1
|
Guimo F, Renard Y, Malafosse C, Dhenin G, Labrousse M, Duprey A. Unraveling Renal Arteries Morphogenesis from Tridimensional Human Embryos Reconstruction. Ann Vasc Surg 2024; 108:65-75. [PMID: 38942378 DOI: 10.1016/j.avsg.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/06/2024] [Accepted: 04/08/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND During human morphogenesis, the definitive kidneys derive from the metanephros during Carnegie Stage 14 to 23. The pronephros and the mesonephros develop previously and successively to finally lead to the formation of the urinary tract. Renal vascularization, first described in 1912 by Félix using a "ladder theory" model, is highly variable and current available morphogenesis descriptions do not explain all reported anatomical variations. The aim of this work was to study the morphogenesis of the human metanephros and its vascularization by three-dimensional reconstructions of human embryos. METHODS Histological sections of 23 human embryos from the Carnegie Collection and 5 human embryos from the French collection (Carnegie stages 14 to 23) were completely digitalized and reconstructed in three dimensions using specific softwares and then analyzed by descriptive method using manual annotation. RESULTS In all studied embryos, the mesonephric arteries did not reach the metanephros irrespective to the position of the metanephros during its cranial ascent. Before the end of the cranial metanephros migration (15 embryos), at the level of the aorto-iliac bifurcation, a "primitive" vascularization was shown in 9 of them. The renal artery originated from the primitive iliac arteries for 8 embryos and from the inferior mesenteric artery in one embryo. Further, a capillary cluster emerging from the lateral wall of the aorta and extending toward the metanephros was found in 2 embryos (Carnegie stages 21 and 22). This may correspond to a phenomenon of neoangiogenesis responsible of the definitive renal artery. CONCLUSIONS The present study reported the morphogenesis of human renal arteries between Carnegie stages 14 and 23 using an original method of tridimensional computerized reconstructions of historical human embryos. Some original findings, in contradiction with the original Felix's description, may explain the most frequently reported anatomical variations.
Collapse
Affiliation(s)
- François Guimo
- Faculty of Medicine, Université de Champagne-Ardennes, Reims, France; Department of Anatomy, Faculty of Medicine, Université de Champagne-Ardennes, Reims, France; Centre Hospitalier Universitaire de Reims, Reims, France.
| | - Yohann Renard
- Department of Anatomy, Faculty of Medicine, Université de Champagne-Ardennes, Reims, France; Centre Hospitalier Universitaire de Reims, Reims, France
| | - Clémentine Malafosse
- Faculty of Medicine, Université de Champagne-Ardennes, Reims, France; Centre Hospitalier Universitaire de Reims, Reims, France
| | - Grégoire Dhenin
- Faculty of Medicine, Université de Champagne-Ardennes, Reims, France
| | - Marc Labrousse
- Department of Anatomy, Faculty of Medicine, Université de Champagne-Ardennes, Reims, France; Centre Hospitalier Universitaire de Reims, Reims, France
| | | |
Collapse
|
2
|
Kühn S, Magno V, Zimmermann R, Limasale YDP, Atallah P, Stoppa A, Männel MJ, Thiele J, Friedrichs J, Freudenberg U, Werner C. Microgels With Electrostatically Controlled Molecular Affinity to Direct Morphogenesis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2409731. [PMID: 39449199 DOI: 10.1002/adma.202409731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/14/2024] [Indexed: 10/26/2024]
Abstract
Concentration gradients of soluble signaling molecules-morphogens-determine the cellular organization in tissue development. Morphogen-releasing microgels have shown potential to recapitulate this principle in engineered tissue constructs, however, with limited control over the molecular cues in space and time. Inspired by the functionality of sulfated glycosaminoglycans (sGAGs) in morphogen signaling in vivo, a library of sGAG-based microgels is developed and designated as µGel Units to Instruct Development (µGUIDEs). Adjustment of the microgel's sGAG sulfation patterns and concentration enabled the programming of electrostatic affinities that control the release of morphogens. Based on computational analyses of molecular transport processes, µGUIDEs provided unprecedented precision in the spatiotemporal modulation of vascular endothelial growth factor (VEGF) gradients in a microgel-in-gel vasculogenesis model and kidney organoid cultures. The versatile approach offers new options for creating morphogen signaling centers to advance the understanding of tissue and organ development.
Collapse
Affiliation(s)
- Sebastian Kühn
- Institute of Biofunctional Polymer Materials/Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden, Hohe Str. 6, 01069, Dresden, Germany
| | - Valentina Magno
- Institute of Biofunctional Polymer Materials/Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden, Hohe Str. 6, 01069, Dresden, Germany
| | - Ralf Zimmermann
- Institute of Biofunctional Polymer Materials/Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden, Hohe Str. 6, 01069, Dresden, Germany
| | - Yanuar Dwi Putra Limasale
- Institute of Biofunctional Polymer Materials/Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden, Hohe Str. 6, 01069, Dresden, Germany
| | - Passant Atallah
- Institute of Biofunctional Polymer Materials/Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden, Hohe Str. 6, 01069, Dresden, Germany
| | - Aukha Stoppa
- Institute of Biofunctional Polymer Materials/Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden, Hohe Str. 6, 01069, Dresden, Germany
| | - Max J Männel
- Institute of Physical Chemistry and Polymer Physics, Leibniz Institute of Polymer Research Dresden, Hohe Str. 6, 01069, Dresden, Germany
| | - Julian Thiele
- Institute of Physical Chemistry and Polymer Physics, Leibniz Institute of Polymer Research Dresden, Hohe Str. 6, 01069, Dresden, Germany
- Institute of Chemistry, Otto von Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Jens Friedrichs
- Institute of Biofunctional Polymer Materials/Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden, Hohe Str. 6, 01069, Dresden, Germany
| | - Uwe Freudenberg
- Institute of Biofunctional Polymer Materials/Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden, Hohe Str. 6, 01069, Dresden, Germany
- Center for Regenerative Therapies Dresden, Cluster of Excellence Physics of Life and Faculty of Chemistry and Food Chemistry, Dresden University of Technology, Fetscherstraße 105, 01307, Dresden, Germany
| | - Carsten Werner
- Institute of Biofunctional Polymer Materials/Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden, Hohe Str. 6, 01069, Dresden, Germany
- Center for Regenerative Therapies Dresden, Cluster of Excellence Physics of Life and Faculty of Chemistry and Food Chemistry, Dresden University of Technology, Fetscherstraße 105, 01307, Dresden, Germany
| |
Collapse
|
3
|
N'Guetta PEY, McLarnon SR, Tassou A, Geron M, Shirvan S, Hill RZ, Scherrer G, O'Brien LL. Comprehensive mapping of sensory and sympathetic innervation of the developing kidney. Cell Rep 2024; 43:114860. [PMID: 39412983 DOI: 10.1016/j.celrep.2024.114860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/23/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024] Open
Abstract
The kidneys act as finely tuned sensors to maintain physiological homeostasis. Both sympathetic and sensory nerves modulate kidney function through precise neural control. However, how the kidneys are innervated during development to support function remains elusive. Using light-sheet and confocal microscopy, we generated anatomical maps of kidney innervation across development. Kidney innervation commences on embryonic day 13.5 (E13.5) as network growth aligns with arterial differentiation. Fibers are synapsin I+, highlighting ongoing axonogenesis and potential signaling crosstalk. By E17.5, axons associate with nephrons, and the network continues to expand postnatally. CGRP+, substance P+, TRPV1+, and PIEZO2+ sensory fibers and TH+ sympathetic fibers innervate the developing kidney. TH+ and PIEZO2+ axons similarly innervate the human kidney, following the arterial tree to reach targets. Retrograde tracing revealed the primary dorsal root ganglia, T10-L2, from which sensory neurons project to the kidneys. Together, our findings elucidate the temporality and neuronal diversity of kidney innervation.
Collapse
Affiliation(s)
- Pierre-Emmanuel Y N'Guetta
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sarah R McLarnon
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Adrien Tassou
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matan Geron
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sepenta Shirvan
- Department of Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Rose Z Hill
- Department of Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lori L O'Brien
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; UNC Kidney Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
4
|
Sean G, Banes AJ, Gangaraju R. Organoids and tissue/organ chips. Stem Cell Res Ther 2024; 15:241. [PMID: 39098898 PMCID: PMC11299405 DOI: 10.1186/s13287-024-03859-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2024] Open
Affiliation(s)
- Graham Sean
- Department of Ophthalmology, University of Tennessee Health Science Center, 930 Madison Ave, Suite 768, Memphis, TN, 38163, USA
| | - Albert J Banes
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, and North Carolina State University, Chapel Hill, NC, USA
- Flexcell International Corporation, Burlington, NC, USA
| | - Rajashekhar Gangaraju
- Department of Ophthalmology, Anatomy & Neurobiology, University of Tennessee Health Science Center, 930 Madison Ave, Suite 768, Memphis, TN, 38163, USA.
| |
Collapse
|
5
|
Maggiore JC, LeGraw R, Przepiorski A, Velazquez J, Chaney C, Vanichapol T, Streeter E, Almuallim Z, Oda A, Chiba T, Silva-Barbosa A, Franks J, Hislop J, Hill A, Wu H, Pfister K, Howden SE, Watkins SC, Little MH, Humphreys BD, Kiani S, Watson A, Stolz DB, Davidson AJ, Carroll T, Cleaver O, Sims-Lucas S, Ebrahimkhani MR, Hukriede NA. A genetically inducible endothelial niche enables vascularization of human kidney organoids with multilineage maturation and emergence of renin expressing cells. Kidney Int 2024:S0085-2538(24)00407-1. [PMID: 38901605 DOI: 10.1016/j.kint.2024.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 05/10/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024]
Abstract
Vascularization plays a critical role in organ maturation and cell-type development. Drug discovery, organ mimicry, and ultimately transplantation hinge on achieving robust vascularization of in vitro engineered organs. Here, focusing on human kidney organoids, we overcame this hurdle by combining a human induced pluripotent stem cell (iPSC) line containing an inducible ETS translocation variant 2 (ETV2) (a transcription factor playing a role in endothelial cell development) that directs endothelial differentiation in vitro, with a non-transgenic iPSC line in suspension organoid culture. The resulting human kidney organoids show extensive endothelialization with a cellular identity most closely related to human kidney endothelia. Endothelialized kidney organoids also show increased maturation of nephron structures, an associated fenestrated endothelium with de novo formation of glomerular and venous subtypes, and the emergence of drug-responsive renin expressing cells. The creation of an engineered vascular niche capable of improving kidney organoid maturation and cell type complexity is a significant step forward in the path to clinical translation. Thus, incorporation of an engineered endothelial niche into a previously published kidney organoid protocol allowed the orthogonal differentiation of endothelial and parenchymal cell types, demonstrating the potential for applicability to other basic and translational organoid studies.
Collapse
Affiliation(s)
- Joseph C Maggiore
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Center for Integrative Organ Systems, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ryan LeGraw
- Department of Pathology, Division of Experimental Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Aneta Przepiorski
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jeremy Velazquez
- Department of Pathology, Division of Experimental Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Christopher Chaney
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Thitinee Vanichapol
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Evan Streeter
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Center for Integrative Organ Systems, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Zainab Almuallim
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Center for Integrative Organ Systems, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Akira Oda
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh Pennsylvania, USA
| | - Takuto Chiba
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh Pennsylvania, USA
| | - Anne Silva-Barbosa
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh Pennsylvania, USA
| | - Jonathan Franks
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Joshua Hislop
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alex Hill
- Department of Pathology, Division of Experimental Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Haojia Wu
- Division of Nephrology, Department of Medicine, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Katherine Pfister
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh Pennsylvania, USA
| | - Sara E Howden
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Simon C Watkins
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Melissa H Little
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia; Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Victoria, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA; Department of Developmental Biology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Samira Kiani
- Department of Pathology, Division of Experimental Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alan Watson
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Donna B Stolz
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alan J Davidson
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Tom Carroll
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Internal Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sunder Sims-Lucas
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh Pennsylvania, USA
| | - Mo R Ebrahimkhani
- Department of Pathology, Division of Experimental Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| | - Neil A Hukriede
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Center for Integrative Organ Systems, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
6
|
Kelam J, Kelam N, Filipović N, Komić L, Racetin A, Komić D, Kostić S, Kuzmić Prusac I, Vukojević K. Expression of Congenital Anomalies of the Kidney and Urinary Tract (CAKUT) Candidate Genes EDA2R, PCDH9, and TRAF7 in Normal Human Kidney Development and CAKUT. Genes (Basel) 2024; 15:702. [PMID: 38927638 PMCID: PMC11203332 DOI: 10.3390/genes15060702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Approximately half of the cases of chronic kidney disease (CKD) in childhood are caused by congenital anomalies of the kidney and urinary tract (CAKUT). Specific genes were identified as having significant importance in regard to the underlying genetic factors responsible for the CAKUT phenotype, and in our research, we focused on analyzing and comparing the expression levels of ectodysplasin A2 receptor (EDA2R), protocadherin9 (PCDH9), and TNF receptor-associated factor 7 (TRAF7) proteins in the cortex and medulla of healthy control kidneys during developmental phases 2, 3, and 4. We also performed an analysis of the area percentages of the mentioned proteins in the cortical and medullary sections of healthy embryonic and fetal kidneys compared to those affected by CAKUT, including duplex kidneys (DK), horseshoe kidneys (HK), hypoplastic kidneys (HYP), and dysplastic kidneys (DYS). We found that the CAKUT candidate gene proteins EDA2R, PCDH9, and TRAF7 are all expressed during normal human kidney development stages. In DYS, the expression of EDA2R was higher than in normal kidneys, likely due to EDA2R's role in apoptosis, which was upregulated in specific cases and could possibly contribute to the formation of DYS. The expression of PCDH9 was lower in HK, which can be attributed to the possible role of PCDH9 in cell migration suppression. Decreased PCDH9 expression is linked to increased cell migration, potentially contributing to the development of HK. The level of TRAF7 expression was reduced in all examined kidney disorders compared to normal kidneys, suggesting that this reduction might be attributed to the crucial role of TRAF7 in the formation of endothelium and ciliogenesis, both of which are essential for normal kidney development. Further research is required to ascertain the function of these proteins in both the typical development of the kidney and in CAKUT.
