1
|
Franks NE, Allen BL. Hedgehog-dependent and hedgehog-independent roles for growth arrest specific 1 in mammalian kidney morphogenesis. Development 2024; 151:dev203012. [PMID: 39629522 DOI: 10.1242/dev.203012] [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] [Accepted: 11/15/2024] [Indexed: 12/19/2024]
Abstract
Growth arrest specific 1 (GAS1) is a key regulator of mammalian embryogenesis, best known for its role in hedgehog (HH) signaling, but with additional described roles in the FGF, RET, and NOTCH pathways. Previous work indicated a later role for GAS1 in kidney development through FGF pathway modulation. Here, we demonstrate that GAS1 is essential for both mesonephrogenesis and metanephrogenesis - most notably, Gas1 deletion in mice results in renal agenesis in a genetic background-dependent fashion. Mechanistically, GAS1 promotes mesonephrogenesis in a HH-dependent fashion, performing a unique co-receptor function, while promoting metanephrogenesis in a HH-independent fashion, acting as a putative secreted RET co-receptor. Our data indicate that Gas1 deletion leads to renal agenesis through a transient reduction in metanephric mesenchyme proliferation - a phenotype that can be rescued by exogenous RET pathway stimulation. Overall, this study indicates that GAS1 contributes to early kidney development through the integration of multiple different signaling pathways.
Collapse
Affiliation(s)
- Nicole E Franks
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Benjamin L Allen
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
2
|
Kesdiren E, Martens H, Brand F, Werfel L, Wedekind L, Trowe MO, Schmitz J, Hennies I, Geffers R, Gucev Z, Seeman T, Schmidt S, Tasic V, Fasano L, Bräsen JH, Kispert A, Christians A, Haffner D, Weber RG. Heterozygous variants in the teashirt zinc finger homeobox 3 (TSHZ3) gene in human congenital anomalies of the kidney and urinary tract. Eur J Hum Genet 2024:10.1038/s41431-024-01710-y. [PMID: 39420202 DOI: 10.1038/s41431-024-01710-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/10/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Around 180 genes have been associated with congenital anomalies of the kidney and urinary tract (CAKUT) in mice, and represent promising novel candidate genes for human CAKUT. In whole-exome sequencing data of two siblings with genetically unresolved multicystic dysplastic kidneys (MCDK), prioritizing variants in murine CAKUT-associated genes yielded a rare variant in the teashirt zinc finger homeobox 3 (TSHZ3) gene. Therefore, the role of TSHZ3 in human CAKUT was assessed. Twelve CAKUT patients from 9/301 (3%) families carried five different rare heterozygous TSHZ3 missense variants predicted to be deleterious. CAKUT patients with versus without TSHZ3 variants were more likely to present with hydronephrosis, hydroureter, ureteropelvic junction obstruction, MCDK, and with genital anomalies, developmental delay, overlapping with the previously described phenotypes in Tshz3-mutant mice and patients with heterozygous 19q12-q13.11 deletions encompassing the TSHZ3 locus. Comparable with Tshz3-mutant mice, the smooth muscle layer was disorganized in the renal pelvis and thinner in the proximal ureter of the nephrectomy specimen of a TSHZ3 variant carrier compared to controls. TSHZ3 was expressed in the human fetal kidney, and strongly at embryonic day 11.5-14.5 in mesenchymal compartments of the murine ureter, kidney, and bladder. TSHZ3 variants in a 5' region were more frequent in CAKUT patients than in gnomAD samples (p < 0.001). Mutant TSHZ3 harboring N-terminal variants showed significantly altered SOX9 and/or myocardin binding, possibly adversely affecting smooth muscle differentiation. Our results provide evidence that heterozygous TSHZ3 variants are associated with human CAKUT, particularly MCDK, hydronephrosis, and hydroureter, and, inconsistently, with specific extrarenal features, including genital anomalies.
Collapse
Affiliation(s)
- Esra Kesdiren
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Helge Martens
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Frank Brand
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Lina Werfel
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
- Department of Pediatric Kidney, Liver, Metabolic and Neurological Diseases, Hannover Medical School, Hannover, Germany
| | - Lukas Wedekind
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
| | - Mark-Oliver Trowe
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
| | - Jessica Schmitz
- Nephropathology, Department of Pathology, Hannover Medical School, Hannover, Germany
| | - Imke Hennies
- Department of Pediatric Kidney, Liver, Metabolic and Neurological Diseases, Hannover Medical School, Hannover, Germany
| | - Robert Geffers
- Genome Analytics Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Zoran Gucev
- Pediatric Nephrology, University Children's Hospital, Skopje, Macedonia
| | - Tomáš Seeman
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Pediatrics, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Sonja Schmidt
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Velibor Tasic
- Pediatric Nephrology, University Children's Hospital, Skopje, Macedonia
| | - Laurent Fasano
- Aix-Marseille Univ, CNRS, IBDM UMR7288, Marseille, France
| | - Jan H Bräsen
- Nephropathology, Department of Pathology, Hannover Medical School, Hannover, Germany
| | - Andreas Kispert
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
| | - Anne Christians
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver, Metabolic and Neurological Diseases, Hannover Medical School, Hannover, Germany
| | - Ruthild G Weber
- Department of Human Genetics, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
3
|
Almet AA, Tsai YC, Watanabe M, Nie Q. Inferring pattern-driving intercellular flows from single-cell and spatial transcriptomics. Nat Methods 2024; 21:1806-1817. [PMID: 39187683 PMCID: PMC11466815 DOI: 10.1038/s41592-024-02380-w] [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: 08/04/2023] [Accepted: 07/23/2024] [Indexed: 08/28/2024]
Abstract
From single-cell RNA-sequencing (scRNA-seq) and spatial transcriptomics (ST), one can extract high-dimensional gene expression patterns that can be described by intercellular communication networks or decoupled gene modules. These two descriptions of information flow are often assumed to occur independently. However, intercellular communication drives directed flows of information that are mediated by intracellular gene modules, in turn triggering outflows of other signals. Methodologies to describe such intercellular flows are lacking. We present FlowSig, a method that infers communication-driven intercellular flows from scRNA-seq or ST data using graphical causal modeling and conditional independence. We benchmark FlowSig using newly generated experimental cortical organoid data and synthetic data generated from mathematical modeling. We demonstrate FlowSig's utility by applying it to various studies, showing that FlowSig can capture stimulation-induced changes to paracrine signaling in pancreatic islets, demonstrate shifts in intercellular flows due to increasing COVID-19 severity and reconstruct morphogen-driven activator-inhibitor patterns in mouse embryogenesis.
Collapse
Affiliation(s)
- Axel A Almet
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | - Yuan-Chen Tsai
- Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA, USA
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Momoko Watanabe
- Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, CA, USA
- Sue & Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA.
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA.
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
4
|
Ibi Y, Nishinakamura R. Generating kidney organoids based on developmental nephrology. Eur J Cell Biol 2024; 103:151450. [PMID: 39137450 DOI: 10.1016/j.ejcb.2024.151450] [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: 03/14/2024] [Revised: 08/05/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024] Open
Abstract
Over the past decade, the induction protocols for the two types of kidney organoids (nephron organoids and ureteric bud organoids) from pluripotent stem cells (PSCs) have been established based on the knowledge gained in developmental nephrology. Kidney organoids are now used for disease modeling and drug screening, but they also have potential as tools for clinical transplantation therapy. One of the options to achieve this goal would be to assemble multiple renal progenitor cells (nephron progenitor, ureteric bud, stromal progenitor) to reproduce the organotypic kidney structure from PSCs. At least from mouse PSCs, all the three progenitors have been induced and assembled into such "higher order" kidney organoids. We will provide an overview of the developmental nephrology required for the induction of renal progenitors and discuss recent advances and remaining challenges of kidney organoids for clinical transplantation therapy.
Collapse
Affiliation(s)
- Yutaro Ibi
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
5
|
Becknell B, El-Harakeh M, Rodriguez-Tirado F, Grounds KM, Li B, Kercsmar M, Wang X, Jackson AR. Keratin 5 basal cells are temporally regulated developmental and tissue repair progenitors in bladder urothelium. Am J Physiol Renal Physiol 2024; 326:F1078-F1090. [PMID: 38634130 PMCID: PMC11386981 DOI: 10.1152/ajprenal.00378.2023] [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: 12/05/2023] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024] Open
Abstract
Urothelium forms a distensible yet impermeable barrier, senses and transduces stimuli, and defends the urinary tract from mechanical, chemical, and bacterial injuries. Biochemical and genetic labeling studies support the existence of one or more progenitor populations with the capacity to rapidly regenerate the urothelium following injury, but slow turnover, a low mitotic index, and inconsistent methodologies obscure progenitor identity. The progenitor properties of basal keratin 5 urothelial cells (K5-UCs) have been previously investigated, but those studies focused on embryonic or adult bladder urothelium. Urothelium undergoes desquamation and apoptosis after birth, which requires postnatal proliferation and restoration. Therefore, we mapped the fate of bladder K5-UCs across postnatal development/maturation and following administration of cyclophosphamide to measure homeostatic and reparative progenitor capacities, respectively. In vivo studies demonstrate that basal K5-UCs are age-restricted progenitors in neonates and juveniles, but not in adult mice. Neonatal K5-UCs retain a superior progenitor capacity in vitro, forming larger and more differentiated urothelial organoids than adult K5-UCs. Accordingly, K5-UC transcriptomes are temporally distinct, with enrichment of transcripts associated with cell proliferation and differentiation in neonates. Induction of urothelial proliferation is sufficient to restore adult K5-UC progenitor capacity. Our findings advance the understanding of urothelial progenitors and support a linear model of urothelial formation and regeneration, which may have significant impact on therapeutic development or tissue engineering strategies.NEW & NOTEWORTHY Fate mapping reveals an important linear relationship, whereby bladder basal urothelial cells give rise to intermediate and superficial cells in an age-restricted manner and contribute to tissue repair. Neonatal basal cells reprise their role as superior progenitors in vitro and display distinct transcriptional signatures, which suggest progenitor function is at least partially cell intrinsic. However, the urothelium progenitor niche cannot be overlooked, since FGF7 rescues adult basal cell progenitor function.
