delgado, J, Eguizábal, C, Fernández-Santos, ME, García-Olmo, D, González-Aseguinolaza, G, Juan, M, Martín, F, Mata, R, Montserrat, N, Pérez-Martínez, A, Pérez-Simón, JA, Prósper, F, Urbano-Ispizua, A, Zapata, AG, Sureda, A, Moraleda, JM, (2023). Role of Hospital Exemption in Europe: position paper from the Spanish Advanced Therapy Network (TERAV) Bone Marrow Transplantation 58, 727-728

Fontcuberta-PiSunyer, M, García-Alamán, A, Prades, É, Téllez, N, Alves-Figueiredo, H, Ramos-Rodríguez, M, Enrich, C, Fernandez-Ruiz, R, Cervantes, S, Clua, L, Ramón-Azcón, J, Broca, C, Wojtusciszyn, A, Montserrat, N, Pasquali, L, Novials, A, Servitja, JM, Vidal, J, Gomis, R, Gasa, R, (2023). Direct reprogramming of human fibroblasts into insulin-producing cells using transcription factors Commun Biol 6, 256

Direct lineage reprogramming of one somatic cell into another without transitioning through a progenitor stage has emerged as a strategy to generate clinically relevant cell types. One cell type of interest is the pancreatic insulin-producing β cell whose loss and/or dysfunction leads to diabetes. To date it has been possible to create β-like cells from related endodermal cell types by forcing the expression of developmental transcription factors, but not from more distant cell lineages like fibroblasts. In light of the therapeutic benefits of choosing an accessible cell type as the cell of origin, in this study we set out to analyze the feasibility of transforming human skin fibroblasts into β-like cells. We describe how the timed-introduction of five developmental transcription factors (Neurog3, Pdx1, MafA, Pax4, and Nkx2-2) promotes conversion of fibroblasts toward a β-cell fate. Reprogrammed cells exhibit β-cell features including β-cell gene expression and glucose-responsive intracellular calcium mobilization. Moreover, reprogrammed cells display glucose-induced insulin secretion in vitro and in vivo. This work provides proof-of-concept of the capacity to make insulin-producing cells from human fibroblasts via transcription factor-mediated direct reprogramming.© 2023. The Author(s).

JTD Keywords: adult, beta-cells, differentiation, direct conversion, genes, in-vivo, islets, maturation, pancreatic progenitors, Pluripotent stem-cells


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


Cillo, U, Weissenbacher, A, Pengel, L, Jochmans, I, Roppolo, D, Amarelli, C, Belli, LS, Berenguer, M, De Vries, A, Ferrer, J, Friedewald, J, Furian, L, Greenwood, S, Monbaliu, D, Nadalin, S, Neyrinck, A, Strazzabosco, M, Toso, C, Zaza, G, Thuraisingham, R, Berney, T, Potena, L, Montserrat, N, Selzner, N, (2022). ESOT Consensus Platform for Organ Transplantation: Setting the Stage for a Rigorous, Regularly Updated Development Process Transplant International 35, 10915

The European Society for Organ Transplantation (ESOT) has created a platform for the development of rigorous and regularly updated evidence based guidelines for clinical practice in the transplantation field. A dedicated Guideline Taskforce, including ESOT-council members, a representative from the Centre for Evidence in Transplantation, editors of the journal Transplant International has developed transparent procedures to guide the development of guidelines, recommendations, and consensus statements. During ESOT's first Consensus Conference in November 2022, leading experts will present in-depth evidence based reviews of nine themes and will propose recommendations aimed at reaching a consensus after public discussion and assessment by an independent jury. All recommendations and consensus statements produced for the nine selected topics will be published including the entire evidence-based consensus-finding process. An extensive literature review of each topic was conducted to provide final evidence and/or expert opinion.Copyright © 2022 Cillo, Weissenbacher, Pengel, Jochmans, Roppolo, Amarelli, Belli, Berenguer, De Vries, Ferrer, Friedewald, Furian, Greenwood, Monbaliu, Nadalin, Neyrinck, Strazzabosco, Toso, Zaza, Thuraisingham, Berney, Potena, Montserrat and Selzner.

JTD Keywords: consensus conference, guidelines, methodology, platform, Consensus conference, Guidelines, Methodology, Organ transplantation, Platform, Quality


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


Monteil, V, Eaton, B, Postnikova, E, Murphy, M, Braunsfeld, B, Crozier, I, Kricek, F, Niederhofer, J, Schwarzbock, A, Breid, H, Devignot, S, Klingstrom, J, Thalin, C, Kellner, MJ, Christ, W, Havervall, S, Mereiter, S, Knapp, S, Jimenez, AS, Bugajska-Schretter, A, Dohnal, A, Ruf, C, Gugenberger, R, Hagelkruys, A, Montserrat, N, Kozieradzki, I, Ali, OH, Stadlmann, J, Holbrook, MR, Schmaljohn, C, Oostenbrink, C, Shoemaker, RH, Mirazimi, A, Wirnsberger, G, Penninger, JM, (2022). Clinical grade ACE2 as a universal agent to block SARS-CoV-2 variants Embo Molecular Medicine 14, e15230