Collapse
Affiliation(s)
- Jelena Kelam
- Department of Family Medicine, Split-Dalmatia County Health Center, 21000 Split, Croatia; (J.K.); (L.K.)
| | - Nela Kelam
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (N.K.); (N.F.); (A.R.); (D.K.); (S.K.)
| | - Natalija Filipović
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (N.K.); (N.F.); (A.R.); (D.K.); (S.K.)
| | - Luka Komić
- Department of Family Medicine, Split-Dalmatia County Health Center, 21000 Split, Croatia; (J.K.); (L.K.)
| | - Anita Racetin
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (N.K.); (N.F.); (A.R.); (D.K.); (S.K.)
| | - Dora Komić
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (N.K.); (N.F.); (A.R.); (D.K.); (S.K.)
| | - Sandra Kostić
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (N.K.); (N.F.); (A.R.); (D.K.); (S.K.)
| | - Ivana Kuzmić Prusac
- Department of Pathology, University Hospital Center Split, 21000 Split, Croatia;
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, 21000 Split, Croatia; (N.K.); (N.F.); (A.R.); (D.K.); (S.K.)
- Department of Anatomy, School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- Center for Translational Research in Biomedicine, School of Medicine, University of Split, 21000 Split, Croatia
| |
Collapse
|
7
|
Pecksen E, Tkachuk S, Schröder C, Vives Enrich M, Neog A, Johnson CP, Lachmann N, Haller H, Kiyan Y. Monocytes prevent apoptosis of iPSCs and promote differentiation of kidney organoids. Stem Cell Res Ther 2024; 15:132. [PMID: 38702808 PMCID: PMC11069262 DOI: 10.1186/s13287-024-03739-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Induced pluripotent stem cells (iPSCs)-derived kidney organoids are a promising model for studying disease mechanisms and renal development. Despite several protocols having been developed, further improvements are needed to overcome existing limitations and enable a wider application of this model. One of the approaches to improve the differentiation of renal organoids in vitro is to include in the system cell types important for kidney organogenesis in vivo, such as macrophages. Another approach could be to improve cell survival. Mesodermal lineage differentiation is the common initial step of the reported protocols. The glycogen synthase kinase-3 (GSK-3) activity inhibitor, CHIR99021 (CHIR), is applied to induce mesodermal differentiation. It has been reported that CHIR simultaneously induces iPSCs apoptosis that can compromise cell differentiation. We thought to interfere with CHIR-induced apoptosis of iPSCs using rapamycin. METHODS Differentiation of kidney organoids from human iPSCs was performed. Cell survival and autophagy were analyzed using Cell counting kit 8 (CCK8) kit and Autophagy detection kit. Cells were treated with rapamycin or co-cultured with human monocytes isolated from peripheral blood or iPSCs-macrophages using a transwell co-culture system. Monocyte-derived extracellular vesicles (EVs) were isolated using polyethylene glycol precipitation. Expression of apoptotic markers cleaved Caspase 3, Poly [ADP-ribose] polymerase 1 (PARP-1) and markers of differentiation T-Box Transcription Factor 6 (TBX6), odd-skipped related 1 (OSR1), Nephrin, E-Cadherin, Paired box gene 2 (Pax2) and GATA Binding Protein 3 (Gata3) was assessed by RT-PCR and western blotting. Organoids were imaged by 3D-confocal microscopy. RESULTS We observed that CHIR induced apoptosis of iPSCs during the initial stage of renal organoid differentiation. Underlying mechanisms implied the accumulation of reactive oxygen species and decreased autophagy. Activation of autophagy by rapamacin and by an indirect co-culture of differentiating iPSCs with iPSCs-macrophages and human peripheral blood monocytes prevented apoptosis induced by CHIR. Furthermore, monocytes (but not rapamycin) strongly promoted expression of renal differentiation markers and organoids development via released extracellular vesicles. CONCLUSION Our data suggest that co-culturing of iPSCs with human monocytes strongly improves differentiation of kidney organoids. An underlying mechanism of monocytic action implies, but not limited to, an increased autophagy in CHIR-treated iPSCs. Our findings enhance the utility of kidney organoid models.
Collapse
Affiliation(s)
- Ekaterina Pecksen
- Clinics for Kidney and Hypertension Disease, Hannover Medical School, Hannover, Germany
| | - Sergey Tkachuk
- Clinics for Kidney and Hypertension Disease, Hannover Medical School, Hannover, Germany
| | - Cristoph Schröder
- Clinics for Kidney and Hypertension Disease, Hannover Medical School, Hannover, Germany
| | - Marc Vives Enrich
- Clinics for Kidney and Hypertension Disease, Hannover Medical School, Hannover, Germany
- Mount Desert Island Biological Laboratory, Bar Harbor, Maine, USA
| | - Anindita Neog
- Clinics for Kidney and Hypertension Disease, Hannover Medical School, Hannover, Germany
- Mount Desert Island Biological Laboratory, Bar Harbor, Maine, USA
| | - Cory P Johnson
- Mount Desert Island Biological Laboratory, Bar Harbor, Maine, USA
| | - Niko Lachmann
- Department of Pediatric Pneumology Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover, Germany
| | - Hermann Haller
- Clinics for Kidney and Hypertension Disease, Hannover Medical School, Hannover, Germany
- Mount Desert Island Biological Laboratory, Bar Harbor, Maine, USA
| | - Yulia Kiyan
- Clinics for Kidney and Hypertension Disease, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
8
|
Dequiedt L, Forjaz A, Lo JO, McCarty O, Wu PH, Rosenberg A, Wirtz D, Kiemen A. Three-dimensional reconstruction of fetal rhesus macaque kidneys at single-cell resolution reveals complex inter-relation of structures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.07.570622. [PMID: 38106004 PMCID: PMC10723390 DOI: 10.1101/2023.12.07.570622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Kidneys are among the most structurally complex organs in the body. Their architecture is critical to ensure proper function and is often impacted by diseases such as diabetes and hypertension. Understanding the spatial interplay between the different structures of the nephron and renal vasculature is crucial. Recent efforts have demonstrated the value of three-dimensional (3D) imaging in revealing new insights into the various components of the kidney; however, these studies used antibodies or autofluorescence to detect structures and so were limited in their ability to compare the many subtle structures of the kidney at once. Here, through 3D reconstruction of fetal rhesus macaque kidneys at cellular resolution, we demonstrate the power of deep learning in exhaustively labelling seventeen microstructures of the kidney. Using these tissue maps, we interrogate the spatial distribution and spatial correlation of the glomeruli, renal arteries, and the nephron. This work demonstrates the power of deep learning applied to 3D tissue images to improve our ability to compare many microanatomical structures at once, paving the way for further works investigating renal pathologies.
Collapse
Affiliation(s)
- Lucie Dequiedt
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - André Forjaz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
| | - Jamie O Lo
- Department of Obstetrics and Gynecology, Oregon Health and Sciences University
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center
| | - Owen McCarty
- Department of Biomedical Engineering, Oregon Health and Sciences University
| | - Pei-Hsun Wu
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
- Institute for NanoBioTechnology, Johns Hopkins University
| | - Avi Rosenberg
- Department of Pathology, Johns Hopkins School of Medicine
| | - Denis Wirtz
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University
- Institute for NanoBioTechnology, Johns Hopkins University
- Department of Pathology, Johns Hopkins School of Medicine
- Department of Oncology, Johns Hopkins School of Medicine
| | - Ashley Kiemen
- Institute for NanoBioTechnology, Johns Hopkins University
- Department of Pathology, Johns Hopkins School of Medicine
- Department of Oncology, Johns Hopkins School of Medicine
| |
Collapse
|
9
|
N’Guetta PEY, McLarnon SR, Tassou A, Geron M, Shirvan S, Hill RZ, Scherrer G, O’Brien LL. Comprehensive mapping of sensory and sympathetic innervation of the developing kidney. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.15.567276. [PMID: 38496522 PMCID: PMC10942422 DOI: 10.1101/2023.11.15.567276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
The kidney functions as a finely tuned sensor to balance body fluid composition and filter out waste through complex coordinated mechanisms. This versatility requires tight neural control, with innervating efferent nerves playing a crucial role in regulating blood flow, glomerular filtration rate, water and sodium reabsorption, and renin release. In turn sensory afferents provide feedback to the central nervous system for the modulation of cardiovascular function. However, the cells targeted by sensory afferents and the physiological sensing mechanisms remain poorly characterized. Moreover, how the kidney is innervated during development to establish these functions remains elusive. Here, we utilized a combination of light-sheet and confocal microscopy to generate anatomical maps of kidney sensory and sympathetic nerves throughout development and resolve the establishment of functional crosstalk. Our analyses revealed that kidney innervation initiates at embryonic day (E)13.5 as the nerves associate with vascular smooth muscle cells and follow arterial differentiation. By E17.5 axonal projections associate with kidney structures such as glomeruli and tubules and the network continues to expand postnatally. These nerves are synapsin I-positive, highlighting ongoing axonogenesis and the potential for functional crosstalk. We show that sensory and sympathetic nerves innervate the kidney concomitantly and classify the sensory fibers as calcitonin gene related peptide (CGRP)+, substance P+, TRPV1+, and PIEZO2+, establishing the presence of PIEZO2 mechanosensory fibers in the kidney. Using retrograde tracing, we identified the primary dorsal root ganglia, T10-L2, from which PIEZO2+ sensory afferents project to the kidney. Taken together our findings elucidate the temporality of kidney innervation and resolve the identity of kidney sympathetic and sensory nerves.
Collapse
Affiliation(s)
- Pierre-Emmanuel Y. N’Guetta
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sarah R. McLarnon
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Adrien Tassou
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matan Geron
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sepenta Shirvan
- Department of Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA 92037
| | - Rose Z. Hill
- Department of Neuroscience, Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA, USA 92037
| | - Grégory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; New York Stem Cell Foundation – Robertson Investigator, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lori L. O’Brien
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
10
|
Luo PM, Gu X, Chaney C, Carroll T, Cleaver O. Stromal netrin 1 coordinates renal arteriogenesis and mural cell differentiation. Development 2023; 150:dev201884. [PMID: 37823339 PMCID: PMC10690105 DOI: 10.1242/dev.201884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 10/02/2023] [Indexed: 10/13/2023]
Abstract
The kidney vasculature has a complex architecture that is essential for renal function. The molecular mechanisms that direct development of kidney blood vessels are poorly characterized. We identified a regionally restricted, stroma-derived signaling molecule, netrin 1 (Ntn1), as a regulator of renal vascular patterning in mice. Stromal progenitor (SP)-specific ablation of Ntn1 (Ntn1SPKO) resulted in smaller kidneys with fewer glomeruli, as well as profound defects of the renal artery and transient blood flow disruption. Notably, Ntn1 ablation resulted in loss of arterial vascular smooth muscle cell (vSMC) coverage and in ectopic SMC deposition at the kidney surface. This was accompanied by dramatic reduction of arterial tree branching that perdured postnatally. Transcriptomic analysis of Ntn1SPKO kidneys revealed dysregulation of vSMC differentiation, including downregulation of Klf4, which we find expressed in a subset of SPs. Stromal Klf4 deletion similarly resulted in decreased smooth muscle coverage and arterial branching without, however, the disruption of renal artery patterning and perfusion seen in Ntn1SPKO. These data suggest a stromal Ntn1-Klf4 axis that regulates stromal differentiation and reinforces stromal-derived smooth muscle as a key regulator of renal blood vessel formation.