Collapse
Affiliation(s)
- Brian Becknell
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
- Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, Ohio, United States
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States
| | - Mohammad El-Harakeh
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Felipe Rodriguez-Tirado
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Kelly M Grounds
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Birong Li
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Macie Kercsmar
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Xin Wang
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
| | - Ashley R Jackson
- Kidney and Urinary Tract Center, Nationwide Children's Hospital, Columbus, Ohio, United States
- Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, Ohio, United States
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States
| |
Collapse
|
6
|
Newton AH, Smith CA. Resolving the mechanisms underlying epithelial-to-mesenchymal transition of the lateral plate mesoderm. Genesis 2024; 62:e23531. [PMID: 37443419 DOI: 10.1002/dvg.23531] [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: 04/09/2023] [Revised: 06/02/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023]
Abstract
Formation of the vertebrate limb buds begins with a localized epithelial-to-mesenchymal transition (EMT) of the somatic lateral plate mesoderm (LPM). While the processes that drive proliferation and outgrowth of the limb mesenchyme are well established, the fundamental mechanisms that precede this process and initiate EMT are less understood. In this review, we outline putative drivers of EMT of the LPM, drawing from analyses across a range of vertebrates and developmental models. We detail the expression patterns of key EMT transcriptional regulators in the somatic LPM of the presumptive limb fields, and their potential role in producing a mesenchymal cell fate. These include a putative cooperative role between the EMT inducers PRRX1 and TWIST1, supported by evidence in zebrafish and chicken models but unconfirmed data from mice. As such, additional functional data are required to definitively determine the mechanisms that initiate and drive EMT of the somatic LPM, a critical transition preceding formation of the limb bud mesenchyme.
Collapse
Affiliation(s)
- Axel H Newton
- School of BioSciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Craig A Smith
- Department of Anatomy and Physiology, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
7
|
D'Cruz R, Kim YK, Mulder J, Ibeh N, Jiang N, Tian Y, Rosenblum ND. Hedgehog signalling in Foxd1+ embryonic kidney stromal progenitors controls nephron formation via Cxcl12 and Wnt5a. J Pathol 2023; 261:385-400. [PMID: 37772431 DOI: 10.1002/path.6195] [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: 04/18/2023] [Revised: 07/05/2023] [Accepted: 07/30/2023] [Indexed: 09/30/2023]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are characterised by a spectrum of structural and histologic abnormalities and are the major cause of childhood kidney failure. During kidney morphogenesis, the formation of a critical number of nephrons is an embryonic process supported, in part, by signalling between nephrogenic precursors and Foxd1-positive stromal progenitor cells. Low nephron number and abnormal patterning of the stroma are signature pathological features among CAKUT phenotypes with decreased kidney function. Despite their critical contribution to CAKUT pathogenesis, the mechanisms that underlie a low nephron number and the functional contribution of a disorganised renal stroma to nephron number are both poorly defined. Here, we identify a primary pathogenic role for increased Hedgehog signalling in embryonic renal stroma in the genesis of congenital low nephron number. Pharmacologic activation of Hedgehog (Hh) signalling in human kidney organoid tissue decreased the number of nephrons and generated excess stroma. The mechanisms underlying these pathogenic effects were delineated in genetic mouse models in which Hh signalling was constitutively activated in a cell lineage-specific manner. Cre-mediated excision of Ptch1 in Foxd1+ stromal progenitor cells, but not in Six2+ nephrogenic precursor cells, generated kidney malformation, identifying the stroma as a driver of low nephron number. Single-cell RNA sequencing analysis identified Cxcl12 and Wnt5a as downstream targets of increased stromal Hh signalling, findings supported by analysis in human kidney organoids. In vivo deficiency of Cxcl12 or Wnt5a in mice with increased stromal Hh signalling improved nephron endowment. These results demonstrate that dysregulated Hh signalling in embryonic renal stromal cells inhibits nephron formation in a manner dependent on Cxcl12 and Wnt5a. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Robert D'Cruz
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Yun-Kyo Kim
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
| | - Jaap Mulder
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
- Division of Nephrology, Hospital for Sick Children, Toronto, Canada
| | - Neke Ibeh
- Princess Margaret Cancer Centre, Unity Health Network, Toronto, Canada
| | - Nan Jiang
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
| | - Yilin Tian
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
| | - Norman D Rosenblum
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Division of Nephrology, Hospital for Sick Children, Toronto, Canada
- Department of Physiology, University of Toronto, Toronto, Canada
- Department of Pediatrics, University of Toronto, Toronto, Canada
| |
Collapse
|
8
|
Yamamoto S, Matsui K, Kinoshita Y, Hiroshi Sasaki, Sekine H, Saito Y, Nakayama Y, Kume H, Kimura T, Yokoo T, Kobayashi E. Successful reconstruction of the rat ureter by a syngeneic collagen tube with a cardiomyocyte sheet. Regen Ther 2023; 24:561-567. [PMID: 37868722 PMCID: PMC10584669 DOI: 10.1016/j.reth.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 10/01/2023] [Accepted: 10/05/2023] [Indexed: 10/24/2023] Open
Abstract
Introduction Ureteral injuries require surgical intervention as they lead to loss of renal function. The current reconstructive techniques for long ureteral defects are problematic. Consequently, this study aimed to reconstruct the ureter in a rat model using subcutaneously prepared autologous collagen tubes (Biotubes). Methods The lower ureter of LEW/SsNSlc rats was ligated to dilate the ureter to make anastomosis easier, and reconstruction was performed six days later by anastomosing the dilated ureter and bladder with a Biotube that was prepared subcutaneously in syngeneic rats. Some rats underwent left nephrectomy and ureter reconstruction simultaneously as negative controls to evaluate the effects of urine flow on patency. The other rats were divided into three groups as follows: a group in which the ureter was reconstructed with the Biotube alone, a group in which cardiomyocyte sheets made from the neonatal hearts of syngeneic rats were wrapped around the Biotube, and a group in which an adipose-derived stem cell sheets made from the inguinal fat of adult syngeneic rats were wrapped. Contrast-enhanced computed tomography and pathological evaluations were performed two weeks after reconstruction. Result In the Biotube alone group, all tubes were occluded and hydronephrosis developed, whereas the urothelium regenerated beyond the anastomosis when the left kidney was not removed, suggesting that urothelial epithelial spread occurred with urinary flow. The patency of the ureteral lumen was obtained in some rats in the cardiomyocyte sheet covered group, whereas stricture or obstruction of the reconstructed ureter was observed in all rats in the other groups. Pathological evaluation revealed a layered urothelial structure in the cardiomyocyte sheet covered group, although only a small amount of cardiomyocyte sheets remained. Conclusion Urinary flow may support the epithelial spread of the urothelium into the reconstructed ureter. Neonatal rat cardiomyocyte sheets supported the patency of the regenerated ureter with a layered urothelium.
Collapse
Affiliation(s)
- Shutaro Yamamoto
- Department of Urology, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Kenji Matsui
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Yoshitaka Kinoshita
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - Hiroshi Sasaki
- Department of Urology, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Hidekazu Sekine
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, Tokyo 162-8666, Japan
| | - Yatsumu Saito
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | | | - Haruki Kume
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - Takahiro Kimura
- Department of Urology, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Eiji Kobayashi
- Department of Kidney Regenerative Medicine, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| |
Collapse
|
9
|
Mannan A, Dhiamn S, Garg N, Singh TG. Pharmacological modulation of Sonic Hedgehog signaling pathways in Angiogenesis: A mechanistic perspective. Dev Biol 2023; 504:58-74. [PMID: 37739118 DOI: 10.1016/j.ydbio.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
The Sonic hedgehog (SHh) signaling pathway is an imperative operating network that helps in regulates the critical events during the development processes like multicellular embryo growth and patterning. Disruptions in SHh pathway regulation can have severe consequences, including congenital disabilities, stem cell renewal, tissue regeneration, and cancer/tumor growth. Activation of the SHh signal occurs when SHh binds to the receptor complex of Patch (Ptc)-mediated Smoothened (Smo) (Ptc-smo), initiating downstream signaling. This review explores how pharmacological modulation of the SHh pathway affects angiogenesis through canonical and non-canonical pathways. The canonical pathway for angiogenesis involves the activation of angiogenic cytokines such as fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), placental growth factor (PGF), hepatocyte growth factor (HGF), platelet-derived growth factor (PDGF), stromal cell-derived factor 1α, transforming growth factor-β1 (TGF-β1), and angiopoietins (Ang-1 and Ang-2), which facilitate the process of angiogenesis. The Non-canonical pathway includes indirect activation of certain pathways like iNOS/Netrin-1/PKC, RhoA/Rock, ERK/MAPK, PI3K/Akt, Wnt/β-catenin, Notch signaling pathway, and so on. This review will provide a better grasp of the mechanistic approach of SHh in mediating angiogenesis, which can aid in the suppression of certain cancer and tumor growths.