The recent emergence of multiple SARS-CoV-2 variants has caused considerable concern due to both reduced vaccine efficacy and escape from neutralizing antibody therapeutics. It is, therefore, paramount to develop therapeutic strategies that inhibit all known and future SARS-CoV-2 variants. Here, we report that all SARS-CoV-2 variants analyzed, including variants of concern (VOC) Alpha, Beta, Gamma, Delta, and Omicron, exhibit enhanced binding affinity to clinical grade and phase 2 tested recombinant human soluble ACE2 (APN01). Importantly, soluble ACE2 neutralized infection of VeroE6 cells and human lung epithelial cells by all current VOC strains with markedly enhanced potency when compared to reference SARS-CoV-2 isolates. Effective inhibition of infections with SARS-CoV-2 variants was validated and confirmed in two independent laboratories. These data show that SARS-CoV-2 variants that have emerged around the world, including current VOC and several variants of interest, can be inhibited by soluble ACE2, providing proof of principle of a pan-SARS-CoV-2 therapeutic.

JTD Keywords: clinical trial, covid-19, treatment, Clinical trial, Converting enzyme 2, Covid-19, Spike, Treatment, Vaccine


Astro, V, Ramirez-Calderon, G, Pennucci, R, Caroli, J, Saera-Vila, A, Cardona-Londono, K, Forastieri, C, Fiacco, E, Maksoud, F, Alowaysi, M, Sogne, E, Falqui, A, Gonzalez, F, Montserrat, N, Battaglioli, E, Mattevi, A, Adamo, A, (2022). Fine-tuned KDM1A alternative splicing regulates human cardiomyogenesis through an enzymatic-independent mechanism Iscience 25, 104665

The histone demethylase KDM1A is a multi- faceted regulator of vital developmental processes, including mesodermal and cardiac tube formation during gastrulation. However, it is unknown whether the fine-tuning of KDM1A splicing isoforms, already shown to regulate neuronal maturation, is crucial for the specification and maintenance of cell identity during cardiogenesis. Here, we discovered a temporal modulation of ubKDM1A and KDM1A+2a during human and mice fetal cardiac development and evaluated their impact on the regulation of cardiac differentiation. We revealed a severely impaired cardiac differentiation in KDM1A(-/-) hESCs that can be rescued by re-expressing ubKDM1A or catalytically impaired ubKDM1A-K661A, but not by KDM1A+2a or KDM1A+2a-K661A. Conversely, KDM1A+2a(-/-) hESCs give rise to functional cardiac cells, displaying increased beating amplitude and frequency and enhanced expression of critical cardiogenic markers. Our findings prove the existence of a divergent scaffolding role of KDM1A splice variants, independent of their enzymatic activity, during hESC differentiation into cardiac cells.

JTD Keywords: cell biology, molecular mechanism of gene regulation, omics, Bhlh transcription factor, Corest, Differentiation, Dna, Embryonic stem-cells, Heart, Lsd1, Phosphorylation, Proteins, Stem cells research, Swirm domain


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

Aydin, O, Passaro, AP, Raman, R, Spellicy, SE, Weinberg, RP, Kamm, RD, Sample, M, Truskey, GA, Zartman, J, Dar, RD, Palacios, S, Wang, J, Tordoff, J, Montserrat, N, Bashir, R, Saif, MTA, Weiss, R, (2022). Principles for the design of multicellular engineered living systems Apl Bioengineering 6, 10903

Remarkable progress in bioengineering over the past two decades has enabled the formulation of fundamental design principles for a variety of medical and non-medical applications. These advancements have laid the foundation for building multicellular engineered living systems (M-CELS) from biological parts, forming functional modules integrated into living machines. These cognizant design principles for living systems encompass novel genetic circuit manipulation, self-assembly, cell–cell/matrix communication, and artificial tissues/organs enabled through systems biology, bioinformatics, computational biology, genetic engineering, and microfluidics. Here, we introduce design principles and a blueprint for forward production of robust and standardized M-CELS, which may undergo variable reiterations through the classic design-build-test-debug cycle. This Review provides practical and theoretical frameworks to forward-design, control, and optimize novel M-CELS. Potential applications include biopharmaceuticals, bioreactor factories, biofuels, environmental bioremediation, cellular computing, biohybrid digital technology, and experimental investigations into mechanisms of multicellular organisms normally hidden inside the “black box” of living cells.

JTD Keywords: cell-fate specification, endothelial-cells, escherichia-coli, extracellular-matrix, gene-expression noise, nuclear hormone-receptors, pluripotent stem-cells, primitive endoderm, transcription factors, Artificial tissues, Assembly cells, Biological parts, Biological systems, Bioremediation, Blood-brain-barrier, Cell engineering, Cell/matrix communication, Design principles, Environmental technology, Functional modules, Fundamental design, Genetic circuits, Genetic engineering, Living machines, Living systems, Medical applications, Molecular biology, Synthetic biology


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


Gawish, R, Starkl, P, Pimenov, L, Hladik, A, Lakovits, K, Oberndorfer, F, Cronin, SJF, Ohradanova-Repic, A, Wirnsberger, G, Agerer, B, Endler, L, Capraz, T, Perthold, JW, Cikes, D, Koglgruber, R, Hagelkruys, A, Montserrat, N, Mirazimi, A, Boon, L, Stockinger, H, Bergthaler, A, Oostenbrink, C, Penninger, JM, Knapp, S, (2022). ACE2 is the critical in vivo receptor for SARS-CoV-2 in a novel COVID-19 mouse model with TNF-and IFNy-driven immunopathology Elife 11, e74623