Collapse
Affiliation(s)
- Peter M. Luo
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
- Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Xiaowu Gu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Christopher Chaney
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
- Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
- Internal Medicine and Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Thomas Carroll
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
- Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
- Internal Medicine and Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
- Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| |
Collapse
|
11
|
Honeycutt SE, N'Guetta PEY, Hardesty DM, Xiong Y, Cooper SL, Stevenson MJ, O'Brien LL. Netrin 1 directs vascular patterning and maturity in the developing kidney. Development 2023; 150:dev201886. [PMID: 37818607 PMCID: PMC10690109 DOI: 10.1242/dev.201886] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 10/02/2023] [Indexed: 10/12/2023]
Abstract
The intricate vascular system of the kidneys supports body fluid and organ homeostasis. However, little is known about how vascular architecture is established during kidney development. More specifically, how signals from the kidney influence vessel maturity and patterning remains poorly understood. Netrin 1 (Ntn1) is a secreted ligand that is crucial for vessel and neuronal guidance. Here, we demonstrate that Ntn1 is expressed by Foxd1+ stromal progenitors in the developing mouse kidney and conditional deletion (Foxd1GC/+;Ntn1fl/fl) results in hypoplastic kidneys with extended nephrogenesis. Wholemount 3D analyses additionally revealed the loss of a predictable vascular pattern in Foxd1GC/+;Ntn1fl/fl kidneys. As vascular patterning has been linked to vessel maturity, we investigated arterialization. Quantification of the CD31+ endothelium at E15.5 revealed no differences in metrics such as the number of branches or branch points, whereas the arterial vascular smooth muscle metrics were significantly reduced at both E15.5 and P0. In support of our observed phenotypes, whole kidney RNA-seq revealed disruptions to genes and programs associated with stromal cells, vasculature and differentiating nephrons. Together, our findings highlight the significance of Ntn1 to proper vascularization and kidney development.
Collapse
Affiliation(s)
- Samuel E. Honeycutt
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Pierre-Emmanuel Y. N'Guetta
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Deanna M. Hardesty
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yubin Xiong
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Shamus L. Cooper
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew J. Stevenson
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lori L. O'Brien
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
12
|
Luxen M, Zwiers PJ, Meester F, Jongman RM, Kuiper T, Moser J, Pultar M, Skalicky S, Diendorfer AB, Hackl M, van Meurs M, Molema G. Unique miRNome and transcriptome profiles underlie microvascular heterogeneity in mouse kidney. Am J Physiol Renal Physiol 2023; 325:F299-F316. [PMID: 37410897 PMCID: PMC10511173 DOI: 10.1152/ajprenal.00005.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/08/2023] Open
Abstract
Endothelial cells in blood vessels in the kidney exert different functions depending on the (micro)vascular bed they are located in. The present study aimed to investigate microRNA and mRNA transcription patterns that underlie these differences. We zoomed in on microvascular compartments in the mouse renal cortex by laser microdissecting the microvessels prior to small RNA- and RNA-sequencing analyses. By these means, we characterized microRNA and mRNA transcription profiles of arterioles, glomeruli, peritubular capillaries, and postcapillary venules. Quantitative RT-PCR, in situ hybridization, and immunohistochemistry were used to validate sequencing results. Unique microRNA and mRNA transcription profiles were found in all microvascular compartments, with dedicated marker microRNAs and mRNAs showing enriched transcription in a single microvascular compartment. In situ hybridization validated the localization of microRNAs mmu-miR-140-3p in arterioles, mmu-miR-322-3p in glomeruli, and mmu-miR-451a in postcapillary venules. Immunohistochemical staining showed that von Willebrand factor protein was mainly expressed in arterioles and postcapillary venules, whereas GABRB1 expression was enriched in glomeruli, and IGF1 was enriched in postcapillary venules. More than 550 compartment-specific microRNA-mRNA interaction pairs were identified that carry functional implications for microvascular behavior. In conclusion, our study identified unique microRNA and mRNA transcription patterns in microvascular compartments of the mouse kidney cortex that underlie microvascular heterogeneity. These patterns provide important molecular information for future studies into differential microvascular engagement in health and disease.NEW & NOTEWORTHY Renal endothelial cells display a high level of heterogeneity depending on the (micro)vascular bed they reside in. The molecular basis contributing to these differences is poorly understood yet of high importance to increase understanding of microvascular engagement in the kidney in health and disease. This report describes m(icro)RNA expression profiles of microvascular beds in the mouse renal cortex and uncovers microvascular compartment-specific m(icro)RNAs and miRNA-mRNA pairs, thereby revealing important molecular mechanisms underlying renal microvascular heterogeneity.
Collapse
Affiliation(s)
- Matthijs Luxen
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Peter J Zwiers
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Femke Meester
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Rianne M Jongman
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Anaesthesiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Timara Kuiper
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jill Moser
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | | | | - Matijs van Meurs
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Grietje Molema
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
13
|
Tarnick J, Elhendawi M, Holland I, Chang Z, Davies JA. Innervation of the developing kidney in vivo and in vitro. Biol Open 2023; 12:bio060001. [PMID: 37439314 PMCID: PMC10411870 DOI: 10.1242/bio.060001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/03/2023] [Indexed: 07/14/2023] Open
Abstract
Within the adult kidney, renal neurites can be observed alongside the arteries where they play a role in regulating blood flow. However, their role and localization during development has so far not been described in detail. In other tissues, such as the skin of developing limb buds, neurons play an important role during arterial differentiation. Here, we aim to investigate whether renal nerves could potentially carry out a similar role during arterial development in the mouse kidney. In order to do so, we used whole-mount immunofluorescence staining to identify whether the timing of neuronal innervation correlates with the recruitment of arterial smooth muscle cells. Our results show that neurites innervate the kidney between day 13.5 and 14.5 of development, arriving after the recruitment of smooth muscle actin-positive cells to the renal arteries.
Collapse
Affiliation(s)
- Julia Tarnick
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Mona Elhendawi
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Ian Holland
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Ziyuan Chang
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Jamie A. Davies
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh EH8 9XD, UK
| |
Collapse
|
14
|
Shi M, Fu P, Bonventre JV, McCracken KW. Directed differentiation of ureteric bud and collecting duct organoids from human pluripotent stem cells. Nat Protoc 2023; 18:2485-2508. [PMID: 37460630 PMCID: PMC11154671 DOI: 10.1038/s41596-023-00847-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 04/24/2023] [Indexed: 08/09/2023]
Abstract
Developing models of human kidney tissue in vitro is an important challenge in regenerative nephrology research, given the paucity of novel and effective therapies in kidney disease. However, the de novo generation of kidney tissues from human pluripotent stem cells (hPSCs) is challenging owing to the structural and functional complexity of the organ, as well its developmental origin from two distinct embryologic populations: the metanephric mesenchyme and the ureteric bud (UB). Directed differentiation strategies have been developed to generate kidney organoids containing nephron-like structures; we recently reported an efficient and practical method to generate UB tissues. Here, we describe a detailed step-by-step protocol for differentiation of hPSCs into three-dimensonal UB organoids that exhibit complex morphological development and the capacity to differentiate into functional collecting duct tissues. Over 3 d, hPSCs are induced into PAX2+GATA3+ pronephric (anterior) intermediate mesoderm fates in monolayer cultures at high efficiency. The cells are aggregated into three-dimensional spheroids, which then assemble and organize into nephric duct-like tissue over 4 d. When embedded into an extracellular matrix, the spheroids grow into UB organoids that recapitulate fetal branching morphogenesis for 1 week of culture. When switched to permissive conditions, the UB organoids spontaneously differentiate to form collecting duct principal cells. This approach provides robust and reproducible methods that can be readily adopted by users with basic experience in hPSC and organoid differentiation to generate UB tissues, which may be used to investigate human kidney development, model disease processes and catalyze further efforts in engineering functional kidney tissue.
Collapse
Affiliation(s)
- Min Shi
- Division of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ping Fu
- Division of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, China
| | - Joseph V Bonventre
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge and Boston, MA, USA.
- Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Kyle W McCracken
- Division of Renal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Division of Nephrology, Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
15
|
Barlow HR, Ahuja N, Bierschenk T, Htike Y, Fassetta L, Azizoglu DB, Flores J, Gao N, de la O S, Sneddon JB, Marciano DK, Cleaver O. Rab11 is essential to pancreas morphogenesis, lumen formation and endocrine mass. Dev Biol 2023; 499:59-74. [PMID: 37172642 DOI: 10.1016/j.ydbio.2023.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/21/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
The molecular links between tissue-level morphogenesis and the differentiation of cell lineages in the pancreas remain elusive despite a decade of studies. We previously showed that in pancreas both processes depend on proper lumenogenesis. The Rab GTPase Rab11 is essential for epithelial lumen formation in vitro, however few studies have addressed its functions in vivo and none have tested its requirement in pancreas. Here, we show that Rab11 is critical for proper pancreas development. Co-deletion of the Rab11 isoforms Rab11A and Rab11B in the developing pancreatic epithelium (Rab11pancDKO) results in ∼50% neonatal lethality and surviving adult Rab11pancDKO mice exhibit defective endocrine function. Loss of both Rab11A and Rab11B in the embryonic pancreas results in morphogenetic defects of the epithelium, including defective lumen formation and lumen interconnection. In contrast to wildtype cells, Rab11pancDKO cells initiate the formation of multiple ectopic lumens, resulting in a failure to coordinate a single apical membrane initiation site (AMIS) between groups of cells. This results in a failure to form ducts with continuous lumens. Here, we show that these defects are due to failures in vesicle trafficking, as apical and junctional components remain trapped within Rab11pancDKO cells. Together, these observations suggest that Rab11 directly regulates epithelial lumen formation and morphogenesis. Our report links intracellular trafficking to organ morphogenesis in vivo and presents a novel framework for decoding pancreatic development.
Collapse
Affiliation(s)
- Haley R Barlow
- Department of Molecular Biology, Center for Regenerative Science and Medicine, USA.
| | - Neha Ahuja
- Department of Molecular Biology, Center for Regenerative Science and Medicine, USA
| | - Tyler Bierschenk
- Department of Molecular Biology, Center for Regenerative Science and Medicine, USA
| | - Yadanar Htike
- Department of Molecular Biology, Center for Regenerative Science and Medicine, USA
| | - Luke Fassetta
- Department of Molecular Biology, Center for Regenerative Science and Medicine, USA
| | - D Berfin Azizoglu
- Department of Developmental Biology, Beckman Center, 279 W. Campus Drive, B300, Stanford, CA, 94305, USA
| | - Juan Flores
- Rutgers University Microbiome Program, 679 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Nan Gao
- Rutgers University Microbiome Program, 679 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Sean de la O
- Department of Cell and Tissue Biology, Department of Anatomy, Diabetes Center, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Julie B Sneddon
- Department of Cell and Tissue Biology, Department of Anatomy, Diabetes Center, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Denise K Marciano
- Internal Medicine and Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Ondine Cleaver
- Department of Molecular Biology, Center for Regenerative Science and Medicine, USA.
| |
Collapse
|
16
|
Honeycutt SE, N’Guetta PEY, Hardesty DM, Xiong Y, Cooper SL, O’Brien LL. Netrin-1 directs vascular patterning and maturity in the developing kidney. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.14.536975. [PMID: 37131589 PMCID: PMC10153117 DOI: 10.1101/2023.04.14.536975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Blood filtering by the kidney requires the establishment of an intricate vascular system that works to support body fluid and organ homeostasis. Despite these critical roles, little is known about how vascular architecture is established during kidney development. More specifically, how signals from the kidney influence vessel maturity and patterning remains poorly understood. Netrin-1 (Ntn1) is a secreted ligand critical for vessel and neuronal guidance. Here, we demonstrate that Ntn1 is expressed by stromal progenitors in the developing kidney, and conditional deletion of Ntn1 from Foxd1+ stromal progenitors (Foxd1GC/+;Ntn1fl/fl) results in hypoplastic kidneys that display extended nephrogenesis. Despite expression of the netrin-1 receptor Unc5c in the adjacent nephron progenitor niche, Unc5c knockout kidneys develop normally. The netrin-1 receptor Unc5b is expressed by embryonic kidney endothelium and therefore we interrogated the vascular networks of Foxd1GC/+;Ntn1fl/fl kidneys. Wholemount, 3D analyses revealed the loss of a predictable vascular pattern in mutant kidneys. As vascular patterning has been linked to vessel maturity, we investigated arterialization in these mutants. Quantification of the CD31+ endothelium at E15.5 revealed no differences in metrics such as the number of branches or branch points, whereas the arterial vascular smooth muscle metrics were significantly reduced at both E15.5 and P0. In support of these results, whole kidney RNA-seq showed upregulation of angiogenic programs and downregulation of muscle-related programs which included smooth muscle-associated genes. Together, our findings highlight the significance of netrin-1 to proper vascularization and kidney development.