Collapse
Affiliation(s)
- Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Sonia Dhiamn
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Nikhil Garg
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
10
|
Alves MBR, Girardet L, Augière C, Moon KH, Lavoie-Ouellet C, Bernet A, Soulet D, Calvo E, Teves ME, Beauparlant CJ, Droit A, Bastien A, Robert C, Bok J, Hinton BT, Belleannée C. Hedgehog signaling regulates Wolffian duct development through the primary cilium†. Biol Reprod 2023; 108:241-257. [PMID: 36525341 PMCID: PMC9930401 DOI: 10.1093/biolre/ioac210] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 11/01/2022] [Accepted: 11/25/2022] [Indexed: 12/23/2022] Open
Abstract
Primary cilia play pivotal roles in embryonic patterning and organogenesis through transduction of the Hedgehog signaling pathway (Hh). Although mutations in Hh morphogens impair the development of the gonads and trigger male infertility, the contribution of Hh and primary cilia in the development of male reproductive ductules, including the epididymis, remains unknown. From a Pax2Cre; IFT88fl/fl knock-out mouse model, we found that primary cilia deletion is associated with imbalanced Hh signaling and morphometric changes in the Wolffian duct (WD), the embryonic precursor of the epididymis. Similar effects were observed following pharmacological blockade of primary cilia formation and Hh modulation on WD organotypic cultures. The expression of genes involved in extracellular matrix, mesenchymal-epithelial transition, canonical Hh and WD development was significantly altered after treatments. Altogether, we identified the primary cilia-dependent Hh signaling as a master regulator of genes involved in WD development. This provides new insights regarding the etiology of sexual differentiation and male infertility issues.
Collapse
Affiliation(s)
- Maíra Bianchi Rodrigues Alves
- Faculty of Medicine, Department of Obstetrics, Gynecology and Reproduction, CHU de Québec Research Center (CHUL)—Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle—Université Laval, Quebec City, QC, Canada
| | - Laura Girardet
- Faculty of Medicine, Department of Obstetrics, Gynecology and Reproduction, CHU de Québec Research Center (CHUL)—Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle—Université Laval, Quebec City, QC, Canada
| | - Céline Augière
- Faculty of Medicine, Department of Obstetrics, Gynecology and Reproduction, CHU de Québec Research Center (CHUL)—Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle—Université Laval, Quebec City, QC, Canada
| | - Kyeong Hye Moon
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Camille Lavoie-Ouellet
- Faculty of Medicine, Department of Obstetrics, Gynecology and Reproduction, CHU de Québec Research Center (CHUL)—Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle—Université Laval, Quebec City, QC, Canada
| | - Agathe Bernet
- Faculty of Medicine, Department of Obstetrics, Gynecology and Reproduction, CHU de Québec Research Center (CHUL)—Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle—Université Laval, Quebec City, QC, Canada
| | - Denis Soulet
- Faculty of Pharmacy, Department of Neurosciences, CHU de Québec Research Center (CHUL)—Université Laval, Quebec City, QC, Canada
| | - Ezequiel Calvo
- Faculty of Medicine, Department of Obstetrics, Gynecology and Reproduction, CHU de Québec Research Center (CHUL)—Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle—Université Laval, Quebec City, QC, Canada
| | - Maria E Teves
- Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, USA
| | - Charles Joly Beauparlant
- Computational Biology Laboratory Research Centre, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Arnaud Droit
- Computational Biology Laboratory Research Centre, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Alexandre Bastien
- Faculty of Agriculture and Food Sciences, Department of Animal Sciences—Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle—Université Laval, Quebec City, QC, Canada
| | - Claude Robert
- Faculty of Agriculture and Food Sciences, Department of Animal Sciences—Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle—Université Laval, Quebec City, QC, Canada
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Barry T Hinton
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Clémence Belleannée
- Faculty of Medicine, Department of Obstetrics, Gynecology and Reproduction, CHU de Québec Research Center (CHUL)—Centre de Recherche en Reproduction, Développement et Santé Intergénérationnelle—Université Laval, Quebec City, QC, Canada
| |
Collapse
|
11
|
Jia D, Zhou Z, Kwon OJ, Zhang L, Wei X, Zhang Y, Yi M, Roudier MP, Regier MC, Dumpit R, Nelson PS, Headley M, True L, Lin DW, Morrissey C, Creighton CJ, Xin L. Stromal FOXF2 suppresses prostate cancer progression and metastasis by enhancing antitumor immunity. Nat Commun 2022; 13:6828. [PMID: 36369237 PMCID: PMC9652358 DOI: 10.1038/s41467-022-34665-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 11/02/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) mediate an immunosuppressive effect, but the underlying mechanism remains incompletely defined. Here we show that increasing prostatic stromal Foxf2 suppresses the growth and progression of both syngeneic and autochthonous mouse prostate cancer models in an immunocompetent context. Mechanistically, Foxf2 moderately attenuates the CAF phenotype and transcriptionally downregulates Cxcl5, which diminish the immunosuppressive myeloid cells and enhance T cell cytotoxicity. Increasing prostatic stromal Foxf2 sensitizes prostate cancer to the immune checkpoint blockade therapies. Augmenting lung stromal Foxf2 also mediates an immunosuppressive milieu and inhibits lung colonization of prostate cancer. FOXF2 is expressed higher in the stroma of human transition zone (TZ) than peripheral zone (PZ) prostate. The stromal FOXF2 expression level in primary prostate cancers inversely correlates with the Gleason grade. Our study establishes Foxf2 as a stromal transcription factor modulating the tumor immune microenvironment and potentially explains why cancers are relatively rare and indolent in the TZ prostate.
Collapse
Affiliation(s)
- Deyong Jia
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Zhicheng Zhou
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Oh-Joon Kwon
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Li Zhang
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Xing Wei
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Yiqun Zhang
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Mingyang Yi
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | - Mary C Regier
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Ruth Dumpit
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter S Nelson
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Mark Headley
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Lawrence True
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Daniel W Lin
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Li Xin
- Department of Urology, University of Washington, Seattle, WA, USA.
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
12
|
Newton AH, Williams SM, Major AT, Smith CA. Cell lineage specification and signalling pathway use during development of the lateral plate mesoderm and forelimb mesenchyme. Development 2022; 149:276597. [DOI: 10.1242/dev.200702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 08/25/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The lateral plate mesoderm (LPM) is a transient tissue that produces a diverse range of differentiated structures, including the limbs. However, the molecular mechanisms that drive early LPM specification and development are poorly understood. In this study, we use single-cell transcriptomics to define the cell-fate decisions directing LPM specification, subdivision and early initiation of the forelimb mesenchyme in chicken embryos. We establish a transcriptional atlas and global cell-cell signalling interactions in progenitor, transitional and mature cell types throughout the developing forelimb field. During LPM subdivision, somatic and splanchnic LPM fate is achieved through activation of lineage-specific gene modules. During the earliest stages of limb initiation, we identify activation of TWIST1 in the somatic LPM as a putative driver of limb bud epithelial-to-mesenchymal transition. Furthermore, we define a new role for BMP signalling during early limb development, revealing that it is necessary for inducing a somatic LPM fate and initiation of limb outgrowth, potentially through activation of TBX5. Together, these findings provide new insights into the mechanisms underlying LPM development, somatic LPM fate choice and early initiation of the vertebrate limb.