Despite tremendous progress in the understanding of COVID-19, mechanistic insight into immunological, disease-driving factors remains limited. We generated maVie16, a mouse-adapted SARS-CoV-2, by serial passaging of a human isolate. In silico modeling revealed how only three Spike mutations of maVie16 enhanced interaction with murine ACE2. maVie16 induced profound pathology in BALB/c and C57BL/6 mice, and the resulting mouse COVID-19 (mCOVID-19) replicated critical aspects of human disease, including early lymphopenia, pulmonary immune cell infiltration, pneumonia, and specific adaptive immunity. Inhibition of the proinflammatory cyto-kines IFN? and TNF substantially reduced immunopathology. Importantly, genetic ACE2-deficiency completely prevented mCOVID-19 development. Finally, inhalation therapy with recombinant ACE2 fully protected mice from mCOVID-19, revealing a novel and efficient treatment. Thus, we here present maVie16 as a new tool to model COVID-19 for the discovery of new therapies and show that disease severity is determined by cytokine-driven immunopathology and critically dependent on ACE2 in vivo. © Gawish et al.

JTD Keywords: covid-19 mouse model, covid-19 therapy, cytokine storm, immunology, inflammation, mavie16, mouse, mouse-adapted sars-cov-2, program, recombinant soluble ace2, tmprss2, Adaptive immunity, Angiotensin converting enzyme 2, Angiotensin-converting enzyme 2, Animal, Animal cell, Animal experiment, Animal model, Animal tissue, Animals, Apoptosis, Article, Bagg albino mouse, Breathing rate, Bronchoalveolar lavage fluid, C57bl mouse, Cell composition, Cell infiltration, Controlled study, Coronavirus disease 2019, Coronavirus spike glycoprotein, Covid-19, Cytokeratin 18, Cytokine production, Dipeptidyl carboxypeptidase, Disease model, Disease models, animal, Disease severity, Drosophila-melanogaster, Enzyme linked immunosorbent assay, Expression vector, Flow cytometry, Gamma interferon, Gene editing, Gene expression, Gene mutation, Genetic engineering, Genetics, Glycosylation, High mobility group b1 protein, Histology, Histopathology, Immune response, Immunocompetent cell, Immunology, Immunopathology, Interferon-gamma, Interleukin 2, Metabolism, Mice, inbred balb c, Mice, inbred c57bl, Mouse-adapted sars-cov-2, Myeloperoxidase, Neuropilin 1, Nonhuman, Nucleocapsid protein, Pathogenicity, Peptidyl-dipeptidase a, Pyroptosis, Recombinant soluble ace2, Renin angiotensin aldosterone system, Rna extraction, Rna isolation, Sars-cov-2, Severe acute respiratory syndrome coronavirus 2, Spike glycoprotein, coronavirus, T lymphocyte activation, Trabecular meshwork, Tumor necrosis factor, Virology, Virus load, Virus replication, Virus transmission, Virus virulence


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


Pilat, N, Lefsihane, K, Brouard, S, Kotsch, K, Falk, C, Steiner, R, Thaunat, O, Fusil, F, Montserrat, N, Amarelli, C, Casiraghi, F, (2021). T- and B-cell therapy in solid organ transplantation: current evidence and future expectations Transplant International 34, 1594-1606

Cell therapy has emerged as an attractive therapeutic option in organ transplantation. During the last decade, the therapeutic potency of Treg immunotherapy has been shown in various preclinical animal models and safety was demonstrated in first clinical trials. However, there are still critical open questions regarding specificity, survival, and migration to the target tissue so the best Treg population for infusion into patients is still under debate. Recent advances in CAR technology hold the promise for Treg-functional superiority. Another exciting strategy is the generation of B-cell antibody receptor (BAR) Treg/cytotoxic T cells to specifically regulate or deplete alloreactive memory B cells. Finally, B cells are also capable of immune regulation, making them promising candidates for immunomodulatory therapeutic strategies. This article summarizes available literature on cell-based innovative therapeutic approaches aiming at modulating alloimmune response for transplantation. Crucial areas of investigation that need a joined effort of the transplant community for moving the field toward successful achievement of tolerance are highlighted.

JTD Keywords: allograft, autoimmune, b-cell antibody receptor t cells, chimeric antigen receptor tregs, expansion, expression, identification, infectious tolerance, mouse, prevention, regulatory b cells, regulatory t cells, signature, B-cell antibody receptor t cells, Chimeric antigen receptor tregs, Kidney-transplantation, Regulatory b cells, Regulatory t cells


Berishvili, E, Casiraghi, F, Amarelli, C, Scholz, H, Piemonti, L, Berney, T, Montserrat, N, (2021). Mini-organs forum: how to advance organoid technology to organ transplant community Transplant International 34, 1588-1593

The generation of human mini-organs, the so-called organoids, is one of the biggest scientific advances in regenerative medicine. This technology exploits traditional three-dimensional culture techniques that support cell-autonomous self-organization responses of stem cells to derive micrometer to millimeter size versions of human organs. The convergence of the organoid technology with organ transplantation is still in its infancy but this alliance is expected to open new venues to change the way we conduct both transplant and organoid research. In this Forum we provide a summary on early achievements facilitating organoid derivation and culture. We further discuss on early advances of organoid transplantation also offering a comprehensive overview of current limitations and challenges to instruct organoid maturation. We expect that this Forum sets the ground for initial discussions between stem cell biologists, bioengineers, and the transplant community to better direct organoid basic research to advance the organ transplantation field.