Collapse
Affiliation(s)
- Samuel Emery Honeycutt
- Department of Cell Biology and Physiology University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Deanna Marie Hardesty
- Department of Cell Biology and Physiology University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yubin Xiong
- Department of Cell Biology and Physiology University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Shamus Luke Cooper
- Department of Cell Biology and Physiology University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lori Lynn O’Brien
- Department of Cell Biology and Physiology University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
17
|
Mierke CT. Physical and biological advances in endothelial cell-based engineered co-culture model systems. Semin Cell Dev Biol 2023; 147:58-69. [PMID: 36732105 DOI: 10.1016/j.semcdb.2023.01.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 02/04/2023]
Abstract
Scientific knowledge in the field of cell biology and mechanobiology heavily leans on cell-based in vitro experiments and models that favor the examination and comprehension of certain biological processes and occurrences across a variety of environments. Cell culture assays are an invaluable instrument for a vast spectrum of biomedical and biophysical investigations. The quality of experimental models in terms of simplicity, reproducibility, and combinability with other methods, and in particular the scale at which they depict cell fate in native tissues, is critical to advancing the knowledge of the comprehension of cell-cell and cell-matrix interactions in tissues and organs. Typically, in vitro models are centered on the experimental tinkering of mammalian cells, most often cultured as monolayers on planar, two-dimensional (2D) materials. Notwithstanding the significant advances and numerous findings that have been accomplished with flat biology models, their usefulness for generating further new biological understanding is constrained because the simple 2D setting does not reproduce the physiological response of cells in natural living tissues. In addition, the co-culture systems in a 2D stetting weakly mirror their natural environment of tissues and organs. Significant advances in 3D cell biology and matrix engineering have resulted in the creation and establishment of a new type of cell culture shapes that more accurately represents the in vivo microenvironment and allows cells and their interactions to be analyzed in a biomimetic approach. Contemporary biomedical and biophysical science has novel advances in technology that permit the design of more challenging and resilient in vitro models for tissue engineering, with a particular focus on scaffold- or hydrogel-based formats, organotypic cultures, and organs-on-chips, which cover the purposes of co-cultures. Even these complex systems must be kept as simplified as possible in order to grasp a particular section of physiology too very precisely. In particular, it is highly appreciated that they bridge the space between conventional animal research and human (patho)physiology. In this review, the recent progress in 3D biomimetic culturation is presented with a special focus on co-cultures, with an emphasis on the technological building blocks and endothelium-based co-culture models in cancer research that are available for the development of more physiologically relevant in vitro models of human tissues under normal and diseased conditions. Through applications and samples of various physiological and disease models, it is possible to identify the frontiers and future engagement issues that will have to be tackled to integrate synthetic biomimetic culture systems far more successfully into biomedical and biophysical investigations.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, Leipzig, Germany.
| |
Collapse
|
18
|
Donnan MD, Deb DK, Onay T, Scott RP, Ni E, Zhou Y, Quaggin SE. Formation of the glomerular microvasculature is regulated by VEGFR-3. Am J Physiol Renal Physiol 2023; 324:F91-F105. [PMID: 36395385 PMCID: PMC9836230 DOI: 10.1152/ajprenal.00066.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 10/12/2022] [Accepted: 11/02/2022] [Indexed: 11/18/2022] Open
Abstract
Microvascular dysfunction is a key driver of kidney disease. Pathophysiological changes in the kidney vasculature are regulated by vascular endothelial growth factor receptors (VEGFRs), supporting them as potential therapeutic targets. The tyrosine kinase receptor VEGFR-3, encoded by FLT4 and activated by the ligands VEGF-C and VEGF-D, is best known for its role in lymphangiogenesis. Therapeutically targeting VEGFR-3 to modulate lymphangiogenesis has been proposed as a strategy to treat kidney disease. However, outside the lymphatics, VEGFR-3 is also expressed in blood vascular endothelial cells in several tissues including the kidney. Here, we show that Vegfr-3 is expressed in fenestrated microvascular beds within the developing and adult mouse kidney, which include the glomerular capillary loops. We found that expression levels of VEGFR-3 are dynamic during glomerular capillary loop development, with the highest expression observed during endothelial cell migration into the S-shaped glomerular body. We developed a conditional knockout mouse model for Vegfr-3 and found that loss of Vegfr-3 resulted in a striking glomerular phenotype characterized by aneurysmal dilation of capillary loops, absence of mesangial structure, abnormal interendothelial cell junctions, and poor attachment between glomerular endothelial cells and the basement membrane. In addition, we demonstrated that expression of the VEGFR-3 ligand VEGF-C by podocytes and mesangial cells is dispensable for glomerular development. Instead, VEGFR-3 in glomerular endothelial cells attenuates VEGFR-2 phosphorylation. Together, the results of our study support a VEGF-C-independent functional role for VEGFR-3 in the kidney microvasculature outside of lymphatic vessels, which has implications for clinical therapies that target this receptor.NEW & NOTEWORTHY Targeting VEGFR-3 in kidney lymphatics has been proposed as a method to treat kidney disease. However, expression of VEGFR-3 is not lymphatic-specific. We demonstrated developmental expression of VEGFR-3 in glomerular endothelial cells, with loss of Vegfr-3 leading to malformation of glomerular capillary loops. Furthermore, we showed that VEGFR-3 attenuates VEGFR-2 activity in glomerular endothelial cells independent of paracrine VEGF-C signaling. Together, these data provide valuable information for therapeutic development targeting these pathways.
Collapse
Affiliation(s)
- Michael D Donnan
- Northwestern University Feinberg School of Medicine, Feinberg Cardiovascular and Renal Research Institute, Chicago, Illinois
| | - Dilip K Deb
- Northwestern University Feinberg School of Medicine, Feinberg Cardiovascular and Renal Research Institute, Chicago, Illinois
| | - Tuncer Onay
- Northwestern University Feinberg School of Medicine, Feinberg Cardiovascular and Renal Research Institute, Chicago, Illinois
| | - Rizaldy P Scott
- Northwestern University Feinberg School of Medicine, Feinberg Cardiovascular and Renal Research Institute, Chicago, Illinois
| | - Eric Ni
- Lake Erie College of Osteopathic Medicine, Greensburg, Pennsylvania
| | - Yalu Zhou
- Northwestern University Feinberg School of Medicine, Feinberg Cardiovascular and Renal Research Institute, Chicago, Illinois
| | - Susan E Quaggin
- Northwestern University Feinberg School of Medicine, Feinberg Cardiovascular and Renal Research Institute, Chicago, Illinois
| |
Collapse
|
19
|
Shi M, Maique JO, Cleaver O, Moe OW, Hu MC. VEGFR2 insufficiency enhances phosphotoxicity and undermines Klotho's protection against peritubular capillary rarefaction and kidney fibrosis. Am J Physiol Renal Physiol 2023; 324:F106-F123. [PMID: 36395384 PMCID: PMC9799155 DOI: 10.1152/ajprenal.00149.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/17/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) and its cognate receptor (VEGFR2) system are crucial for cell functions associated with angiogenesis and vasculogenesis. Klotho contributes to vascular health maintenance in the kidney and other organs in mammals, but it is unknown whether renoprotection by Klotho is dependent on VEGF/VEGFR2 signaling. We used heterozygous VEGFR2-haploinsufficient (VEGFR2+/-) mice resulting from heterozygous knockin of green fluorescent protein in the locus of fetal liver kinase 1 encoding VEGFR2 to test the interplay of Klotho, phosphate, and VEGFR2 in kidney function, the vasculature, and fibrosis. VEGFR2+/- mice displayed downregulated VEGF/VEGFR2 signaling in the kidney, lower density of peritubular capillaries, and accelerated kidney fibrosis, all of which were also found in the homozygous Klotho hypomorphic mice. High dietary phosphate induced higher plasma phosphate, greater peritubular capillary rarefaction, and more kidney fibrosis in VEGFR2+/- mice compared with wild-type mice. Genetic overexpression of Klotho significantly attenuated the elevated plasma phosphate, kidney dysfunction, peritubular capillary rarefaction, and kidney fibrosis induced by a high-phosphate diet in wild-type mice but only modestly ameliorated these changes in the VEGFR2+/- background. In cultured endothelial cells, VEGFR2 inhibition reduced free VEGFR2 but enhanced its costaining of an endothelial marker (CD31) and exacerbated phosphotoxicity. Klotho protein maintained VEGFR2 expression and attenuated high phosphate-induced cell injury, which was reduced by VEGFR2 inhibition. In conclusion, normal VEGFR2 function is required for vascular integrity and for Klotho to exert vascular protective and antifibrotic actions in the kidney partially through the regulation of VEGFR2 function.NEW & NOTEWORTHY This research paper studied the interplay of vascular endothelial growth factor receptor type 2 (VEGFR2), high dietary phosphate, and Klotho, an antiaging protein, in peritubular structure and kidney fibrosis. Klotho protein was shown to maintain VEGFR2 expression in the kidney and reduce high phosphate-induced cell injury. However, Klotho cytoprotection was attenuated by VEGFR2 inhibition. Thus, normal VEGFR2 function is required for vascular integrity and Klotho to exert vascular protective and antifibrotic actions in the kidney.
Collapse
Affiliation(s)
- Mingjun Shi
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas
| | - Jenny Omega Maique
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Dallas, Texas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
20
|
Bautch VL, Mukouyama YS. The Beauty and Complexity of Blood Vessel Patterning. Cold Spring Harb Perspect Med 2022; 12:a041167. [PMID: 35379659 PMCID: PMC9619359 DOI: 10.1101/cshperspect.a041167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
This review highlights new concepts in vascular patterning in the last 10 years, with emphasis on its beauty and complexity. Endothelial cell signaling pathways that respond to molecular or mechanical signals are described, and examples of vascular patterning that use these pathways in brain, skin, heart, and kidney are highlighted. The pathological consequences of patterning loss are discussed in the context of arteriovenous malformations (AVMs), and prospects for the next 10 years presented.
Collapse
Affiliation(s)
- Victoria L Bautch
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Development Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
21
|
Tobeiha M, Jafari A, Fadaei S, Mirazimi SMA, Dashti F, Amiri A, Khan H, Asemi Z, Reiter RJ, Hamblin MR, Mirzaei H. Evidence for the Benefits of Melatonin in Cardiovascular Disease. Front Cardiovasc Med 2022; 9:888319. [PMID: 35795371 PMCID: PMC9251346 DOI: 10.3389/fcvm.2022.888319] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/10/2022] [Indexed: 12/13/2022] Open
Abstract
The pineal gland is a neuroendocrine gland which produces melatonin, a neuroendocrine hormone with critical physiological roles in the circadian rhythm and sleep-wake cycle. Melatonin has been shown to possess anti-oxidant activity and neuroprotective properties. Numerous studies have shown that melatonin has significant functions in cardiovascular disease, and may have anti-aging properties. The ability of melatonin to decrease primary hypertension needs to be more extensively evaluated. Melatonin has shown significant benefits in reducing cardiac pathology, and preventing the death of cardiac muscle in response to ischemia-reperfusion in rodent species. Moreover, melatonin may also prevent the hypertrophy of the heart muscle under some circumstances, which in turn would lessen the development of heart failure. Several currently used conventional drugs show cardiotoxicity as an adverse effect. Recent rodent studies have shown that melatonin acts as an anti-oxidant and is effective in suppressing heart damage mediated by pharmacologic drugs. Therefore, melatonin has been shown to have cardioprotective activity in multiple animal and human studies. Herein, we summarize the most established benefits of melatonin in the cardiovascular system with a focus on the molecular mechanisms of action.
Collapse
Affiliation(s)
- Mohammad Tobeiha
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Ameneh Jafari
- Advanced Therapy Medicinal Product (ATMP) Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Fadaei
- Department of Internal Medicine and Endocrinology, Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Ali Mirazimi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Dashti
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Atefeh Amiri
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health. Long School of Medicine, San Antonio, TX, United States
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Johannesburg, South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
22
|
Eldeeb MA, Esmaili M, Hassan M, Ragheb MA. The Role of PTEN-L in Modulating PINK1-Parkin-Mediated Mitophagy. Neurotox Res 2022; 40:1103-1114. [PMID: 35699891 DOI: 10.1007/s12640-022-00475-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 12/27/2021] [Accepted: 01/13/2022] [Indexed: 11/24/2022]
Abstract
An inherent challenge that mitochondria face is the continuous exposure to diverse stresses which increase their likelihood of dysregulation. In response, human cells have evolved sophisticated quality control mechanisms to identify and eliminate abnormal dysfunctional mitochondria. One pivotal mitochondrial quality control pathway is PINK1/Parkin-dependent mitophagy which mediates the selective removal of the dysfunctional mitochondria from the cell by autophagy. PTEN-induced putative kinase 1 (PINK1) is a mitochondrial Ser/Thr kinase that was originally identified as a gene responsible for autosomal recessive early-onset Parkinson's disease (PD). Notably, upon failure of mitochondrial import, Parkin, another autosomal-recessive PD gene, is recruited to mitochondria and mediates the autophagic clearance of deregulated mitochondria. Importantly, recruitment of Parkin to damaged mitochondria hinges on the accumulation of PINK1 on the outer mitochondrial membrane (OMM). Normally, PINK1 is imported from the cytosol through the translocase of the outer membrane (TOM) complex, a large multimeric channel responsible for the import of most mitochondrial proteins. After import, PINK1 is rapidly degraded. Thus, at steady-state, PINK1 levels are kept low. However, upon mitochondrial import failure, PINK1 accumulates and forms a high-molecular weight > 700 kDa complex with TOM on the OMM. Thus, PINK1 functions as sensor, tagging dysfunctional mitochondria for Parkin-mediated mitophagy. Although much has been learned about the function of PINK1 in mitophagy, the biochemical and structural basis of negative regulation of PINK1 operation and functions is far from clear. Recent work unveiled new players as PTEN-l as negative regulator of PINK1 function. Herein, we review key aspects of mitophagy and PINK1/Parkin-mediated mitophagy with highlighting the role of negative regulation of PINK1 function and presenting some of the key future directions in PD cell biology.