Collapse
Affiliation(s)
- Axel H. Newton
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University 1 , Victoria , Australia
- BioScience 4, School of BioSciences, The University of Melbourne 2 , Victoria , Australia
| | - Sarah M. Williams
- Monash University 3 Monash Bioinformatics Platform , , Victoria , Australia
- Queensland Cyber Infrastructure Foundation, University of Queensland 4 , Queensland , Australia
| | - Andrew T. Major
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University 1 , Victoria , Australia
| | - Craig A. Smith
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University 1 , Victoria , Australia
| |
Collapse
|
13
|
Deuper L, Meuser M, Thiesler H, Jany UWH, Rudat C, Hildebrandt H, Trowe MO, Kispert A. Mesenchymal FGFR1 and FGFR2 control patterning of the ureteric mesenchyme by balancing SHH and BMP4 signaling. Development 2022; 149:276592. [DOI: 10.1242/dev.200767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 08/19/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The coordinated development of the mesenchymal and epithelial progenitors of the murine ureter depends on a complex interplay of diverse signaling activities. We have recently shown that epithelial FGFR2 signaling regulates stratification and differentiation of the epithelial compartment by enhancing epithelial Shh expression, and mesenchymal SHH and BMP4 activity. Here, we show that FGFR1 and FGFR2 expression in the mesenchymal primordium impinges on the SHH/BMP4 signaling axis to regulate mesenchymal patterning and differentiation. Mouse embryos with conditional loss of Fgfr1 and Fgfr2 in the ureteric mesenchyme exhibited reduced mesenchymal proliferation and prematurely activated lamina propria formation at the expense of the smooth muscle cell program. They also manifested hydroureter at birth. Molecular profiling detected increased SHH, WNT and retinoic acid signaling, whereas BMP4 signaling in the mesenchyme was reduced. Pharmacological activation of SHH signaling in combination with inhibition of BMP4 signaling recapitulated the cellular changes in explant cultures of wild-type ureters. Additional experiments suggest that mesenchymal FGFR1 and FGFR2 act as a sink for FGF ligands to dampen activation of Shh and BMP receptor gene expression by epithelial FGFR2 signaling.
Collapse
Affiliation(s)
- Lena Deuper
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| | - Max Meuser
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| | - Hauke Thiesler
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover 2 , 30625 Hannover , Germany
| | - Ulrich W. H. Jany
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| | - Carsten Rudat
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| | - Herbert Hildebrandt
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover 2 , 30625 Hannover , Germany
| | - Mark-Oliver Trowe
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| | - Andreas Kispert
- Institute of Molecular Biology, Medizinische Hochschule Hannover 1 , 30625 Hannover , Germany
| |
Collapse
|
14
|
Li L, Li Y, Yang J, Xie X, Chen H. The immune responses to different Uropathogens call individual interventions for bladder infection. Front Immunol 2022; 13:953354. [PMID: 36081496 PMCID: PMC9445553 DOI: 10.3389/fimmu.2022.953354] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Urinary tract infection (UTI) caused by uropathogens is the most common infectious disease and significantly affects all aspects of the quality of life of the patients. However, uropathogens are increasingly becoming antibiotic-resistant, which threatens the only effective treatment option available-antibiotic, resulting in higher medical costs, prolonged hospital stays, and increased mortality. Currently, people are turning their attention to the immune responses, hoping to find effective immunotherapeutic interventions which can be alternatives to the overuse of antibiotic drugs. Bladder infections are caused by the main nine uropathogens and the bladder executes different immune responses depending on the type of uropathogens. It is essential to understand the immune responses to diverse uropathogens in bladder infection for guiding the design and development of immunotherapeutic interventions. This review firstly sorts out and comparatively analyzes the immune responses to the main nine uropathogens in bladder infection, and summarizes their similarities and differences. Based on these immune responses, we innovatively propose that different microbial bladder infections should adopt corresponding immunomodulatory interventions, and the same immunomodulatory intervention can also be applied to diverse microbial infections if they share the same effective therapeutic targets.
Collapse
Affiliation(s)
- Linlong Li
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Yangyang Li
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Jiali Yang
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiang Xie
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- *Correspondence: Xiang Xie, ; Huan Chen,
| | - Huan Chen
- The School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, China
- *Correspondence: Xiang Xie, ; Huan Chen,
| |
Collapse
|
15
|
Fink EE, Sona S, Tran U, Desprez PE, Bradley M, Qiu H, Eltemamy M, Wee A, Wolkov M, Nicolas M, Min B, Haber GP, Wessely O, Lee BH, Ting AH. Single-cell and spatial mapping Identify cell types and signaling Networks in the human ureter. Dev Cell 2022; 57:1899-1916.e6. [PMID: 35914526 PMCID: PMC9381170 DOI: 10.1016/j.devcel.2022.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 03/18/2022] [Accepted: 07/05/2022] [Indexed: 01/16/2023]
Abstract
Tissue engineering offers a promising treatment strategy for ureteral strictures, but its success requires an in-depth understanding of the architecture, cellular heterogeneity, and signaling pathways underlying tissue regeneration. Here, we define and spatially map cell populations within the human ureter using single-cell RNA sequencing, spatial gene expression, and immunofluorescence approaches. We focus on the stromal and urothelial cell populations to enumerate the distinct cell types composing the human ureter and infer potential cell-cell communication networks underpinning the bi-directional crosstalk between these compartments. Furthermore, we analyze and experimentally validate the importance of the sonic hedgehog (SHH) signaling pathway in adult progenitor cell maintenance. The SHH-expressing basal cells support organoid generation in vitro and accurately predict the differentiation trajectory from basal progenitor cells to terminally differentiated umbrella cells. Our results highlight the essential processes involved in adult ureter tissue homeostasis and provide a blueprint for guiding ureter tissue engineering.
Collapse
Affiliation(s)
- Emily E Fink
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Surbhi Sona
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Nutrition, Center for Proteomics and Bioinformatics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Uyen Tran
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Pierre-Emmanuel Desprez
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Urology, CHU Lille, Claude Huriez Hospital, Université Lille, 59000 Lille, France
| | - Matthew Bradley
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Hong Qiu
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Mohamed Eltemamy
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Alvin Wee
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Madison Wolkov
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Marlo Nicolas
- Pathology & Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Booki Min
- Department of Microbiology and Immunology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Georges-Pascal Haber
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Oliver Wessely
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Byron H Lee
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - Angela H Ting
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
16
|
Kurz J, Weiss AC, Lüdtke THW, Deuper L, Trowe MO, Thiesler H, Hildebrandt H, Heineke J, Duncan SA, Kispert A. GATA6 is a crucial factor for Myocd expression in the visceral smooth muscle cell differentiation program of the murine ureter. Development 2022; 149:dev200522. [PMID: 35905011 PMCID: PMC10656427 DOI: 10.1242/dev.200522] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/29/2022] [Indexed: 11/20/2023]
Abstract
Smooth muscle cells (SMCs) are a crucial component of the mesenchymal wall of the ureter, as they account for the efficient removal of the urine from the renal pelvis to the bladder by means of their contractile activity. Here, we show that the zinc-finger transcription factor gene Gata6 is expressed in mesenchymal precursors of ureteric SMCs under the control of BMP4 signaling. Mice with a conditional loss of Gata6 in these precursors exhibit a delayed onset and reduced level of SMC differentiation and peristaltic activity, as well as dilatation of the ureter and renal pelvis (hydroureternephrosis) at birth and at postnatal stages. Molecular profiling revealed a delayed and reduced expression of the myogenic driver gene Myocd, but the activation of signaling pathways and transcription factors previously implicated in activation of the visceral SMC program in the ureter was unchanged. Additional gain-of-function experiments suggest that GATA6 cooperates with FOXF1 in Myocd activation and SMC differentiation, possibly as pioneer and lineage-determining factors, respectively.
Collapse
Affiliation(s)
- Jennifer Kurz
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Anna-Carina Weiss
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Timo H.-W. Lüdtke
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Lena Deuper
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Hauke Thiesler
- Institut für Klinische Biochemie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Herbert Hildebrandt
- Institut für Klinische Biochemie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | - Joerg Heineke
- Abteilung für Kardiovaskuläre Physiologie, European Center for Angioscience, Medizinische Fakultät Mannheim, Universität Heidelberg, D-68167 Mannheim, Germany
| | - Stephen A. Duncan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Andreas Kispert
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| |
Collapse
|
17
|
Kurz J, Weiss AC, Thiesler H, Qasrawi F, Deuper L, Kaur J, Rudat C, Lüdtke TH, Wojahn I, Hildebrandt H, Trowe MO, Kispert A. Notch signaling is a novel regulator of visceral smooth muscle cell differentiation in the murine ureter. Development 2022; 149:274136. [DOI: 10.1242/dev.199735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 12/31/2021] [Indexed: 01/13/2023]
Abstract
ABSTRACT
The contractile phenotype of smooth muscle cells (SMCs) is transcriptionally controlled by a complex of the DNA-binding protein SRF and the transcriptional co-activator MYOCD. The pathways that activate expression of Myocd and of SMC structural genes in mesenchymal progenitors are diverse, reflecting different intrinsic and extrinsic signaling inputs. Taking the ureter as a model, we analyzed whether Notch signaling, a pathway previously implicated in vascular SMC development, also affects visceral SMC differentiation. We show that mice with a conditional deletion of the unique Notch mediator RBPJ in the undifferentiated ureteric mesenchyme exhibit altered ureter peristalsis with a delayed onset, and decreased contraction frequency and intensity at fetal stages. They also develop hydroureter 2 weeks after birth. Notch signaling is required for precise temporal activation of Myocd expression and, independently, for expression of a group of late SMC structural genes. Based on additional expression analyses, we suggest that a mesenchymal JAG1-NOTCH2/NOTCH3 module regulates visceral SMC differentiation in the ureter in a biphasic and bimodal manner, and that its molecular function differs from that in the vascular system.