JTD Keywords: in-vitro, matrix, mice, organoids, regenerative medicine, vivo, Intestinal stem-cell, Organoids, Regenerative medicine


Calistri, A, Luganini, A, Mognetti, B, Elder, E, Sibille, G, Conciatori, V, Del Vecchio, C, Sainas, S, Boschi, D, Montserrat, N, Mirazimi, A, Lolli, ML, Gribaudo, G, Parolin, C, (2021). The new generation hdhodh inhibitor meds433 hinders the in vitro replication of sars-cov-2 and other human coronaviruses Microorganisms 9, 1731

Although coronaviruses (CoVs) have long been predicted to cause zoonotic diseases and pandemics with high probability, the lack of effective anti-pan-CoVs drugs rapidly usable against the emerging SARS-CoV-2 actually prevented a promptly therapeutic intervention for COVID-19. Development of host-targeting antivirals could be an alternative strategy for the control of emerging CoVs infections, as they could be quickly repositioned from one pandemic event to another. To contribute to these pandemic preparedness efforts, here we report on the broad-spectrum CoVs antiviral activity of MEDS433, a new inhibitor of the human dihydroorotate dehydrogenase (hDHODH), a key cellular enzyme of the de novo pyrimidine biosynthesis pathway. MEDS433 in-hibited the in vitro replication of hCoV-OC43 and hCoV-229E, as well as of SARS-CoV-2, at low nanomolar range. Notably, the anti-SARS-CoV-2 activity of MEDS433 against SARS-CoV-2 was also observed in kidney organoids generated from human embryonic stem cells. Then, the antiviral activity of MEDS433 was reversed by the addition of exogenous uridine or the product of hDHODH, the orotate, thus confirming hDHODH as the specific target of MEDS433 in hCoVs-infected cells. Taken together, these findings suggest MEDS433 as a potential candidate to develop novel drugs for COVID-19, as well as broad-spectrum antiviral agents exploitable for future CoVs threats.

JTD Keywords: antiviral activity, biosynthesis, broad-spectrum antiviral, combination treatment, coronavirus, dipyridamole, hdhodh inhibitor, organoids, pyrimidine, pyrimidine biosynthesis, sars-cov-2, target, virus-infection, Antiviral activ-ity, Broad-spectrum antiviral, Combination treatment, Coronavirus, Gene-expression, Hdhodh inhibitor, Organoids, Pyrimidine biosynthesis, Sars-cov-2


Lovell-Badge, R, Anthony, E, Barker, RA, Bubela, T, Brivanlou, AH, Carpenter, M, Charo, RA, Clark, A, Clayton, E, Cong, YL, Daley, GQ, Fu, JP, Fujita, M, Greenfield, A, Goldman, SA, Hill, L, Hyun, I, Isasi, R, Kahn, J, Kato, K, Kim, JS, Kimmelman, J, Knoblich, JA, Mathews, D, Montserrat, N, Mosher, J, Munsie, M, Nakauchi, H, Naldini, L, Naughton, G, Niakan, K, Ogbogu, U, Pedersen, R, Rivron, N, Rooke, H, Rossant, J, Round, J, Saitou, M, Sipp, D, Steffann, J, Sugarman, J, Surani, A, Takahashi, J, Tang, FC, Turner, L, Zettler, PJ, Zhai, XM, (2021). ISSCR Guidelines for Stem Cell Research and Clinical Translation: The 2021 update Stem Cell Reports 16, 1398-1408

The International Society for Stem Cell Research has updated its Guidelines for Stem Cell Research and Clinical Translation in order to address advances in stem cell science and other relevant fields, together with the associated ethical, social, and policy issues that have arisen since the last update in 2016. While growing to encompass the evolving science, clinical applications of stem cells, and the increasingly complex implications of stem cell research for society, the basic principles underlying the Guidelines remain unchanged, and they will continue to serve as the standard for the field and as a resource for scientists, regulators, funders, physicians, and members of the public, including patients. A summary of the key updates and issues is presented here.

JTD Keywords: self-organization, Human embryo research


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


Dhillon, P, Park, J, del Pozo, CH, Li, LZ, Doke, T, Huang, SZ, Zhao, JJ, Kang, HM, Shrestra, R, Balzer, MS, Chatterjee, S, Prado, P, Han, SY, Liu, HB, Sheng, X, Dierickx, P, Batmanov, K, Romero, JP, Prósper, F, Li, MY, Pei, LM, Kim, J, Montserrat, N, Susztak, K, (2021). The Nuclear Receptor ESRRA Protects from Kidney Disease by Coupling Metabolism and Differentiation Cell Metabolism 33, 379-394.e8

© 2020 Elsevier Inc. Using single-cell RNA sequencing, Susztak and colleagues, show major changes in cell diversity in mouse models of kidney fibrosis. Proximal tubule (PT) cells are highly vulnerable to dysfunction in fibrosis and show altered differentiation. Nuclear receptors such as ESRRA maintain both PT cell metabolism and differentiation by directly regulating PT-cell-specific genes.