Collapse
Affiliation(s)
- Mohamed A Eldeeb
- Department of Neurology and Neurosurgery, Montreal Neurological Institute McGill University, Montreal, QC, Canada. .,Department of Chemistry, Biochemistry Division, Cairo University, Giza, Egypt.
| | - Mansoore Esmaili
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Marwa Hassan
- Department of Chemistry, Biochemistry Division, Cairo University, Giza, Egypt
| | - Mohamed A Ragheb
- Department of Chemistry, Biochemistry Division, Cairo University, Giza, Egypt
| |
Collapse
|
23
|
Ahuja N, Hiltabidle MS, Rajasekhar H, Voss S, Lu SZ, Barlow HR, Cowdin MA, Daniel E, Vaddaraju V, Anandakumar T, Black E, Cleaver O, Maynard C. Endothelial Cyp26b1 restrains murine heart valve growth during development. Dev Biol 2022; 486:81-95. [DOI: 10.1016/j.ydbio.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 11/28/2022]
|
24
|
Ryan AR, Cleaver O. Plumbing our organs: Lessons from vascular development to instruct lab generated tissues. Curr Top Dev Biol 2022; 148:165-194. [DOI: 10.1016/bs.ctdb.2022.02.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
25
|
Donnan MD, Kenig-Kozlovsky Y, Quaggin SE. The lymphatics in kidney health and disease. Nat Rev Nephrol 2021; 17:655-675. [PMID: 34158633 DOI: 10.1038/s41581-021-00438-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 02/07/2023]
Abstract
The mammalian vascular system consists of two networks: the blood vascular system and the lymphatic vascular system. Throughout the body, the lymphatic system contributes to homeostatic mechanisms by draining extravasated interstitial fluid and facilitating the trafficking and activation of immune cells. In the kidney, lymphatic vessels exist mainly in the kidney cortex. In the medulla, the ascending vasa recta represent a hybrid lymphatic-like vessel that performs lymphatic-like roles in interstitial fluid reabsorption. Although the lymphatic network is mainly derived from the venous system, evidence supports the existence of lymphatic beds that are of non-venous origin. Following their development and maturation, lymphatic vessel density remains relatively stable; however, these vessels undergo dynamic functional changes to meet tissue demands. Additionally, new lymphatic growth, or lymphangiogenesis, can be induced by pathological conditions such as tissue injury, interstitial fluid overload, hyperglycaemia and inflammation. Lymphangiogenesis is also associated with conditions such as polycystic kidney disease, hypertension, ultrafiltration failure and transplant rejection. Although lymphangiogenesis has protective functions in clearing accumulated fluid and immune cells, the kidney lymphatics may also propagate an inflammatory feedback loop, exacerbating inflammation and fibrosis. Greater understanding of lymphatic biology, including the developmental origin and function of the lymphatics and their response to pathogenic stimuli, may aid the development of new therapeutic agents that target the lymphatic system.
Collapse
Affiliation(s)
- Michael D Donnan
- Feinberg Cardiovascular & Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Susan E Quaggin
- Feinberg Cardiovascular & Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Division of Nephrology & Hypertension, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
26
|
Ryan AR, England AR, Chaney CP, Cowdin MA, Hiltabidle M, Daniel E, Gupta AK, Oxburgh L, Carroll TJ, Cleaver O. Vascular deficiencies in renal organoids and ex vivo kidney organogenesis. Dev Biol 2021; 477:98-116. [PMID: 34000274 PMCID: PMC8382085 DOI: 10.1016/j.ydbio.2021.04.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 04/24/2021] [Accepted: 04/27/2021] [Indexed: 12/18/2022]
Abstract
Chronic kidney disease (CKD) and end stage renal disease (ESRD) are increasingly frequent and devastating conditions that have driven a surge in the need for kidney transplantation. A stark shortage of organs has fueled interest in generating viable replacement tissues ex vivo for transplantation. One promising approach has been self-organizing organoids, which mimic developmental processes and yield multicellular, organ-specific tissues. However, a recognized roadblock to this approach is that many organoid cell types fail to acquire full maturity and function. Here, we comprehensively assess the vasculature in two distinct kidney organoid models as well as in explanted embryonic kidneys. Using a variety of methods, we show that while organoids can develop a wide range of kidney cell types, as previously shown, endothelial cells (ECs) initially arise but then rapidly regress over time in culture. Vasculature of cultured embryonic kidneys exhibit similar regression. By contrast, engraftment of kidney organoids under the kidney capsule results in the formation of a stable, perfused vasculature that integrates into the organoid. This work demonstrates that kidney organoids offer a promising model system to define the complexities of vascular-nephron interactions, but the establishment and maintenance of a vascular network present unique challenges when grown ex vivo.
Collapse
Affiliation(s)
- Anne R Ryan
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alicia R England
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christopher P Chaney
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mitzy A Cowdin
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Max Hiltabidle
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Edward Daniel
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | - Thomas J Carroll
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
27
|
Li J, Geraldo LH, Dubrac A, Zarkada G, Eichmann A. Slit2-Robo Signaling Promotes Glomerular Vascularization and Nephron Development. J Am Soc Nephrol 2021; 32:2255-2272. [PMID: 34341180 PMCID: PMC8729857 DOI: 10.1681/asn.2020111640] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/22/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Kidney function requires continuous blood filtration by glomerular capillaries. Disruption of glomerular vascular development or maintenance contributes to the pathogenesis of kidney diseases, but the signaling events regulating renal endothelium development remain incompletely understood. Here, we discovered a novel role of Slit2-Robo signaling in glomerular vascularization. Slit2 is a secreted polypeptide that binds to transmembrane Robo receptors and regulates axon guidance as well as ureteric bud branching and angiogenesis. METHODS We performed Slit2-alkaline phosphatase binding to kidney cryosections from mice with or without tamoxifen-inducible Slit2 or Robo1 and -2 deletions, and we characterized the phenotypes using immunohistochemistry, electron microscopy, and functional intravenous dye perfusion analysis. RESULTS Only the glomerular endothelium, but no other renal endothelial compartment, responded to Slit2 in the developing kidney vasculature. Induced Slit2 gene deletion or Slit2 ligand trap at birth affected nephrogenesis and inhibited vascularization of developing glomeruli by reducing endothelial proliferation and migration, leading to defective cortical glomerular perfusion and abnormal podocyte differentiation. Global and endothelial-specific Robo deletion showed that both endothelial and epithelial Robo receptors contributed to glomerular vascularization. CONCLUSIONS Our study provides new insights into the signaling pathways involved in glomerular vascular development and identifies Slit2 as a potential tool to enhance glomerular angiogenesis.
Collapse
Affiliation(s)
- Jinyu Li
- Department of Cellular and Molecular Physiology, Yale University Medical School, New Haven, Connecticut
- Cardiovascular Research Center, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Luiz Henrique Geraldo
- Cardiovascular Research Center, Department of Internal Medicine, Yale University, New Haven, Connecticut
- Université de Paris, Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale U907, Paris, France
| | - Alexandre Dubrac
- Cardiovascular Research Center, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Georgia Zarkada
- Cardiovascular Research Center, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Anne Eichmann
- Department of Cellular and Molecular Physiology, Yale University Medical School, New Haven, Connecticut
- Cardiovascular Research Center, Department of Internal Medicine, Yale University, New Haven, Connecticut
- Université de Paris, Paris Cardiovascular Research Center, Institut National de la Santé et de la Recherche Médicale U907, Paris, France
| |
Collapse
|
28
|
Little MH. Returning to kidney development to deliver synthetic kidneys. Dev Biol 2021; 474:22-36. [PMID: 33333068 PMCID: PMC8052282 DOI: 10.1016/j.ydbio.2020.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/09/2020] [Indexed: 12/27/2022]
Abstract
There is no doubt that the development of transplantable synthetic kidneys could improve the outcome for the many millions of people worldwide suffering from chronic kidney disease. Substantial progress has been made in the last 6 years in the generation of kidney tissue from stem cells. However, the limited scale, incomplete cellular complexity and functional immaturity of such structures suggests we are some way from this goal. While developmental biology has successfully guided advances to date, these human kidney models are limited in their capacity for ongoing nephrogenesis and lack corticomedullary definition, a unified vasculature and a coordinated exit path for urinary filtrate. This review will reassess our developmental understanding of how the mammalian embryo manages to create kidneys, how this has informed our progress to date and how both engineering and developmental biology can continue to guide us towards a synthetic kidney.
Collapse
Affiliation(s)
- Melissa H Little
- Murdoch Children's Research Institute, Flemington Rd, Parkville, VIC, Australia; Department of Paediatrics, The University of Melbourne, VIC, Australia; Department of Anatomy and Neuroscience, The University of Melbourne, VIC, Australia.
| |
Collapse
|
29
|
Abstract
The kidney plays an integral role in filtering the blood-removing metabolic by-products from the body and regulating blood pressure. This requires the establishment of large numbers of efficient and specialized blood filtering units (nephrons) that incorporate a system for vascular exchange and nutrient reabsorption as well as a collecting duct system to remove waste (urine) from the body. Kidney development is a dynamic process which generates these structures through a delicately balanced program of self-renewal and commitment of nephron progenitor cells that inhabit a constantly evolving cellular niche at the tips of a branching ureteric "tree." The former cells build the nephrons and the latter the collecting duct system. Maintaining these processes across fetal development is critical for establishing the normal "endowment" of nephrons in the kidney and perturbations to this process are associated both with mutations in integral genes and with alterations to the fetal environment.
Collapse
Affiliation(s)
- Ian M Smyth
- Department of Anatomy and Developmental Biology, Department of Biochemistry and Molecular Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
30
|
Holloway EM, Wu JH, Czerwinski M, Sweet CW, Wu A, Tsai YH, Huang S, Stoddard AE, Capeling MM, Glass I, Spence JR. Differentiation of Human Intestinal Organoids with Endogenous Vascular Endothelial Cells. Dev Cell 2020; 54:516-528.e7. [PMID: 32841595 PMCID: PMC7480827 DOI: 10.1016/j.devcel.2020.07.023] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/11/2020] [Accepted: 07/29/2020] [Indexed: 12/17/2022]
Abstract
Human pluripotent stem cell (hPSC)-derived intestinal organoids (HIOs) lack some cellular populations found in the native organ, including vasculature. Using single-cell RNA sequencing (scRNA-seq), we have identified a population of endothelial cells (ECs) present early in HIO differentiation that declines over time in culture. Here, we developed a method to expand and maintain this endogenous population of ECs within HIOs (vHIOs). Given that ECs possess organ-specific gene expression, morphology, and function, we used bulk RNA-seq and scRNA-seq to interrogate the developing human intestine, lung, and kidney in order to identify organ-enriched EC gene signatures. By comparing these gene signatures and validated markers to HIO ECs, we find that HIO ECs grown in vitro share the highest similarity with native intestinal ECs relative to kidney and lung. Together, these data demonstrate that HIOs can co-differentiate a native EC population that is properly patterned with an intestine-specific EC transcriptional signature in vitro.
Collapse
Affiliation(s)
- Emily M Holloway
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Joshua H Wu
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Michael Czerwinski
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Caden W Sweet
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Angeline Wu
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Yu-Hwai Tsai
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sha Huang
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Amy E Stoddard
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA
| | - Meghan M Capeling
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA
| | - Ian Glass
- Department of Pediatrics, Genetic Medicine, University of Washington, Seattle, WA 98195, USA
| | - Jason R Spence
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA.
| |
Collapse
|
31
|
Li P, Wang J, Zhao X, Ru J, Tian T, An Y, Tang L, Bai Y. PTEN inhibition attenuates endothelial cell apoptosis in coronary heart disease via modulating the AMPK-CREB-Mfn2-mitophagy signaling pathway. J Cell Physiol 2020; 235:4878-4889. [PMID: 31654396 DOI: 10.1002/jcp.29366] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 10/07/2019] [Indexed: 12/11/2022]
Abstract
Atherosclerosis (AS) is a major pathogenic factor in patients with cardiovascular diseases, and endothelial dysfunction (ED) plays a primary role in the occurrence and development of AS. In our study, we attempted to evaluate the role of phosphatase and tensin homolog (PTEN) in endothelial cell apoptosis under oxidized low-density lipoprotein (ox-LDL) stimulation and identify the associated mechanisms. The results of our study demonstrated that ox-LDL induced human umbilical vein endothelial cell (HUVEC) death via mitochondrial apoptosis, as evidenced by reduced mitochondrial potential, increased mitochondria permeability transition pore opening, cellular calcium overload, and caspase-9/-3 activation. In addition, ox-LDL also suppressed cellular energy production via downregulating the mitochondrial respiratory complex. Moreover, ox-LDL impaired HUVECs migration. Western blot analysis showed that PTEN expression was upregulated after exposure to ox-LDL and knockdown of PTEN could attenuate ox-LDL-mediated endothelial cell damage. Furthermore, we found that ox-LDL impaired mitophagy activity, whereas PTEN deletion could improve mitophagic flux and this effect relied on the activity of the AMP-activated protein kinase (AMPK)-cAMP-response element-binding protein (CREB)-Mitofusin-2 (Mfn2) axis. When the AMPK-CREB-Mfn2 pathway was inhibited, PTEN deletion-associated HUVECs protection was significantly reduced, suggesting that the AMPK-CREB-Mfn2-mitophagy axis is required for PTEN deletion-mediated endothelial cell survival under ox-LDL. Taken together, our results indicate that ox-LDL-induced endothelial cell damage is associated with PTEN overexpression, and inhibition of PTEN could promote endothelial survival via activating the AMPK-CREB-Mfn2-mitophagy signaling pathway.