Collapse
Affiliation(s)
- Jennifer Kurz
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Anna-Carina Weiss
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Hauke Thiesler
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Fairouz Qasrawi
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Lena Deuper
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Jaskiran Kaur
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Carsten Rudat
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Timo H. Lüdtke
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Irina Wojahn
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Herbert Hildebrandt
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Andreas Kispert
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| |
Collapse
|
18
|
Yin W, Liontos A, Koepke J, Ghoul M, Mazzocchi L, Liu X, Lu C, Wu H, Fysikopoulos A, Sountoulidis A, Seeger W, Ruppert C, Günther A, Stainier DYR, Samakovlis C. An essential function for autocrine hedgehog signaling in epithelial proliferation and differentiation in the trachea. Development 2022; 149:274222. [PMID: 35112129 PMCID: PMC8918789 DOI: 10.1242/dev.199804] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 12/07/2021] [Indexed: 12/12/2022]
Abstract
The tracheal epithelium is a primary target for pulmonary diseases as it provides a conduit for air flow between the environment and the lung lobes. The cellular and molecular mechanisms underlying airway epithelial cell proliferation and differentiation remain poorly understood. Hedgehog (HH) signaling orchestrates communication between epithelial and mesenchymal cells in the lung, where it modulates stromal cell proliferation, differentiation and signaling back to the epithelium. Here, we reveal a previously unreported autocrine function of HH signaling in airway epithelial cells. Epithelial cell depletion of the ligand sonic hedgehog (SHH) or its effector smoothened (SMO) causes defects in both epithelial cell proliferation and differentiation. In cultured primary human airway epithelial cells, HH signaling inhibition also hampers cell proliferation and differentiation. Epithelial HH function is mediated, at least in part, through transcriptional activation, as HH signaling inhibition leads to downregulation of cell type-specific transcription factor genes in both the mouse trachea and human airway epithelial cells. These results provide new insights into the role of HH signaling in epithelial cell proliferation and differentiation during airway development. Summary: A conserved autocrine role for HH signaling in tracheal epithelial cell proliferation and differentiation is revealed, suggesting potential new interventions for airway epithelial proliferation and differentiation defects.
Collapse
Affiliation(s)
- Wenguang Yin
- Cardio-Pulmonary Institute, Member of the German Center for Lung Research (DZL), University of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University of Giessen, Giessen 35392, Germany.,State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, People's Republic of China.,Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim 61231, Germany
| | - Andreas Liontos
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, S-10691 Stockholm, Sweden.,Science for Life Laboratory, Stockholm University, Solna 171 21, Sweden
| | - Janine Koepke
- Cardio-Pulmonary Institute, Member of the German Center for Lung Research (DZL), University of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University of Giessen, Giessen 35392, Germany
| | - Maroua Ghoul
- Cardio-Pulmonary Institute, Member of the German Center for Lung Research (DZL), University of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University of Giessen, Giessen 35392, Germany
| | - Luciana Mazzocchi
- Cardio-Pulmonary Institute, Member of the German Center for Lung Research (DZL), University of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University of Giessen, Giessen 35392, Germany
| | - Xinyuan Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, People's Republic of China
| | - Chunyan Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, People's Republic of China
| | - Haoyu Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510182, People's Republic of China
| | - Athanasios Fysikopoulos
- Cardio-Pulmonary Institute, Member of the German Center for Lung Research (DZL), University of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University of Giessen, Giessen 35392, Germany
| | - Alexandros Sountoulidis
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, S-10691 Stockholm, Sweden.,Science for Life Laboratory, Stockholm University, Solna 171 21, Sweden
| | - Werner Seeger
- Cardio-Pulmonary Institute, Member of the German Center for Lung Research (DZL), University of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University of Giessen, Giessen 35392, Germany.,Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| | - Clemens Ruppert
- Cardio-Pulmonary Institute, Member of the German Center for Lung Research (DZL), University of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University of Giessen, Giessen 35392, Germany
| | - Andreas Günther
- Cardio-Pulmonary Institute, Member of the German Center for Lung Research (DZL), University of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University of Giessen, Giessen 35392, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim 61231, Germany
| | - Christos Samakovlis
- Cardio-Pulmonary Institute, Member of the German Center for Lung Research (DZL), University of Giessen and Marburg Lung Center (UGMLC), Justus Liebig University of Giessen, Giessen 35392, Germany.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, S-10691 Stockholm, Sweden.,Science for Life Laboratory, Stockholm University, Solna 171 21, Sweden.,Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim 61231, Germany
| |
Collapse
|
19
|
Junga A, Siņicins I, Pētersons A, Pilmane M. Evaluation of PGP 9.5, NGFR, TGFβ1, FGFR1, MMP-2, AT2R2, SHH, and TUNEL in Primary Obstructive Megaureter Tissue. J Histochem Cytochem 2022; 70:139-149. [PMID: 34915763 PMCID: PMC8777373 DOI: 10.1369/00221554211063515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Primary obstructive megaureter (POM) morphogenesis is not fully known. The aim of the study was to evaluate the appearance of different factors that might take part in the pathogenesis of POM. Megaureter tissues of 14 children were stained with hematoxylin and eosin as well as with immunohistochemistry for protein gene product 9.5, nerve growth factor receptor, transforming growth factor beta 1 (TGFβ1), fibroblast growth factor receptor 1 (FGFR1), matrix metalloproteinase 2 (MMP-2), angiotensin 2 receptor type 2, and sonic hedgehog (SHH) protein. Apoptosis was detected by terminal dUTP nick-end labeling reaction. POM tissues revealed transitional epithelium with scattered vacuolization, submucosa with inflammatory cells, and focally vacuolized and chaotically organized muscle layers. Apoptosis, appearance of MMP-2, FGFR1, and SHH prevailed, but TGFβ1 positive cell number was lower in patients. Correlation between MMP-2 in epithelium and endothelium, FGFR1 and MMP-2 in epithelium, and TGFβ1 in epithelium and connective tissue in patients was detected. POM morphopathogenesis involves an apoptotic cell death of epithelium and smooth muscle as well as tissue degradation in epithelium and connective tissue of the ureter wall. The decrease of tissue growth through diminished TGFβ1 expression and stimulation of FGFR1 and MMP-2 suggests a disbalance of tissue remodelation in the megaureter wall.
Collapse
Affiliation(s)
- Anna Junga
- Anna Junga, Institute of Anatomy and
Anthropology, Riga Stradins University, Kronvalda bulv 9, Riga LV-1010, Latvia.
E-mail:
| | - Ivo Siņicins
- Institute of Anatomy and Anthropology, Riga
Stradins University, Riga, Latvia
| | - Aigars Pētersons
- Department of Children Surgery, Riga Stradins
University, Riga, Latvia
| | - Māra Pilmane
- Institute of Anatomy and Anthropology, Riga
Stradins University, Riga, Latvia
| |
Collapse
|
20
|
Meuser M, Deuper L, Rudat C, Aydoğdu N, Thiesler H, Zarnovican P, Hildebrandt H, Trowe MO, Kispert A. FGFR2 signaling enhances the SHH-BMP4 signaling axis in early ureter development. Development 2022; 149:273983. [DOI: 10.1242/dev.200021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/09/2021] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The patterned array of basal, intermediate and superficial cells in the urothelium of the mature ureter arises from uncommitted epithelial progenitors of the distal ureteric bud. Urothelial development requires signaling input from surrounding mesenchymal cells, which, in turn, depend on cues from the epithelial primordium to form a layered fibro-muscular wall. Here, we have identified FGFR2 as a crucial component in this reciprocal signaling crosstalk in the murine ureter. Loss of Fgfr2 in the ureteric epithelium led to reduced proliferation, stratification, intermediate and basal cell differentiation in this tissue, and affected cell survival and smooth muscle cell differentiation in the surrounding mesenchyme. Loss of Fgfr2 impacted negatively on epithelial expression of Shh and its mesenchymal effector gene Bmp4. Activation of SHH or BMP4 signaling largely rescued the cellular defects of mutant ureters in explant cultures. Conversely, inhibition of SHH or BMP signaling in wild-type ureters recapitulated the mutant phenotype in a dose-dependent manner. Our study suggests that FGF signals from the mesenchyme enhance, via epithelial FGFR2, the SHH-BMP4 signaling axis to drive urothelial and mesenchymal development in the early ureter.
Collapse
Affiliation(s)
- Max Meuser
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Lena Deuper
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Carsten Rudat
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Nurullah Aydoğdu
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Hauke Thiesler
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Patricia Zarnovican
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Herbert Hildebrandt
- Institute of Clinical Biochemistry, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Andreas Kispert
- Institute of Molecular Biology, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| |
Collapse
|
21
|
Wang Z, Shan Y, Yang Y, Wang T, Guo Z. MicroRNA-155 is upregulated in the placentas of patients with preeclampsia and affects trophoblast apoptosis by targeting SHH/GLi1/BCL2. Hum Exp Toxicol 2021; 40:439-451. [PMID: 32909851 DOI: 10.1177/0960327120954252] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The pathogenesis of preeclampsia (PE) is complicated and multiple risk factors have been associated with its occurrence. Still, the underlying molecular mechanisms involved in PE remain elusive. Aberrant apoptosis and insufficient invasion of trophoblasts have been observed and are considered vital pathological features in PE. Herein, we found that miR-155 can specifically degrade the mRNA of the Hedgehog ligand sonic hedgehog (SHH), using dual luciferase reporter assays. Quantitative real-time PCR found that administering miR-155 mimics or inhibitors could significantly decrease or increase the expression of SHH in the trophoblasts, respectively. The transcription levels of miR-155 in the placenta were higher in patients with PE compared to the levels in healthy pregnant women, as shown by quantitative real-time PCR. Serum levels of miR-155 could predict the diagnosis of PE by receiver operating characteristic curve analysis and diagnosis evaluation tests. A significant increase in apoptosis was observed after administering miR-155 in HTR8/SVneo cells cultured ex vivo, accompanied by reduced proliferation. Mechanistically, transcriptional activity and expression of GLi1 were also inhibited under treatment of miR-155, and could be recovered after supplying additional recombinant human SHH to primary trophoblasts from patients, as determined by luciferase activity assays and western blotting. We further found that inhibiting miR-155 increased the production of SHH and improved the phenotype in primary trophoblasts from patients with PE. Our data show that miR-155 regulates apoptosis of trophoblasts in PE, which has potential value for predicting PE risk and might be deemed as a therapeutic target for treating PE.