JTD Keywords: chronic kidney disease, esrra, fatty-acid oxidation, fibrosis, kidney, organoids, ppara, proximal tubule cells, single-cell atac sequencing, Chronic kidney disease, Esrra, Fatty-acid oxidation, Fibrosis, Kidney, Organoids, Ppara, Proximal tubule cells, Single-cell atac sequencing, Single-cell rna sequencing


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


Monteil, V, Dyczynski, M, Lauschke, VM, Kwon, H, Wirnsberger, G, Youhanna, S, Zhang, HB, Slutsky, AS, del Pozo, CH, Horn, M, Montserrat, N, Penninger, JM, Mirazimi, A, (2021). Human soluble ACE2 improves the effect of remdesivir in SARS-CoV-2 infection Embo Molecular Medicine 13, e13426

© 2020 The Authors. Published under the terms of the CC BY 4.0 license There is a critical need for safe and effective drugs for COVID-19. Only remdesivir has received authorization for COVID-19 and has been shown to improve outcomes but not decrease mortality. However, the dose of remdesivir is limited by hepatic and kidney toxicity. ACE2 is the critical cell surface receptor for SARS-CoV-2. Here, we investigated additive effect of combination therapy using remdesivir with recombinant soluble ACE2 (high/low dose) on Vero E6 and kidney organoids, targeting two different modalities of SARS-CoV-2 life cycle: cell entry via its receptor ACE2 and intracellular viral RNA replication. This combination treatment markedly improved their therapeutic windows against SARS-CoV-2 in both models. By using single amino-acid resolution screening in haploid ES cells, we report a singular critical pathway required for remdesivir toxicity, namely, Adenylate Kinase 2. The data provided here demonstrate that combining two therapeutic modalities with different targets, common strategy in HIV treatment, exhibit strong additive effects at sub-toxic concentrations. Our data lay the groundwork for the study of combinatorial regimens in future COVID-19 clinical trials.

JTD Keywords: clinical trial, combination therapy, covid-19, Clinical trial, Combination therapy, Covid-19, Treatment


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


Hoogduijn, M.J., Montserrat, N., van der Laan, L.J.W., Dazzi, F., Perico, N., Kastrup, J., Gilbo, N., Ploeg, R.J., Roobrouck, V., Casiraghi, F., Johnson, C.L., Franquesa, M., Dahlke, M.H., Massey, E., Hosgood, S., Reinders, M.E.J., (2020). The emergence of regenerative medicine in organ transplantation: 1st European Cell Therapy and Organ Regeneration Section meeting Transplant International 33, (8), 833-840

Regenerative medicine is emerging as a novel field in organ transplantation. In September 2019, the European Cell Therapy and Organ Regeneration Section (ECTORS) of the European Society for Organ Transplantation (ESOT) held its first meeting to discuss the state-of-the-art of regenerative medicine in organ transplantation. The present article highlights the key areas of interest and major advances in this multidisciplinary field in organ regeneration and discusses its implications for the future of organ transplantation.

JTD Keywords: Cell therapy, Machine perfusion, Mesenchymal stromal cell, Organoid, Regeneration, Transplantation


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


Zoufaly, Alexander, Poglitsch, Marko, Aberle, Judith H., Hoepler, Wolfgang, Seitz, Tamara, Traugott, Marianna, Grieb, Alexander, Pawelka, Erich, Laferl, Hermann, Wenisch, Christoph, Neuhold, Stephanie, Haider, Doris, Stiasny, Karin, Bergthaler, Andreas, Puchhammer-Stoeckl, Elisabeth, Mirazimi, Ali, Montserrat, Nuria, Zhang, Haibo, Slutsky, Arthur S., Penninger, Josef M., (2020). Human recombinant soluble ACE2 in severe COVID-19 The Lancet Respiratory Medicine 8, (11), 1154-1158

Angiotensin converting enzyme 2 (ACE2) is the crucial severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor and protects multiple tissues, including the lung, from injury as a regulator of the renin–angiotensin system.1 Therefore, ACE2 has become the focus of COVID-19 research and a plethora of drug development efforts. Among the novel compounds under development is human recombinant soluble ACE2 (hrsACE2 [APN01; Apeiron Biologics, Vienna, Austria]), which has two mechanisms of action that theoretically should be of benefit in COVID-19.2 The first involves binding the viral spike protein and thereby neutralising SARS-CoV-2,3 and the second is minimising injury to multiple organs, including the lungs, kidneys, and heart, because of unabated renin–angiotensin system hyperactivation and increased angiotensin II concentrations.4, 5, 6 hrsACE2 has been tested in 89 patients, namely in healthy volunteers in phase 1 studies and in patients with acute respiratory distress syndrome (ARDS) in phase 2 clinical studies, with a good safety profile.7, 8 Moreover, hrsACE2 can reduce SARS-CoV-2 load by a factor of 1000–5000 in in-vitro cell-culture experiments and engineered organoids, directly demonstrating that ACE2 can effectively neutralise SARS-CoV-2.3 We describe in this Case Report the first course of treatment with hrsACE2 of a patient with severe COVID-19.