Collapse
Affiliation(s)
- Pei Li
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Jing Wang
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xia Zhao
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Jing Ru
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Tian Tian
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yun An
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Liying Tang
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yuzhi Bai
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
32
|
Hu C, Lu K, Liu W. Exendin-4 attenuates inflammation-mediated endothelial cell apoptosis in varicose veins through inhibiting the MAPK-JNK signaling pathway. J Recept Signal Transduct Res 2020; 40:464-470. [PMID: 32338116 DOI: 10.1080/10799893.2020.1756326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Context: Inflammation response has been found to be associated with endothelial cell death in the progression of varicose veins. Exendin-4 is able to reduce inflammation and thus attenuate cell apoptosis.Aim: The aim of our study is to explore the influence of Exendin-4 on LPS-treated endothelial cells.Methods: Cells were treated with LPS. Exendin-4 was added into the medium of cells. Western blots, qPCR, and ELISA were used to analyze the role of Exendin-4 in LPS-mediated cell death.Results: We found that LPS treatment caused significantly cell death. Whereas this trend could be attenuated by Exendin-4. After treatment with Exendin-4, inflammation factors upregulation and oxidative stress activation were significantly repressed, an effect that was followed by a drop in the levels of glucose production and lactic acid generation. At the molecular levels, Exendin-4 treatment inhibited the activity of MAPK-JNK signaling pathway in the presence of LPS treatment.Conclusions: LPS causes cell apoptosis through inducing inflammation response, oxidative stress and energy stress. Exendin-4 treatment enhances cell survival, reduces inflammation, and improves energy stress through inhibiting the MAPK-JNK signaling pathway.
Collapse
Affiliation(s)
- Changfu Hu
- Shenzhen University General Hospital, Shenzhen, China
| | - Kai Lu
- Daqing Oilfield General Hospital, Daqing, China
| | - Weili Liu
- Daqing Oilfield General Hospital, Daqing, China
| |
Collapse
|
33
|
Jafree DJ, Long DA. Beyond a Passive Conduit: Implications of Lymphatic Biology for Kidney Diseases. J Am Soc Nephrol 2020; 31:1178-1190. [PMID: 32295825 DOI: 10.1681/asn.2019121320] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The kidney contains a network of lymphatic vessels that clear fluid, small molecules, and cells from the renal interstitium. Through modulating immune responses and via crosstalk with surrounding renal cells, lymphatic vessels have been implicated in the progression and maintenance of kidney disease. In this Review, we provide an overview of the development, structure, and function of lymphatic vessels in the healthy adult kidney. We then highlight the contributions of lymphatic vessels to multiple forms of renal pathology, emphasizing CKD, transplant rejection, and polycystic kidney disease and discuss strategies to target renal lymphatics using genetic and pharmacologic approaches. Overall, we argue the case for lymphatics playing a fundamental role in renal physiology and pathology and treatments modulating these vessels having therapeutic potential across the spectrum of kidney disease.
Collapse
Affiliation(s)
- Daniyal J Jafree
- Developmental Biology and Cancer Programme, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.,MB/PhD Programme, Faculty of Medical Sciences, University College London, London, United Kingdom
| | - David A Long
- Developmental Biology and Cancer Programme, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
34
|
Lu K, Zhao J, Liu W. Macrophage stimulating 1-induced inflammation response promotes aortic aneurysm formation through triggering endothelial cells death and activating the NF-κB signaling pathway. J Recept Signal Transduct Res 2020; 40:374-382. [PMID: 32156191 DOI: 10.1080/10799893.2020.1738484] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aortic aneurysm formation is associated with endothelial cells dysfunction through an undefined mechanism. Macrophage stimulating 1 (Mst1) and NF-κB signaling pathway have been found to be related to inflammation response in endothelial cell damage. The goal of our study is to explore the role of Mst1 in regulating endothelial cell viability with a focus on NF-κB signaling pathway and inflammation response. Endothelial cell viability and death were determined via immunofluorescence and ELISA. Agonist of NF-κB signaling pathway and siRNA against Mst1 were used. The results in our study demonstrated that Mst1 transcription and expression were significantly elevated after exposure to oxidative stress in endothelial cells. Once loss of Mst1 through transfection of siRNA (si-Mst1), endothelial cell viability and survival rate were rapidly increased in response to oxidative stress. In addition, we also found that Mst1 controlled inflammation response and mitochondrial function in endothelial cells. Re-activation of NF-κB signaling pathway was followed by an activation of inflammation response and mitochondrial dysfunction, as evidenced by increased expression of inflammation factors and decreased ATP synthesis. Altogether, our results identify Mst1 as the primary factors responsible for endothelial cells dysfunction in aneurysms formation through inducing inflammation response, endothelial apoptosis, and NF-κB signaling pathway activation.
Collapse
Affiliation(s)
- Kai Lu
- Daqing Oilfield General Hospital, Daqing, P. R. China
| | - Jianfei Zhao
- Daqing Oilfield General Hospital, Daqing, P. R. China
| | - Weili Liu
- Daqing Oilfield General Hospital, Daqing, P. R. China
| |
Collapse
|
35
|
Daniel E, Barlow HR, Sutton GI, Gu X, Htike Y, Cowdin MA, Cleaver O. Cyp26b1 is an essential regulator of distal airway epithelial differentiation during lung development. Development 2020; 147:dev181560. [PMID: 32001436 PMCID: PMC7044453 DOI: 10.1242/dev.181560] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 01/23/2020] [Indexed: 12/16/2022]
Abstract
Proper organ development depends on coordinated communication between multiple cell types. Retinoic acid (RA) is an autocrine and paracrine signaling molecule essential for the development of most organs, including the lung. Despite extensive work detailing effects of RA deficiency in early lung morphogenesis, little is known about how RA regulates late gestational lung maturation. Here, we investigate the role of the RA catabolizing protein Cyp26b1 in the lung. Cyp26b1 is highly enriched in lung endothelial cells (ECs) throughout development. We find that loss of Cyp26b1 leads to reduction of alveolar type 1 cells, failure of alveolar inflation and early postnatal lethality in mouse. Furthermore, we observe expansion of distal epithelial progenitors, but no appreciable changes in proximal airways, ECs or stromal populations. Exogenous administration of RA during late gestation partially mimics these defects; however, transcriptional analyses comparing Cyp26b1-/- with RA-treated lungs reveal overlapping, but distinct, responses. These data suggest that defects observed in Cyp26b1-/- lungs are caused by both RA-dependent and RA-independent mechanisms. This work reports crucial cellular crosstalk during lung development involving Cyp26b1-expressing endothelium and identifies a novel RA modulator in lung development.
Collapse
Affiliation(s)
- Edward Daniel
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Haley R Barlow
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Gabrielle I Sutton
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaowu Gu
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yadanar Htike
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mitzy A Cowdin
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ondine Cleaver
- Department of Molecular Biology and Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
36
|
Tian Y, Song H, Jin D, Hu N, Sun L. MST1-Hippo pathway regulates inflammation response following myocardial infarction through inhibiting HO-1 signaling pathway. J Recept Signal Transduct Res 2020; 40:231-236. [PMID: 32054389 DOI: 10.1080/10799893.2020.1726954] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Context: Mammalian STE20-like protein kinases 1 (MST1) has been found to be associated with cardiomyocyte damage following acute myocardial infarction.Aim: The aim of our study is to explore the influence of MST1 in inflammation response following myocardial infarction.Methods: Cardiomyocyte cell line was used in vitro with hypoxia treatment to establish myocardial infarction model. ELISA, qPCR, Western blots, and siRNA technology were used to analyze the role of MST1 in inflammation response following myocardial infarction.Results: The transcription and expression of MST1 was significantly elevated following myocardial infarction. Loss of MST1 attenuated the levels of inflammation response and thus contributed to the survival of cardiomyocyte in vitro. Mechanistically, MST1 deletion reversed the activity of heme oxygenase-1 (HO-1) and thus reduced hypoxia-mediated cardiomyocyte death.Conclusions: Altogether, in this study, we found that MST1-Hippo pathway is activated in myocardial infarction and contributes to the inflammation response in cardiomyocytes through inhibiting the HO-1 signaling pathway. This finding would provide a potential target to reverse cardiomyocyte viability and reduce inflammation response in myocardial infarction.
Collapse
Affiliation(s)
- Yanan Tian
- Department of Cardiology, The Affiliated Hospital of Chengde Medical College, Chengde, P. R. China
| | - Haijiu Song
- The First Department of Medicine, Chengde City Hospital of traditional Chinese Medicine, Chengde, P. R. China
| | - Dapeng Jin
- Department of Cardiology, The Affiliated Hospital of Chengde Medical College, Chengde, P. R. China
| | - Na Hu
- Department of Cardiology, The Affiliated Hospital of Chengde Medical College, Chengde, P. R. China
| | - Lixian Sun
- Department of Cardiology, The Affiliated Hospital of Chengde Medical College, Chengde, P. R. China
| |
Collapse
|
37
|
Nishikawa M, Sakai Y, Yanagawa N. Design and strategy for manufacturing kidney organoids. Biodes Manuf 2020. [DOI: 10.1007/s42242-020-00060-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
38
|
Shang X, Zhang Y, Xu J, Li M, Wang X, Yu R. SRV2 promotes mitochondrial fission and Mst1-Drp1 signaling in LPS-induced septic cardiomyopathy. Aging (Albany NY) 2020; 12:1417-1432. [PMID: 31951593 PMCID: PMC7053598 DOI: 10.18632/aging.102691] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022]
Abstract
Mitochondrial fission is associated with cardiomyocyte death and myocardial depression, and suppressor of ras val-2 (SRV2) is a newly discovered pro-fission protein. In this study, we examined the mechanisms of SRV2-mediated mitochondrial fission in septic cardiomyopathy. Western blotting, ELISA, and immunofluorescence were used to evaluate mitochondrial function, oxidative balance, energy metabolism and caspase-related death, and siRNA and adenoviruses were used to perform loss- and gain-of-function assays. Our results demonstrated that increased SRV2 expression promotes, while SRV2 knockdown attenuates, cardiomyocyte death in LPS-induced septic cardiomyopathy. Mechanistically, SRV2 activation promoted mitochondrial fission and physiological abnormalities by upregulating oxidative injury, ATP depletion, and caspase-9-related apoptosis. Our results also demonstrated that SRV2 promotes mitochondrial fission via a Mst1-Drp1 axis. SRV2 knockdown decreased Mst1 and Drp1 levels, while Mst1 overexpression abolished the mitochondrial protection and cardiomyocyte survival-promoting effects of SRV2 knockdown. SRV2 is thus a key novel promotor of mitochondrial fission and Mst1-Drp1 axis activity in septic cardiomyopathy.
Collapse
Affiliation(s)
- Xiuling Shang
- Department of Critical Care Medicine, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Medical University, Fuzhou, Fujian 350001, China
| | - Yingrui Zhang
- Department of Critical Care Medicine, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Medical University, Fuzhou, Fujian 350001, China
| | - Jingqing Xu
- Department of Critical Care Medicine, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Medical University, Fuzhou, Fujian 350001, China
| | - Min Li
- Department of Critical Care Medicine, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Medical University, Fuzhou, Fujian 350001, China
| | - Xiaoting Wang
- Department of Critical Care Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing 100730, China
| | - Rongguo Yu
- Department of Critical Care Medicine, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Medical University, Fuzhou, Fujian 350001, China
| |
Collapse
|
39
|
Gao J, Wang H, Li Y, Li W. Resveratrol attenuates cerebral ischaemia reperfusion injury via modulating mitochondrial dynamics homeostasis and activating AMPK-Mfn1 pathway. Int J Exp Pathol 2019; 100:337-349. [PMID: 31867811 DOI: 10.1111/iep.12336] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/04/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022] Open
Abstract
The pathogenesis of cerebral ischaemia reperfusion injury (IRI) has not been fully described. Accordingly, there is little effective drug available for the treatment of cerebral IRI. The aim of our study was to explore the exact role played by Mfn1-mediated mitochondrial protection in cerebral IRI and evaluate the beneficial action of resveratrol on reperfused brain. Our study demonstrated that hypoxia-reoxygenation (HR) injury caused N2a cell apoptosis and this process was highly affected by mitochondrial dysfunction. Decreased mitochondrial membrane potential, increased mitochondrial oxidative stress, and an activated mitochondrial apoptosis pathway were noted in HR-treated N2a cells. Interestingly, resveratrol treatment could attenuate N2a cell apoptosis via sustaining mitochondrial homeostasis. Further, we found that resveratrol modulated mitochondrial performance via activating the Mfn1-related mitochondrial protective system. Knockdown of Mfn1 could abolish the beneficial effects of resveratrol on HR-treated N2a cells. Besides, we also report that resveratrol regulated Mfn1 expression via the AMPK pathway; inhibition of AMPK pathway also neutralized the anti-apoptotic effect of resveratrol on N2a cells in the setting of cerebral IRI. Taken together our results show that mitochondrial damage is closely associated with the progression of cerebral IRI. In addition we also demonstrate the protective action played by resveratrol on reperfused brain and show that this effect is achieved via activating the AMPK-Mfn1 pathway.