Collapse
Affiliation(s)
- Zhenpeng Wang
- Department of Gynecologic Oncologic, 117971The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yanhong Shan
- Department of Obstetrics, 117971The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yi Yang
- Center of Reproductive Medicine, Center of Prenatal Diagnosis, 117971The First Hospital of Jilin University, Changchun, Jilin, China
| | - Tianshu Wang
- Department of Obstetrics, 117971The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhiheng Guo
- Department of Obstetrics, 117971The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
22
|
Nasr T, Holderbaum AM, Chaturvedi P, Agarwal K, Kinney JL, Daniels K, Trisno SL, Ustiyan V, Shannon JM, Wells JM, Sinner D, Kalinichenko VV, Zorn AM. Disruption of a hedgehog-foxf1-rspo2 signaling axis leads to tracheomalacia and a loss of sox9+ tracheal chondrocytes. Dis Model Mech 2020; 14:dmm.046573. [PMID: 33328171 PMCID: PMC7875488 DOI: 10.1242/dmm.046573] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/09/2020] [Indexed: 12/14/2022] Open
Abstract
Congenital tracheomalacia, resulting from incomplete tracheal cartilage development, is a relatively common birth defect that severely impairs breathing in neonates. Mutations in the Hedgehog (HH) pathway and downstream Gli transcription factors are associated with tracheomalacia in patients and mouse models; however, the underlying molecular mechanisms are unclear. Using multiple HH/Gli mouse mutants including one that mimics Pallister-Hall Syndrome, we show that excessive Gli repressor activity prevents specification of tracheal chondrocytes. Lineage tracing experiments show that Sox9+ chondrocytes arise from HH-responsive splanchnic mesoderm in the fetal foregut that expresses the transcription factor Foxf1. Disrupted HH/Gli signaling results in 1) loss of Foxf1 which in turn is required to support Sox9+ chondrocyte progenitors and 2) a dramatic reduction in Rspo2, a secreted ligand that potentiates Wnt signaling known to be required for chondrogenesis. These results reveal a HH-Foxf1-Rspo2 signaling axis that governs tracheal cartilage development and informs the etiology of tracheomalacia.
Collapse
Affiliation(s)
- Talia Nasr
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267
| | - Andrea M Holderbaum
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267
| | - Praneet Chaturvedi
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229
| | - Kunal Agarwal
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229
| | - Jessica L Kinney
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229
| | - Keziah Daniels
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229
| | - Stephen L Trisno
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267
| | - Vladimir Ustiyan
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229
| | - John M Shannon
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229
| | - James M Wells
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267
| | - Debora Sinner
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229
| | - Vladimir V Kalinichenko
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229
| | - Aaron M Zorn
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45267
| |
Collapse
|
23
|
Cao P, Walker NM, Braeuer RR, Mazzoni-Putman S, Aoki Y, Misumi K, Wheeler DS, Vittal R, Lama VN. Loss of FOXF1 expression promotes human lung-resident mesenchymal stromal cell migration via ATX/LPA/LPA1 signaling axis. Sci Rep 2020; 10:21231. [PMID: 33277571 PMCID: PMC7718269 DOI: 10.1038/s41598-020-77601-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 11/09/2020] [Indexed: 02/08/2023] Open
Abstract
Forkhead box F1 (FOXF1) is a lung embryonic mesenchyme-associated transcription factor that demonstrates persistent expression into adulthood in mesenchymal stromal cells. However, its biologic function in human adult lung-resident mesenchymal stromal cells (LR-MSCs) remain to be elucidated. Here, we demonstrate that FOXF1 expression acts as a restraint on the migratory function of LR-MSCs via its role as a novel transcriptional repressor of autocrine motility-stimulating factor Autotaxin (ATX). Fibrotic human LR-MSCs demonstrated lower expression of FOXF1 mRNA and protein, compared to non-fibrotic controls. RNAi-mediated FOXF1 silencing in LR-MSCs was associated with upregulation of key genes regulating proliferation, migration, and inflammatory responses and significantly higher migration were confirmed in FOXF1-silenced LR-MSCs by Boyden chamber. ATX is a secreted lysophospholipase D largely responsible for extracellular lysophosphatidic acid (LPA) production, and was among the top ten upregulated genes upon Affymetrix analysis. FOXF1-silenced LR-MSCs demonstrated increased ATX activity, while mFoxf1 overexpression diminished ATX expression and activity. The FOXF1 silencing-induced increase in LR-MSC migration was abrogated by genetic and pharmacologic targeting of ATX and LPA1 receptor. Chromatin immunoprecipitation analyses identified three putative FOXF1 binding sites in the 1.5 kb ATX promoter which demonstrated transcriptional repression of ATX expression. Together these findings identify FOXF1 as a novel transcriptional repressor of ATX and demonstrate that loss of FOXF1 promotes LR-MSC migration via the ATX/LPA/LPA1 signaling axis.
Collapse
Affiliation(s)
- Pengxiu Cao
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 1500 W Medical Center Drive, 3916 Taubman Center, Ann Arbor, MI, 48109-0360, USA
| | - Natalie M Walker
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 1500 W Medical Center Drive, 3916 Taubman Center, Ann Arbor, MI, 48109-0360, USA
| | - Russell R Braeuer
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 1500 W Medical Center Drive, 3916 Taubman Center, Ann Arbor, MI, 48109-0360, USA
| | - Serina Mazzoni-Putman
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 1500 W Medical Center Drive, 3916 Taubman Center, Ann Arbor, MI, 48109-0360, USA
| | - Yoshiro Aoki
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 1500 W Medical Center Drive, 3916 Taubman Center, Ann Arbor, MI, 48109-0360, USA
| | - Keizo Misumi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 1500 W Medical Center Drive, 3916 Taubman Center, Ann Arbor, MI, 48109-0360, USA
| | - David S Wheeler
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 1500 W Medical Center Drive, 3916 Taubman Center, Ann Arbor, MI, 48109-0360, USA
| | - Ragini Vittal
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 1500 W Medical Center Drive, 3916 Taubman Center, Ann Arbor, MI, 48109-0360, USA
| | - Vibha N Lama
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, 1500 W Medical Center Drive, 3916 Taubman Center, Ann Arbor, MI, 48109-0360, USA.
| |
Collapse
|
24
|
Little MH. Patterning a Ureter Is All in the Stroma. J Am Soc Nephrol 2020; 31:2231-2232. [PMID: 32999037 DOI: 10.1681/asn.2020071055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Affiliation(s)
- Melissa H Little
- Cell Biology Theme, Murdoch Children's Research Institute, Parkville, Victoria, Australia .,Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia .,Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
25
|
Sallam M, Palakkan AA, Mills CG, Tarnick J, Elhendawi M, Marson L, Davies JA. Differentiation of a Contractile, Ureter-Like Tissue, from Embryonic Stem Cell-Derived Ureteric Bud and Ex Fetu Mesenchyme. J Am Soc Nephrol 2020; 31:2253-2262. [PMID: 32826325 DOI: 10.1681/asn.2019101075] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND There is intense interest in replacing kidneys from stem cells. It is now possible to produce, from embryonic or induced pluripotent stem cells, kidney organoids that represent immature kidneys and display some physiologic functions. However, current techniques have not yet resulted in renal tissue with a ureter, which would be needed for engineered kidneys to be clinically useful. METHODS We used a published sequence of growth factors and drugs to induce mouse embryonic stem cells to differentiate into ureteric bud tissue. We characterized isolated engineered ureteric buds differentiated from embryonic stem cells in three-dimensional culture and grafted them into ex fetu mouse kidney rudiments. RESULTS Engineered ureteric buds branched in three-dimensional culture and expressed Hoxb7, a transcription factor that is part of a developmental regulatory system and a ureteric bud marker. When grafted into the cortex of ex fetu kidney rudiments, engineered ureteric buds branched and induced nephron formation; when grafted into peri-Wolffian mesenchyme, still attached to a kidney rudiment or in isolation, they did not branch but instead differentiated into multilayer ureter-like epithelia displaying robust expression of the urothelial marker uroplakin. This engineered ureteric bud tissue also organized the mesenchyme into smooth muscle that spontaneously contracted, with a period a little slower than that of natural ureteric peristalsis. CONCLUSIONS Mouse embryonic stem cells can be differentiated into ureteric bud cells. Grafting those UB-like structures into peri-Wolffian mesenchyme of cultured kidney rudiments can induce production of urothelium and organize the mesenchyme to produce rhythmically contracting smooth muscle layers. This development may represent a significant step toward the goal of renal regeneration.