JTD


Monteil, Vanessa, Dyczynski, Matheus, Lauschke, Volker M., Kwon, Hyesoo, Wirnsberger, Gerald, Youhanna, Sonia, Zhang, Haibo, Slutsky, Arthur S., Hurtado del Pozo, Carmen, Horn, Moritz, Montserrat, Nuria, Penninger, Josef M., Mirazimi, Ali, (2020). Human soluble ACE2 improves the effect of remdesivir in SARS-CoV-2 infection EMBO Molecular Medicine , e13426

There is a critical need for safe and effective drugs for COVID-19. Only remdesivir has received authorization for COVID-19 and has been shown to improve outcomes but not decrease mortality. However, the dose of remdesivir is limited by hepatic and kidney toxicity. ACE2 is the critical cell surface receptor for SARS-CoV-2. Here, we investigated additive effect of combination therapy using remdesivir with recombinant soluble ACE2 (high/low dose) on Vero E6 and kidney organoids, targeting two different modalities of SARS-CoV-2 life cycle: cell entry via its receptor ACE2 and intracellular viral RNA replication. This combination treatment markedly improved their therapeutic windows against SARS-CoV-2 in both models. By using single amino-acid resolution screening in haploid ES cells, we report a singular critical pathway required for remdesivir toxicity, namely, Adenylate Kinase 2. The data provided here demonstrate that combining two therapeutic modalities with different targets, common strategy in HIV treatment, exhibit strong additive effects at sub-toxic concentrations. Our data lay the groundwork for the study of combinatorial regimens in future COVID-19 clinical trials.

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


Cilloni, Daniela, Petiti, Jessica, Campia, Valentina, Podestà , Marina, Squillario, Margherita, Montserrat, Nuria, Bertaina, Alice, Sabatini, Federica, Carturan, Sonia, Berger, Massimo, Saglio, Francesco, Bandini, Giuseppe, Bonifazi, Francesca, Fagioli, Franca, Moretta, Lorenzo, Saglio, Giuseppe, Verri, Alessandro, Barla, Annalisa, Locatelli, Franco, Frassoni, Francesco, (2020). Transplantation induces profound changes in the transcriptional asset of hematopoietic stem cells: Identification of specific signatures using machine learning techniques Journal of Clinical Medicine 9, (6), 1670

During the phase of proliferation needed for hematopoietic reconstitution following transplantation, hematopoietic stem/progenitor cells (HSPC) must express genes involved in stem cell self-renewal. We investigated the expression of genes relevant for self-renewal and expansion of HSPC (operationally defined as CD34+ cells) in steady state and after transplantation. Specifically, we evaluated the expression of ninety-one genes that were analyzed by real-time PCR in CD34+ cells isolated from (i) 12 samples from umbilical cord blood (UCB); (ii) 15 samples from bone marrow healthy donors; (iii) 13 samples from bone marrow after umbilical cord blood transplant (UCBT); and (iv) 29 samples from patients after transplantation with adult hematopoietic cells. The results show that transplanted CD34+ cells from adult cells acquire an asset very different from transplanted CD34+ cells from cord blood. Multivariate machine learning analysis (MMLA) showed that four specific gene signatures can be obtained by comparing the four types of CD34+ cells. In several, but not all cases, transplanted HSPC from UCB overexpress reprogramming genes. However, these remarkable changes do not alter the commitment to hematopoietic lineage. Overall, these results reveal undisclosed aspects of transplantation biology.

JTD Keywords: Hematopoietic stem/progenitor cell, Cord blood, Stem cell transplantation


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


Sample, Matthew, Boulicault, Marion, Allen, Caley, Bashir, Rashid, Hyun, Insoo, Levis, Megan, Lowenthal, Caroline, Mertz, David, Montserrat, Nuria, Palmer, Megan J., Saha, Krishanu, Zartman, Jeremiah, (2019). Multi-cellular engineered living systems: building a community around responsible research on emergence Biofabrication 11, (4), 043001

Ranging from miniaturized biological robots to organoids, multi-cellular engineered living systems (M-CELS) pose complex ethical and societal challenges. Some of these challenges, such as how to best distribute risks and benefits, are likely to arise in the development of any new technology. Other challenges arise specifically because of the particular characteristics of M-CELS. For example, as an engineered living system becomes increasingly complex, it may provoke societal debate about its moral considerability, perhaps necessitating protection from harm or recognition of positive moral and legal rights, particularly if derived from cells of human origin. The use of emergence-based principles in M-CELS development may also create unique challenges, making the technology difficult to fully control or predict in the laboratory as well as in applied medical or environmental settings. In response to these challenges, we argue that the M-CELS community has an obligation to systematically address the ethical and societal aspects of research and to seek input from and accountability to a broad range of stakeholders and publics. As a newly developing field, M-CELS has a significant opportunity to integrate ethically responsible norms and standards into its research and development practices from the start. With the aim of seizing this opportunity, we identify two general kinds of salient ethical issues arising from M-CELS research, and then present a set of commitments to and strategies for addressing these issues. If adopted, these commitments and strategies would help define M-CELS as not only an innovative field, but also as a model for responsible research and engineering.