Collapse
Affiliation(s)
- Jinbao Gao
- Department of Neurosurgery, the Seventh Medical Center, the PLA Army General Hospital, Beijing, China
| | - Haijiang Wang
- Department of Neurosurgery, the Seventh Medical Center, the PLA Army General Hospital, Beijing, China
| | - Yunjun Li
- Department of Neurosurgery, the Seventh Medical Center, the PLA Army General Hospital, Beijing, China
| | - Wende Li
- Department of Neurosurgery, the Seventh Medical Center, the PLA Army General Hospital, Beijing, China
| |
Collapse
|
40
|
Barry DM, McMillan EA, Kunar B, Lis R, Zhang T, Lu T, Daniel E, Yokoyama M, Gomez-Salinero JM, Sureshbabu A, Cleaver O, Di Lorenzo A, Choi ME, Xiang J, Redmond D, Rabbany SY, Muthukumar T, Rafii S. Molecular determinants of nephron vascular specialization in the kidney. Nat Commun 2019; 10:5705. [PMID: 31836710 PMCID: PMC6910926 DOI: 10.1038/s41467-019-12872-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 09/22/2019] [Indexed: 12/13/2022] Open
Abstract
Although kidney parenchymal tissue can be generated in vitro, reconstructing the complex vasculature of the kidney remains a daunting task. The molecular pathways that specify and sustain functional, phenotypic and structural heterogeneity of the kidney vasculature are unknown. Here, we employ high-throughput bulk and single-cell RNA sequencing of the non-lymphatic endothelial cells (ECs) of the kidney to identify the molecular pathways that dictate vascular zonation from embryos to adulthood. We show that the kidney manifests vascular-specific signatures expressing defined transcription factors, ion channels, solute transporters, and angiocrine factors choreographing kidney functions. Notably, the ontology of the glomerulus coincides with induction of unique transcription factors, including Tbx3, Gata5, Prdm1, and Pbx1. Deletion of Tbx3 in ECs results in glomerular hypoplasia, microaneurysms and regressed fenestrations leading to fibrosis in subsets of glomeruli. Deciphering the molecular determinants of kidney vascular signatures lays the foundation for rebuilding nephrons and uncovering the pathogenesis of kidney disorders. The kidney is vascularized with highly specialized and zonated endothelial cells that are essential for its filtration function. Here, Barry et al. provide a single-cell RNA sequencing analysis of the kidney vasculature that highlights its transcriptional heterogeneity and uncovers pathways important for its development and function.
Collapse
Affiliation(s)
- David M Barry
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA.
| | - Elizabeth A McMillan
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Balvir Kunar
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Raphael Lis
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Tuo Zhang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Tyler Lu
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Edward Daniel
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Masataka Yokoyama
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Jesus M Gomez-Salinero
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Angara Sureshbabu
- Division of Nephrology and Hypertension, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Annarita Di Lorenzo
- Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Mary E Choi
- Division of Nephrology and Hypertension, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Jenny Xiang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY, 10065, USA
| | - David Redmond
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Sina Y Rabbany
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA.,Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, Hempstead, NY, 11549, USA
| | - Thangamani Muthukumar
- Division of Nephrology and Hypertension, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Shahin Rafii
- Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
41
|
Jafree DJ, Moulding D, Kolatsi-Joannou M, Perretta Tejedor N, Price KL, Milmoe NJ, Walsh CL, Correra RM, Winyard PJ, Harris PC, Ruhrberg C, Walker-Samuel S, Riley PR, Woolf AS, Scambler PJ, Long DA. Spatiotemporal dynamics and heterogeneity of renal lymphatics in mammalian development and cystic kidney disease. eLife 2019; 8:48183. [PMID: 31808745 PMCID: PMC6948954 DOI: 10.7554/elife.48183] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 11/30/2019] [Indexed: 12/11/2022] Open
Abstract
Heterogeneity of lymphatic vessels during embryogenesis is critical for organ-specific lymphatic function. Little is known about lymphatics in the developing kidney, despite their established roles in pathology of the mature organ. We performed three-dimensional imaging to characterize lymphatic vessel formation in the mammalian embryonic kidney at single-cell resolution. In mouse, we visually and quantitatively assessed the development of kidney lymphatic vessels, remodeling from a ring-like anastomosis under the nascent renal pelvis; a site of VEGF-C expression, to form a patent vascular plexus. We identified a heterogenous population of lymphatic endothelial cell clusters in mouse and human embryonic kidneys. Exogenous VEGF-C expanded the lymphatic population in explanted mouse embryonic kidneys. Finally, we characterized complex kidney lymphatic abnormalities in a genetic mouse model of polycystic kidney disease. Our study provides novel insights into the development of kidney lymphatic vasculature; a system which likely has fundamental roles in renal development, physiology and disease. In most organs in the body, fluid tends to build up in the spaces between cells, especially if the organs become inflamed. Each organ has a ‘waste disposal system’; a set of specialized tubes called lymphatic vessels, to clear away this excess fluid and keep a check on inflammation. Defects in these tubes have been linked to a wide range of diseases including heart attacks, obesity, dementia and cancer. The kidneys are responsible for filtering blood and balancing many of the body’s chemical processes. Polycystic kidney disease (PKD) is the most common genetic kidney disorder and it results in cysts filled with fluid building up in the kidney. The growth of cysts in PKD may be due to a problem with the lymphatic vessels. However, compared to other organs, how lymphatic vessels first form within the kidney and what they do is not well understood. Now, Jafree et al. have used three-dimensional imaging to study how lymphatic vessels form in the kidneys of mice and humans. The experiments showed that lymphatic vessels first appear when mouse kidneys are about half developed, and start to grow rapidly when the kidneys are thought to begin filtering blood. Clusters of cells that may help lymphatic vessels to grow were also found hidden deep within the kidneys of mouse embryos. Treating the kidneys with a factor that stimulates the growth of lymphatic vessels increased the numbers of these clusters. Jafree et al. found similar clusters of cells in human kidneys, suggesting that lymphatic vessels in the kidneys of different mammals may develop in the same way. Further experiments showed that the lymphatic vessels of kidneys in mice with PKD become distorted early on in the disease, when cysts are still small and before the mice develop symptoms. In the future, identifying drugs that target kidney lymphatic vessels may lead to more effective treatments for patients with PKD and other kidney diseases.
Collapse
Affiliation(s)
- Daniyal J Jafree
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.,MB/PhD Programme, Faculty of Medical Sciences, University College London, London, United Kingdom
| | - Dale Moulding
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Maria Kolatsi-Joannou
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Nuria Perretta Tejedor
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Karen L Price
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Natalie J Milmoe
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Claire L Walsh
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, United Kingdom
| | - Rosa Maria Correra
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Paul Jd Winyard
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, United States
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Simon Walker-Samuel
- Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, United Kingdom
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Adrian S Woolf
- School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom.,Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Peter J Scambler
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - David A Long
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
42
|
Ma C, Wang J, Fan L. Therapeutic effects of bone mesenchymal stem cells on oral and maxillofacial defects: a novel signaling pathway involving miR-31/CXCR4/Akt axis. J Recept Signal Transduct Res 2019; 39:321-330. [PMID: 31573375 DOI: 10.1080/10799893.2019.1669054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Context: Although bone mesenchymal stem cells (BMSCs) have been used for the treatment of oral and maxillofacial defects, the survival rate and limited proliferation reduces the therapeutic efficiency of BMSC.Objective: The aim of our study is to explore the role of miR-31 in regulating survival, proliferation, and migration of BMSC in vitro.Materials and methods: LPS was used in vitro to induce BMSC damage and then miR-31 was used to incubate with BMSC. Subsequently, BMSC proliferation, survival, and migration were determined via ELISA, qPCR, western blots, and immunofluorescence.Results: The expression of miR-31 was downregulated in response to LPS stress. Interestingly, supplementation of miR-31 could reverse the survival, proliferation and migration of BMSC under LPS. Mechanically, miR-31 treatment inhibited the activation of caspase, and thus promoted BMSC survival. Besides, miR-31 upregulated the genes related to cell proliferation, an effect that was followed by an increase in the levels of migratory factors. Further, we found that miR-31 treatment activated the CXCR4/Akt pathway and blockade of CXCR4/Akt could abolish the beneficial effects of miR-31 on BMSC proliferation, survival, and migration.Conclusions: miR-31 could increase the therapeutic efficiency of BMSC via the CXCR4/Akt pathway.
Collapse
Affiliation(s)
- Chao Ma
- Department of Oral and Maxillofacial Surgery, Cangzhou Central Hospital of Hebei Province, Cangzhou, China
| | - Jingxian Wang
- Department of Oral and Maxillofacial Surgery, Cangzhou Central Hospital of Hebei Province, Cangzhou, China
| | - Longkun Fan
- Department of Oral and Maxillofacial Surgery, Cangzhou Central Hospital of Hebei Province, Cangzhou, China
| |
Collapse
|
43
|
Abstract
There are now many reports of human kidney organoids generated via the directed differentiation of human pluripotent stem cells (PSCs) based on an existing understanding of mammalian kidney organogenesis. Such kidney organoids potentially represent tractable tools for the study of normal human development and disease with improvements in scale, structure, and functional maturation potentially providing future options for renal regeneration. The utility of such organotypic models, however, will ultimately be determined by their developmental accuracy. While initially inferred from mouse models, recent transcriptional analyses of human fetal kidney have provided greater insight into nephrogenesis. In this review, we discuss how well human kidney organoids model the human fetal kidney and how the remaining differences challenge their utility.
Collapse
Affiliation(s)
- Melissa H Little
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3052, Australia
- Department of Paediatrics, The University of Melbourne, Victoria 3052, Australia
| | - Alexander N Combes
- Murdoch Children's Research Institute, Parkville, Victoria 3052, Australia
- Department of Anatomy and Neuroscience, The University of Melbourne, Victoria 3052, Australia
| |
Collapse
|
44
|
Yamanaka S, Saito Y, Fujimoto T, Takamura T, Tajiri S, Matsumoto K, Yokoo T. Kidney Regeneration in Later-Stage Mouse Embryos via Transplanted Renal Progenitor Cells. J Am Soc Nephrol 2019; 30:2293-2305. [PMID: 31548350 DOI: 10.1681/asn.2019020148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/12/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The limited availability of donor kidneys for transplantation has spurred interest in investigating alternative strategies, such as regenerating organs from stem cells transplanted into animal embryos. However, there is no known method for transplanting cells into later-stage embryos, which may be the most suitable host stages for organogenesis, particularly into regions useful for kidney regeneration. METHODS We demonstrated accurate transplantation of renal progenitor cells expressing green fluorescent protein to the fetal kidney development area by incising the opaque uterine muscle layer but not the transparent amniotic membrane. We allowed renal progenitor cell-transplanted fetuses to develop for 6 days postoperatively before removal for analysis. We also transplanted renal progenitor cells into conditional kidney-deficient mouse embryos. We determined growth and differentiation of transplanted cells in all cases. RESULTS Renal progenitor cell transplantation into the retroperitoneal cavity of fetuses at E13-E14 produced transplant-derived, vascularized glomeruli with filtration function and did not affect fetal growth or survival. Cells transplanted to the nephrogenic zone produced a chimera in the cap mesenchyme of donor and host nephron progenitor cells. Renal progenitor cells transplanted to conditional kidney-deficient fetuses induced the formation of a new nephron in the fetus that is connected to the host ureteric bud. CONCLUSIONS We developed a cell transplantation method for midstage to late-stage fetuses. In vivo kidney regeneration from renal progenitor cells using the renal developmental environment of the fetus shows promise. Our findings suggest that fetal transplantation methods may contribute to organ regeneration and developmental research.
Collapse
Affiliation(s)
- Shuichiro Yamanaka
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yatsumu Saito
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Toshinari Fujimoto
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Tsuyoshi Takamura
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Susumu Tajiri
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kei Matsumoto
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
45
|
Zhong J, Tan Y, Lu J, Liu J, Xiao X, Zhu P, Chen S, Zheng S, Chen Y, Hu Y, Guo Z. Therapeutic contribution of melatonin to the treatment of septic cardiomyopathy: A novel mechanism linking Ripk3-modified mitochondrial performance and endoplasmic reticulum function. Redox Biol 2019; 26:101287. [PMID: 31386965 PMCID: PMC6692063 DOI: 10.1016/j.redox.2019.101287] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 07/09/2019] [Accepted: 07/26/2019] [Indexed: 12/30/2022] Open
Abstract
The basic pathophysiological mechanisms underlying septic cardiomyopathy have not yet been completely clarified. Disease-specific treatments are lacking, and care is still based on supportive modalities. The aim of our study was to assess the protective effects of melatonin on septic cardiomyopathy, with a focus on the interactions between receptor-interacting protein kinase 3 (Ripk3), the mitochondria, endoplasmic reticulum (ER) and cytoskeletal degradation in cardiomyocytes. Ripk3 expression was increased in heart samples challenged with LPS, followed by myocardial inflammation, cardiac dysfunction, myocardial breakdown and cardiomyocyte death. The melatonin treatment attenuated septic myocardial injury in a comparable manner to the genetic depletion of Ripk3. Molecular investigations revealed that Ripk3 intimately regulated mitochondrial function, ER stress, cytoskeletal homeostasis and cardioprotective signaling pathways. Melatonin-mediated inhibition of Ripk3 improved mitochondrial bioenergetics, reduced mitochondria-initiated oxidative damage, sustained mitochondrial dynamics, ameliorated ER stress, normalized calcium recycling, and activated cardioprotective signaling pathways (including AKT, ERK and AMPK) in cardiomyocytes. Interestingly, Ripk3 overexpression mediated resistance to melatonin therapy following the infection of LPS-treated hearts with an adenovirus expressing Ripk3. Altogether, our findings identify Ripk3 upregulation as a novel risk factor for the development of sepsis-related myocardial injury, and melatonin restores the physiological functions of the mitochondria, ER, contractile cytoskeleton and cardioprotective signaling pathways. Additionally, our data also reveal a new, potentially therapeutic mechanism by which melatonin protects the heart from sepsis-mediated dysfunction, possibly by targeting Ripk3.