Collapse
Affiliation(s)
- May Sallam
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh, UK .,Human Anatomy and Embryology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Anwar A Palakkan
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh, UK
| | | | - Julia Tarnick
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh, UK
| | - Mona Elhendawi
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh, UK.,Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Lorna Marson
- Edinburgh Transplant Centre, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Jamie A Davies
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
26
|
Gilliam EA, Schlieve CR, Fowler KL, Rea JN, Schall KA, Huang S, Spence JR, Grikscheit TC. Grading TESI: Crypt and villus formation in tissue-engineered small intestine alters with stem/progenitor cell source. Am J Physiol Gastrointest Liver Physiol 2020; 319:G261-G279. [PMID: 32597710 DOI: 10.1152/ajpgi.00387.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The small intestine has a remarkable ability to enhance its absorptive and digestive surface area through the formation of villi, a process known as villification. We sought to learn whether developing mouse and human tissue-engineered small intestine (TESI) followed known developmental biology routes to villification, such as Sonic hedgehog (SHH)/Indian hedgehog (IHH) and bone morphogenetic protein 4 (BMP4)/forkhead box F1 (FOXF1) signaling to identify targets to enhance the development of TESI. After generating TESI from prenatal and postnatal stem cell sources, we evaluated the effect of cell source derivation on villification with a grading scheme to approximate developmental stage. χ2 analysis compared the prevalence of TESI grade from each stem cell source. RNAscope probes detected genes known to direct villification and the development of the crypt-villus axis in mouse and human development. These were compared in TESI derived from various pluripotent and progenitor cell donor cell types as well as native human fetal and postnatal tissues. Prenatal and pluripotent cell sources form mature villus and crypt-like structures more frequently than postnatal donor sources, and there are alternate routes to villus formation. Human TESI recapitulates epithelial to mesenchymal crosstalk of several genes identified in development, with fetal and pluripotent donor-derived TESI arriving at villus formation following described developmental patterns. However, postnatal TESI is much less likely to form complete villus-crypt patterns and demonstrates alternate SHH/IHH and BMP4/FOXF1 signaling patterns. Grading TESI and other cellular constructs may assist discoveries to support future human therapies.NEW & NOTEWORTHY The small intestine can enhance its absorptive and digestive surface area through a process known as villification. Tissue-engineered small intestine achieves mature villification at varying levels of success between differing sources. We have developed a consistent grading schema of morphology and characterized it across multiple developmental pathways, allowing objective comparison between differing constructs and sources.
Collapse
Affiliation(s)
- Elizabeth A Gilliam
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California.,Department of Surgery, Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California
| | - Christopher R Schlieve
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California.,Department of Surgery, Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California
| | - Kathryn L Fowler
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California
| | - Jessica N Rea
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California.,Department of Surgery, Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California
| | - Kathy A Schall
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California.,Department of Surgery, Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California
| | - Sha Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jason R Spence
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan.,Program of Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Tracy C Grikscheit
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute at Children's Hospital Los Angeles, Los Angeles, California.,Department of Surgery, Division of Pediatric Surgery, Children's Hospital Los Angeles, Los Angeles, California.,Keck Medical School, University of Southern California, Los Angeles, California
| |
Collapse
|
27
|
Jackson AR, Ching CB, McHugh KM, Becknell B. Roles for urothelium in normal and aberrant urinary tract development. Nat Rev Urol 2020; 17:459-468. [PMID: 32647226 DOI: 10.1038/s41585-020-0348-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2020] [Indexed: 12/11/2022]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUTs) represent the leading cause of chronic kidney disease and end-stage kidney disease in children. Increasing evidence points to critical roles for the urothelium in the developing urinary tract and in the genesis of CAKUTs. The involvement of the urothelium in patterning the urinary tract is supported by evidence that CAKUTs can arise as a result of abnormal urothelial development. Emerging evidence indicates that congenital urinary tract obstruction triggers urothelial remodelling that stabilizes the obstructed kidney and limits renal injury. Finally, the diagnostic potential of radiological findings and urinary biomarkers derived from the urothelium of patients with CAKUTs might aid their contribution to clinical care.
Collapse
Affiliation(s)
- Ashley R Jackson
- Nephrology and Urology Research Affinity Group, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Christina B Ching
- Nephrology and Urology Research Affinity Group, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Division of Pediatric Urology, Department of Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kirk M McHugh
- Nephrology and Urology Research Affinity Group, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Anatomy, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Brian Becknell
- Nephrology and Urology Research Affinity Group, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA. .,Center for Clinical and Translational Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA. .,Nephrology Division, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
28
|
Shen G, Ren H, Shang Q, Zhao W, Zhang Z, Yu X, Tang K, Tang J, Yang Z, Liang D, Jiang X. Foxf1 knockdown promotes BMSC osteogenesis in part by activating the Wnt/β-catenin signalling pathway and prevents ovariectomy-induced bone loss. EBioMedicine 2020; 52:102626. [PMID: 31981979 PMCID: PMC6992955 DOI: 10.1016/j.ebiom.2020.102626] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/22/2019] [Accepted: 12/03/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Forkhead box protein f1 (Foxf1) is associated with cell differentiation, and may be a key player in bone homoeostasis. However, the effect of Foxf1 on osteogenesis of bone marrow-derived mesenchymal stem cells (BMSCs) and ovariectomy-induced bone loss, as well as its clinical implications, is unknown. METHODS By quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) and western blotting, we assayed Foxf1 expression in bone tissue, BMSCs, and bone marrow-derived macrophages (BMMs), derived from ovariectomised (OVX) mice, and during osteogenic differentiation and osteoclast differentiation. Using a loss-of-function approach (small interfering RNA [siRNA]-mediated knockdown) in vitro, we examined whether Foxf1 regulates osteoblast differentiation of BMSCs via the Wnt/β-catenin signalling pathway. Furthermore, we assessed the anabolic effect of Foxf1 knockdown (siFoxf1) in OVX mice in vivo. We also assayed the expression of Foxf1 in bone tissue derived from postmenopausal osteoporosis (PMOP) patients and its link with bone mineral density (BMD). Finally, we examined the effect of Foxf1 knockdown on the osteoblastic differentiation of human BMSCs. FINDINGS Foxf1 expression was significantly increased in bone extract and BMSCs from OVX mice and gradually decreased during osteoblastic differentiation of BMSCs but did not differ significantly in OVX mouse-derived BMMs or during osteoclast differentiation. In vitro, Foxf1 knockdown markedly increased the expression of osteoblast specific genes, alkaline phosphatase (ALP) activity, and mineralisation. Moreover, siFoxf1 activated the Wnt/β-catenin signalling pathway. The siFoxf1-induced increase in osteogenic differentiation was partly rescued by inhibitor of Wnt signalling (DKK1). In OVX mice, Foxf1 siRNA significantly reduced bone loss by enhancing bone formation. Foxf1 expression levels negatively correlated with reduced bone mass and bone formation in bone tissue from PMOP patients. Finally, Foxf1 knockdown significantly promoted osteogenesis by human BMSCs. INTERPRETATION Our findings indicate that Foxf1 knockdown promotes BMSC osteogenesis and prevents OVX-induced bone loss. Therefore, Foxf1 has potential as a biomarker of osteogenesis and may be a therapeutic target for PMOP.
Collapse
Affiliation(s)
- Gengyang Shen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Hui Ren
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qi Shang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Wenhua Zhao
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhida Zhang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiang Yu
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Kai Tang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jingjing Tang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhidong Yang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - De Liang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xiaobing Jiang
- Department of Spinal Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Lingnan Medical Research Center of Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
29
|
Regulation of bone morphogenetic protein 4 on epithelial tissue. J Cell Commun Signal 2020; 14:283-292. [PMID: 31912367 DOI: 10.1007/s12079-019-00537-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/14/2019] [Indexed: 01/01/2023] Open
Abstract
Epithelial tissues provide tissue barriers and specialize in organs and glands. When epithelial homeostasis is physiologically or pathologically stimulated, epithelial cells produce mesenchymal cells through the epithelial-mesenchymal transition, forming new tissues, promoting the cure of diseases or leading to illness. A variety of cytokines are involved in the regulation of epithelial cell differentiation. Bone morphogenetic proteins (BMPs), especially the bone morphogenetic protein 4 (BMP4) has a variety of biological functions and plays a prominent role in the regulation of epithelial cell differentiation. BMP4 is an important regulatory factor of a series of life activities in vertebrates, which is also related to cell proliferation, differentiation and mobility, it also has relation with tumor development. This paper mainly reviews the mechanism of BMP4's regulation on epithelial tissues, as well as its effect on the growth, differentiation, benign lesions and malignant lesions of epithelial tissues, and expounds the function of BMP4 in epithelial tissues, to provide theoretical support for the research on reducing epithelial diseases.