JTD Keywords: Ethics, Society, Governance, Emergence, Moral considerability, Responsible innovation


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


Hernandez-Benitez, R., Llanos Martinez-Martinez, M., Lajara, J., Magistretti, P., Montserrat, N., Izpisua Belmonte, Juan Carlos, (2018). At the heart of genome editing and cardiovascular diseases Circulation Research 123, (2), 221-223

Cardiovascular disease (CVD) is still the leading cause of death worldwide, but the knowledge and technologies for counteracting this disease may already be in our hands. Scientific advances over the past few years, such as the isolation and differentiation of induced pluripotent stem cells, and the development of gene-editing tools, have enabled us to model CVD, but more importantly, may represent tools for CVD early diagnosis, patient stratification, and treatment.

JTD


Niederberger, Craig, Pellicer, Antonio, Cohen, Jacques, Gardner, David K., Palermo, Gianpiero D., O'Neill, Claire L., Chow, Stephen, Rosenwaks, Zev, Cobo, Ana, Swain, Jason E., Schoolcraft, William B., Frydman, René, Bishop, Lauren A., Aharon, Davora, Gordon, Catherine, New, Erika, Decherney, Alan, Tan, Seang Lin, Paulson, Richard J., Goldfarb, James M., Brännström, Mats, Donnez, Jacques, Silber, Sherman, Dolmans, Marie-Madeleine, Simpson, Joe Leigh, Handyside, Alan H., Munné, Santiago, Eguizabal, Cristina, Montserrat, Nuria, Izpisua Belmonte, Juan Carlos, Trounson, Alan, Simon, Carlos, Tulandi, Togas, Giudice, Linda C., Norman, Robert J., Hsueh, Aaron J., Sun, Yingpu, Laufer, Neri, Kochman, Ronit, Eldar-Geva, Talia, Lunenfeld, Bruno, Ezcurra, Diego, D'Hooghe, Thomas, Fauser, Bart C. J. M., Tarlatzis, Basil C., Meldrum, David R., Casper, Robert F., Fatemi, Human M., Devroey, Paul, Galliano, Daniela, Wikland, Matts, Sigman, Mark, Schoor, Richard A., Goldstein, Marc, Lipshultz, Larry I., Schlegel, Peter N., Hussein, Alayman, Oates, Robert D., Brannigan, Robert E., Ross, Heather E., Pennings, Guido, Klock, Susan C., Brown, Simon, Van Steirteghem, André, Rebar, Robert W., LaBarbera, Andrew R., (2018). Forty years of IVF Fertility and Sterility 110, (2), 185-324

This monograph, written by the pioneers of IVF and reproductive medicine, celebrates the history, achievements, and medical advancements made over the last 40 years in this rapidly growing field.

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


Climent, A. M., Hernandez-Romero, I., Guillem, M. S., Montserrat, N., Fernandez, M. E., Atienza, F., Fernandez-Aviles, F., (2017). High resolution microscopic optical mapping of anatomical and functional reentries in human cardiac cell cultures IEEE Conference Publications Computing in Cardiology Conference (CinC), 2016 , IEEE (Vancouver, Canada) 43, 233-236

Anatomical and/or functional reentries have been proposed as one of the main mechanism of perpetuation of cardiac fibrillation processes. However, technical limitations have difficult the characterization of those reentries and are hampering the development of effective anti-arrhythmic treatments. The goal of this study is to present a novel technology to map with high resolution the center of fibrillation drivers in order to characterize the mechanisms of reentry. Cell cultures of human cardiac-like cells differentiated from pluripotent stem cells were analyzed with a novel microscopic optical mapping system. The pharmacological response to verapamil administration of each type of reentry was analyzed. In all analyzed cell cultures, a reentry was identified as the mechanism of maintenance of the arrhythmia. Interestingly, the administration of verapamil produced opposite effects on activation rate depending on the mechanisms of reentry (i.e. anatomical or functional). Microscopic optical mapping of reentries allows the identification of perpetuation mechanisms which has been demonstrated to be linked with different pharmacological response.

JTD Keywords: Stem cells, Rotors, Microscopy, Optical filters, Calcium, Optical microscopy, Biomedical optical imaging


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


Xia, Yun, Montserrat, Nuria, Campistol, Josep M., Izpisua Belmonte, Juan Carlos, Remuzzi, Giuseppe, Williams, David F., (2017). Lineage reprogramming toward kidney regeneration Kidney Transplantation, Bioengineering and Regeneration (ed. Orlando, G., Remuzzi, Giuseppe, Williams, David F.), Academic Press (London, UK) , 1167-1175

We have known for decades that it is possible to switch the phenotype of one somatic cell type into another. Such epigenetic rewiring processes can be artificially managed and even reversed by using a defined set of transcription factors. Lineage reprogramming is very often defined as a process of converting one cell type into another without going through a pluripotent state, providing great promise for regenerative medicine. However, the identification of key transcription factors for lineage reprogramming is limited, due to the exhaustive and expensive experimental processes. Accumulating knowledge of genetic and epigenetic regulatory networks that are critical for defining a specific lineage provides unprecedented opportunities to model and predict pioneering factors that may drive directional lineage reprogramming to obtain the desired cell type.