Collapse
Affiliation(s)
- Jiankai Zhong
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528308, Guangdong, China
| | - Ying Tan
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Jianhua Lu
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528308, Guangdong, China
| | - Jichen Liu
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaochan Xiao
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Pinji Zhu
- Medical School of Chinese PLA, PLA General Hospital, Beijing, 100853, China
| | - Sainan Chen
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Sulin Zheng
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528308, Guangdong, China
| | - Yuying Chen
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528308, Guangdong, China
| | - Yunzhao Hu
- Department of Cardiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde Foshan), Foshan, 528308, Guangdong, China
| | - Zhigang Guo
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
46
|
Song X, Li T. Ripk3 mediates cardiomyocyte necrosis through targeting mitochondria and the JNK-Bnip3 pathway under hypoxia-reoxygenation injury. J Recept Signal Transduct Res 2019; 39:331-340. [PMID: 31658855 DOI: 10.1080/10799893.2019.1676259] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/29/2019] [Accepted: 10/01/2019] [Indexed: 12/13/2022]
Abstract
Context: Cardiomyocyte necrosis following myocardial infarction drastically the progression of heart failure.Objective: In the current study, we explored the upstream mediator for cardiomyocytes necrosis induced by hypoxia-reoxygenation (HR) injury with a focus on mitochondrial function and JNK-Bnip3 pathway.Materials and methods: Cell necrosis was determined via MTT assay, TUNEL staining and PI staining. siRNA transfection was performed to inhibit Ripk3 activation in response to HR injury. Pathway blocker was applied to prevent JNK activation.Results: Ripk3 was rapidly increased in HR-treated cardiomyocytes and correlated with the necrosis of cardiomyocytes. Interestingly, silencing of Ripk3 attenuated HR-mediated cardiomyocytes necrosis. At the molecular levels, Ripk3 deletion sustained mitochondrial bioenergetics and stabilized mitochondrial glucose metabolism. Besides, Ripk3 deletion also reduced mitochondrial oxidative stress and inhibited mPTP opening. To the end, we found Ripk3 activation was along with JNK pathway activation and Bnip3 upregulation. Interestingly, blockade of JNK pathway abolished the harmful effects of HR injury on mitochondrial function, energy metabolism and redox balance. Moreover, overexpression of Bnip3 abrogated the protection action played by Ripk3 deletion on cardiomyocytes survival.Conclusions: Taken together, these data may identify Ripk3 upregulation, mitochondrial dysfunction and JNK-Bnip3 axis activation as the novel mechanisms underlying cardiomyocytes necrosis achieved by HR injury. Thereby, approaches targeted to the Ripk3-JNK-Bnip3-mitochondria cascade have the potential to ameliorate the progression of HR-related cardiomyocytes necrosis in the clinical practice.
Collapse
Affiliation(s)
- Xinyu Song
- Department of Cardiology, Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Tianchang Li
- Department of Cardiology, Sixth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
47
|
Braitsch CM, Azizoglu DB, Htike Y, Barlow HR, Schnell U, Chaney CP, Carroll TJ, Stanger BZ, Cleaver O. LATS1/2 suppress NFκB and aberrant EMT initiation to permit pancreatic progenitor differentiation. PLoS Biol 2019; 17:e3000382. [PMID: 31323030 PMCID: PMC6668837 DOI: 10.1371/journal.pbio.3000382] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 07/31/2019] [Accepted: 07/02/2019] [Indexed: 12/25/2022] Open
Abstract
The Hippo pathway directs cell differentiation during organogenesis, in part by restricting proliferation. How Hippo signaling maintains a proliferation-differentiation balance in developing tissues via distinct molecular targets is only beginning to be understood. Our study makes the unexpected finding that Hippo suppresses nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) signaling in pancreatic progenitors to permit cell differentiation and epithelial morphogenesis. We find that pancreas-specific deletion of the large tumor suppressor kinases 1 and 2 (Lats1/2PanKO) from mouse progenitor epithelia results in failure to differentiate key pancreatic lineages: acinar, ductal, and endocrine. We carried out an unbiased transcriptome analysis to query differentiation defects in Lats1/2PanKO. This analysis revealed increased expression of NFκB activators, including the pantetheinase vanin1 (Vnn1). Using in vivo and ex vivo studies, we show that VNN1 activates a detrimental cascade of processes in Lats1/2PanKO epithelium, including (1) NFκB activation and (2) aberrant initiation of epithelial-mesenchymal transition (EMT), which together disrupt normal differentiation. We show that exogenous stimulation of VNN1 or NFκB can trigger this cascade in wild-type (WT) pancreatic progenitors. These findings reveal an unexpected requirement for active suppression of NFκB by LATS1/2 during pancreas development, which restrains a cell-autonomous deleterious transcriptional program and thereby allows epithelial differentiation.
Collapse
Affiliation(s)
- Caitlin M. Braitsch
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - D. Berfin Azizoglu
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Yadanar Htike
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Haley R. Barlow
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Ulrike Schnell
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Christopher P. Chaney
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Thomas J. Carroll
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Ben Z. Stanger
- Department of Medicine and Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Ondine Cleaver
- Department of Molecular Biology and the Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
48
|
Abstract
Current kidney-on-chip models lack the 3D geometry, complexity, and functionality vital for recapitulating in vivo renal tissue. We report the fabrication and perfusion of 3D vascularized proximal tubules embedded within an engineered ECM that exhibit active reabsorption of solutes via tubular–vascular exchange. Using this model, we quantified albumin and glucose reabsorption over time. We also studied hyperglycemic effects in the absence and presence of a glucose transport inhibitor. Our 3D kidney tissue provides a platform for in vitro studies of kidney function, disease modeling, and pharmacology. Three-dimensional renal tissues that emulate the cellular composition, geometry, and function of native kidney tissue would enable fundamental studies of filtration and reabsorption. Here, we have created 3D vascularized proximal tubule models composed of adjacent conduits that are lined with confluent epithelium and endothelium, embedded in a permeable ECM, and independently addressed using a closed-loop perfusion system to investigate renal reabsorption. Our 3D kidney tissue allows for coculture of proximal tubule epithelium and vascular endothelium that exhibits active reabsorption via tubular–vascular exchange of solutes akin to native kidney tissue. Using this model, both albumin uptake and glucose reabsorption are quantified as a function of time. Epithelium–endothelium cross-talk is further studied by exposing proximal tubule cells to hyperglycemic conditions and monitoring endothelial cell dysfunction. This diseased state can be rescued by administering a glucose transport inhibitor. Our 3D kidney tissue provides a platform for in vitro studies of kidney function, disease modeling, and pharmacology.
Collapse
|
49
|
Munro DAD, Wineberg Y, Tarnick J, Vink CS, Li Z, Pridans C, Dzierzak E, Kalisky T, Hohenstein P, Davies JA. Macrophages restrict the nephrogenic field and promote endothelial connections during kidney development. eLife 2019; 8:43271. [PMID: 30758286 PMCID: PMC6374076 DOI: 10.7554/elife.43271] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/29/2019] [Indexed: 12/17/2022] Open
Abstract
The origins and functions of kidney macrophages in the adult have been explored, but their roles during development remain largely unknown. Here we characterise macrophage arrival, localisation, heterogeneity, and functions during kidney organogenesis. Using genetic approaches to ablate macrophages, we identify a role for macrophages in nephron progenitor cell clearance as mouse kidney development begins. Throughout renal organogenesis, most kidney macrophages are perivascular and express F4/80 and CD206. These macrophages are enriched for mRNAs linked to developmental processes, such as blood vessel morphogenesis. Using antibody-mediated macrophage-depletion, we show macrophages support vascular anastomoses in cultured kidney explants. We also characterise a subpopulation of galectin-3+ (Gal3+) myeloid cells within the developing kidney. Our findings may stimulate research into macrophage-based therapies for renal developmental abnormalities and have implications for the generation of bioengineered kidney tissues. The kidneys clean our blood by filtering out waste products while ensuring that useful components, like nutrients, remain in the bloodstream. Blood enters the kidneys through a network of intricately arranged blood vessels, which associate closely with the ‘cleaning tubes’ that carry out filtration. Human kidneys start developing during the early phases of embryonic development. During this process, the newly forming blood vessels and cleaning tubes must grow in the right places for the adult kidney to work properly. Macrophages are cells of the immune system that clear away foreign, diseased, or damaged cells. They are also thought to encourage growth of the developing kidney, but how exactly they do this has remained unknown. Munro et al. therefore wanted to find out when macrophages first appeared in the embryonic kidney and how they might help control their development. Experiments using mice revealed that the first macrophages arrived in the kidney early during its development, alongside newly forming blood vessels. Further investigation using genetically modified mice that did not have macrophages revealed that these immune cells were needed at this stage to clear away misplaced kidney cells and help ‘set the scene’ for future development. At later stages, macrophages in the kidney interacted closely with growing blood vessels. As well as producing molecules linked with blood vessel formation, the macrophages wrapped around the vessels themselves, sometimes even eating cells lining the vessels and the blood cells carried within them. These observations suggested that macrophages actively shaped the network of blood vessels developing within the kidneys. Experiments removing macrophages from kidney tissue confirmed this: in normal kidneys, the blood vessels grew into a continuous network, but in kidneys lacking macrophages, far fewer connections formed between the vessels. This work sheds new light on how the complex structures in the adult kidney first arise and could be useful in future research. For example, adding macrophages to simplified, laboratory-grown ‘mini-kidneys’ could make them better models to study kidney growth, while patients suffering from kidney diseases might benefit from new drugs targeting macrophages.
Collapse
Affiliation(s)
- David AD Munro
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Yishay Wineberg
- Department of Bioengineering, Bar-Ilan University, Ramat Gan, Israel.,Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Julia Tarnick
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Chris S Vink
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Zhuan Li
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Clare Pridans
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Elaine Dzierzak
- Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Tomer Kalisky
- Department of Bioengineering, Bar-Ilan University, Ramat Gan, Israel.,Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Peter Hohenstein
- Leiden University Medical Center, Leiden University, Leiden, The Netherlands.,The Roslin Institute, The University of Edinburgh, Midlothian, United Kingdom
| | - Jamie A Davies
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
50
|
Homan KA, Gupta N, Kroll KT, Kolesky DB, Skylar-Scott M, Miyoshi T, Mau D, Valerius MT, Ferrante T, Bonventre JV, Lewis JA, Morizane R. Flow-enhanced vascularization and maturation of kidney organoids in vitro. Nat Methods 2019; 16:255-262. [PMID: 30742039 PMCID: PMC6488032 DOI: 10.1038/s41592-019-0325-y] [Citation(s) in RCA: 508] [Impact Index Per Article: 101.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/21/2018] [Indexed: 01/01/2023]
Abstract
Kidney organoids derived from human pluripotent stem cells have glomerular- and tubular-like compartments that are largely avascular and immature in static culture. Here we report an in vitro method for culturing kidney organoids under flow on millifluidic chips, which expands their endogenous pool of endothelial progenitor cells and generates vascular networks with perfusable lumens surrounded by mural cells. We found that vascularized kidney organoids cultured under flow had more mature podocyte and tubular compartments with enhanced cellular polarity and adult gene expression compared with that in static controls. Glomerular vascular development progressed through intermediate stages akin to those involved in the embryonic mammalian kidney's formation of capillary loops abutting foot processes. The association of vessels with these compartments was reduced after disruption of the endogenous VEGF gradient. The ability to induce substantial vascularization and morphological maturation of kidney organoids in vitro under flow opens new avenues for studies of kidney development, disease, and regeneration.
Collapse
Affiliation(s)
- Kimberly A Homan
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | - Navin Gupta
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Katharina T Kroll
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | - David B Kolesky
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | - Mark Skylar-Scott
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | - Tomoya Miyoshi
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Donald Mau
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | - M Todd Valerius
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Thomas Ferrante
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | - Joseph V Bonventre
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jennifer A Lewis
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA. .,Harvard Stem Cell Institute, Cambridge, MA, USA.
| | - Ryuji Morizane
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA. .,Harvard Stem Cell Institute, Cambridge, MA, USA. .,Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|