JTD Keywords: Reprogramming, Pluripotency, Differentiation, Lineage specification, Epigenetic regulatory network, Regeneration


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


Eguizabal, C., Herrera, L., De Oñate, L., Montserrat, N., Hajkova, P., Izpisua Belmonte, J. C., (2016). Characterization of the epigenetic changes during human gonadal primordial germ cells reprogramming Stem Cells , 34, (9), 2418-2428

Abstract: Epigenetic reprogramming is a central process during mammalian germline development. Genome-wide DNA demethylation in primordial germ cells (PGCs) is a prerequisite for the erasure of epigenetic memory, preventing the transmission of epimutations to the next generation. Apart from DNA demethylation, germline reprogramming has been shown to entail reprogramming of histone marks and chromatin remodelling. Contrary to other animal models, there is limited information about the epigenetic dynamics during early germ cell development in humans. Here, we provide further characterization of the epigenetic configuration of the early human gonadal PGCs. We show that early gonadal human PGCs are DNA hypomethylated and their chromatin is characterized by low H3K9me2 and high H3K27me3 marks. Similarly to previous observations in mice, human gonadal PGCs undergo dynamic chromatin changes concomitant with the erasure of genomic imprints. Interestingly, and contrary to mouse early germ cells, expression of BLIMP1/PRDM1 persists in through all gestational stages in human gonadal PGCs and is associated with nuclear lysine-specific demethylase-1. Our work provides important additional information regarding the chromatin changes associated with human PGCs development between 6 and 13 weeks of gestation in male and female gonads.

JTD Keywords: Epigenetic, Human primordial germ cells, Reprograming


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


Reddy, Pradeep, Ocampo, Alejandro, Suzuki, Keiichiro, Luo, Jinping, Bacman, Sandra , Williams, Sion, Sugawara, Atsushi, Okamura, Daiji, Tsunekawa, Yuji, Wu, Jun, Lam, David, Xiong, Xiong, Montserrat, Nuria, Esteban, Concepcion, Liu, Guang-Hui, Sancho-Martinez, Ignacio, Manau, Dolors, Civico, Salva, Cardellach, Francesc, del Mar O'Callaghan, Maria, Campistol, Jaime, Zhao, Huimin, Campistol, Josep, Moraes, Carlos, Izpisua Belmonte, Juan Carlos, (2015). Selective elimination of mitochondrial mutations in the germline by genome editing Cell 161, (3), 459-469

Mitochondrial diseases include a group of maternally inherited genetic disorders caused by mutations in mtDNA. In most of these patients, mutated mtDNA coexists with wild-type mtDNA, a situation known as mtDNA heteroplasmy. Here, we report on a strategy toward preventing germline transmission of mitochondrial diseases by inducing mtDNA heteroplasmy shift through the selective elimination of mutated mtDNA. As a proof of concept, we took advantage of NZB/BALB heteroplasmic mice, which contain two mtDNA haplotypes, BALB and NZB, and selectively prevented their germline transmission using either mitochondria-targeted restriction endonucleases or TALENs. In addition, we successfully reduced human mutated mtDNA levels responsible for Leber?s hereditary optic neuropathy (LHOND), and neurogenic muscle weakness, ataxia, and retinitis pigmentosa (NARP), in mammalian oocytes using mitochondria-targeted TALEN (mito-TALENs). Our approaches represent a potential therapeutic avenue for preventing the transgenerational transmission of human mitochondrial diseases caused by mutations in mtDNA. Mitochondrial diseases include a group of maternally inherited genetic disorders caused by mutations in mtDNA. In most of these patients, mutated mtDNA coexists with wild-type mtDNA, a situation known as mtDNA heteroplasmy. Here, we report on a strategy toward preventing germline transmission of mitochondrial diseases by inducing mtDNA heteroplasmy shift through the selective elimination of mutated mtDNA. As a proof of concept, we took advantage of NZB/BALB heteroplasmic mice, which contain two mtDNA haplotypes, BALB and NZB, and selectively prevented their germline transmission using either mitochondria-targeted restriction endonucleases or TALENs. In addition, we successfully reduced human mutated mtDNA levels responsible for Leber?s hereditary optic neuropathy (LHOND), and neurogenic muscle weakness, ataxia, and retinitis pigmentosa (NARP), in mammalian oocytes using mitochondria-targeted TALEN (mito-TALENs). Our approaches represent a potential therapeutic avenue for preventing the transgenerational transmission of human mitochondrial diseases caused by mutations in mtDNA.

JTD


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


Garcia, A., Hortigüela, V., Lagunas, A., Cortina, C., Montserrat, N., Samitier, J., Martinez, Elena, (2014). Protein patterning on hydrogels by direct microcontact printing: application to cardiac differentiation RSC Advances 4, (55), 29120-29123

An extended microcontact printing technique to chemically pattern hydrogels is reported. The procedure employs standard polydimethylsiloxane stamps and requires minor pre-processing of the hydrogels by freeze-drying. Micropatterned Matrigel[trade mark sign] and gelatin hydrogels induce NIH-3T3 cell alignment and elongation. Furthermore, human embryonic stem cells cultured on fibronectin-patterned hydrogels display beating foci earlier than those cultured on non-patterned substrates.

JTD