Collapse
|
30
|
Weiss AC, Bohnenpoll T, Kurz J, Blank P, Airik R, Lüdtke TH, Kleppa MJ, Deuper L, Kaiser M, Mamo TM, Costa R, von Hahn T, Trowe MO, Kispert A. Delayed onset of smooth muscle cell differentiation leads to hydroureter formation in mice with conditional loss of the zinc finger transcription factor gene Gata2 in the ureteric mesenchyme. J Pathol 2019; 248:452-463. [PMID: 30916783 DOI: 10.1002/path.5270] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/03/2019] [Accepted: 03/21/2019] [Indexed: 12/22/2022]
Abstract
The establishment of the peristaltic machinery of the ureter is precisely controlled to cope with the onset of urine production in the fetal kidney. Retinoic acid (RA) has been identified as a signal that maintains the mesenchymal progenitors of the contractile smooth muscle cells (SMCs), while WNTs, SHH, and BMP4 induce their differentiation. How the activity of the underlying signalling pathways is controlled in time, space, and quantity to activate coordinately the SMC programme is poorly understood. Here, we provide evidence that the Zn-finger transcription factor GATA2 is involved in this crosstalk. In mice, Gata2 is expressed in the undifferentiated ureteric mesenchyme under control of RA signalling. Conditional deletion of Gata2 by a Tbx18cre driver results in hydroureter formation at birth, associated with a loss of differentiated SMCs. Analysis at earlier stages and in explant cultures revealed that SMC differentiation is not abrogated but delayed and that dilated ureters can partially regain peristaltic activity when relieved of urine pressure. Molecular analysis identified increased RA signalling as one factor contributing to the delay in SMC differentiation, possibly caused by reduced direct transcriptional activation of Cyp26a1, which encodes an RA-degrading enzyme. Our study identified GATA2 as a feedback inhibitor of RA signalling important for precise onset of ureteric SMC differentiation, and suggests that in a subset of cases of human congenital ureter dilatations, temporary relief of urine pressure may ameliorate the differentiation status of the SMC coat. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Anna-Carina Weiss
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Tobias Bohnenpoll
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Jennifer Kurz
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Patrick Blank
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Rannar Airik
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Timo H Lüdtke
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Marc-Jens Kleppa
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Lena Deuper
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Marina Kaiser
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Tamrat M Mamo
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Rui Costa
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany.,Klinik für Gastroenterologie, Hepatologie und Endokrinologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Thomas von Hahn
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany.,Klinik für Gastroenterologie, Hepatologie und Endokrinologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Mark-Oliver Trowe
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Andreas Kispert
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| |
Collapse
|
31
|
Huang L, Tjakra M, Luo D, Wen L, Lei D, Wang J, Yin T, Zhang X, Huang J, Wang Y, Wang G. FoxF1 is Required for Ciliogenesis and Distribution of Sonic Hedgehog Signaling Components in Cilium. Curr Mol Med 2019; 19:326-334. [PMID: 30950350 DOI: 10.2174/1566524019666190405115420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 03/21/2019] [Accepted: 03/27/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND In vertebrates, cilium is crucial for Hedgehog signaling transduction. Forkhead box transcriptional factor FoxF1 is reported to be associated with Sonic Hedgehog (Shh) signaling in many cases. However, the role of FoxF1 in cilium remains unknown. Here, we showed an essential role of FoxF1 in the regulation of ciliogenesis and in the distribution of Shh signaling components in cilium. METHODS NIH/3T3 cells were serum starved for 24h to induce cilium. Meanwhile, shRNA was used to knockdown the FoxF1 expression in the cells and CRISPR/Cas9 was used to generate the FoxF1 zebrafish mutant. The mRNA and protein expression of indicated genes were detected by the qRT-PCR and western blot, respectively. Immunofluorescence staining was performed to detect the cilium and Shh components distribution. RESULTS FoxF1 knockdown decreased the cilium length in NIH/3T3 cells. Meanwhile, the disruption of FoxF1 function inhibited the expression of cilium-related genes and caused an abnormal distribution of Shh components in the cilium. Furthermore, homozygous FoxF1 mutants exhibited defective development of pronephric cilium in early zebrafish embryos. CONCLUSION Together, our data illustrated that FoxF1 is required for ciliogenesis in vitro and in vivo and for the proper localization of Shh signaling components in cilium.
Collapse
Affiliation(s)
- Lu Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Chongqing University, Chongqing, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing University, Chongqing, China.,Bioengineering College of Chongqing University, Chongqing, China
| | - Marco Tjakra
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Chongqing University, Chongqing, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing University, Chongqing, China.,Bioengineering College of Chongqing University, Chongqing, China
| | - Desha Luo
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Chongqing University, Chongqing, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing University, Chongqing, China.,Bioengineering College of Chongqing University, Chongqing, China
| | - Lin Wen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Chongqing University, Chongqing, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing University, Chongqing, China.,Bioengineering College of Chongqing University, Chongqing, China
| | - Daoxi Lei
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Chongqing University, Chongqing, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing University, Chongqing, China.,Bioengineering College of Chongqing University, Chongqing, China
| | - Jinxuan Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Chongqing University, Chongqing, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing University, Chongqing, China.,Bioengineering College of Chongqing University, Chongqing, China
| | - Tieying Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Chongqing University, Chongqing, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing University, Chongqing, China.,Bioengineering College of Chongqing University, Chongqing, China
| | - Xiaojuan Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Chongqing University, Chongqing, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing University, Chongqing, China.,Bioengineering College of Chongqing University, Chongqing, China
| | - Junli Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Chongqing University, Chongqing, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing University, Chongqing, China.,Bioengineering College of Chongqing University, Chongqing, China
| | - Yeqi Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Chongqing University, Chongqing, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing University, Chongqing, China.,Bioengineering College of Chongqing University, Chongqing, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, Chongqing University, Chongqing, China.,State and Local Joint Engineering Laboratory for Vascular Implants, Chongqing University, Chongqing, China.,Bioengineering College of Chongqing University, Chongqing, China
| |
Collapse
|
32
|
Abstract
The ability to explant and then maintain embryonic tissues in organ culture makes it feasible to study the growth and differentiation of whole organs, or parts or combinations of them, in three dimensions. Moreover, the possible effects of biochemical manipulations or mutations can be explored by visualizing a growing organ. The mammalian renal tract comprises the kidney, ureter, and urinary bladder, and the focus of this chapter is organ culture of the embryonic mouse ureter in serum-free defined medium. Over the culture period, rudiments grow in length, smooth muscle differentiates, and the ureters then undergo peristalsis in a proximal to distal direction.
Collapse
Affiliation(s)
- Filipa M Lopes
- Faculty of Biology Medicine and Health, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, University of Manchester, Manchester, UK.
| | - Adrian S Woolf
- Faculty of Biology Medicine and Health, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, University of Manchester, Manchester, UK.,Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
33
|
Aydoğdu N, Rudat C, Trowe MO, Kaiser M, Lüdtke TH, Taketo MM, Christoffels VM, Moon A, Kispert A. TBX2 and TBX3 act downstream of canonical WNT signaling in patterning and differentiation of the mouse ureteric mesenchyme. Development 2018; 145:145/23/dev171827. [PMID: 30478225 DOI: 10.1242/dev.171827] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/24/2018] [Indexed: 12/20/2022]
Abstract
The organized array of smooth muscle cells (SMCs) and fibroblasts in the walls of visceral tubular organs arises by patterning and differentiation of mesenchymal progenitors surrounding the epithelial lumen. Here, we show that the TBX2 and TBX3 transcription factors have novel and required roles in regulating these processes in the murine ureter. Co-expression of TBX2 and TBX3 in the inner mesenchymal region of the developing ureter requires canonical WNT signaling. Loss of TBX2/TBX3 in this region disrupts activity of two crucial drivers of the SMC program, Foxf1 and BMP4 signaling, resulting in decreased SMC differentiation and increased extracellular matrix. Transcriptional profiling and chromatin immunoprecipitation experiments revealed that TBX2/TBX3 directly repress expression of the WNT antagonists Dkk2 and Shisa2, the BMP antagonist Bmper and the chemokine Cxcl12 These findings suggest that TBX2/TBX3 are effectors of canonical WNT signaling in the ureteric mesenchyme that promote SMC differentiation by maintaining BMP4 and WNT signaling in the inner region, while restricting CXCL12 signaling to the outer layer of fibroblast-fated mesenchyme.
Collapse
Affiliation(s)
- Nurullah Aydoğdu
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Carsten Rudat
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Mark-Oliver Trowe
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Marina Kaiser
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Timo H Lüdtke
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Makoto Mark Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Vincent M Christoffels
- Department of Anatomy, Embryology and Physiology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Anne Moon
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Clinic, Danville PA 17822, USA.,Departments of Pediatrics and Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Andreas Kispert
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| |
Collapse
|
34
|
Huang M, He H, Belenkaya T, Lin X. Multiple roles of epithelial heparan sulfate in stomach morphogenesis. J Cell Sci 2018; 131:jcs.210781. [PMID: 29700203 DOI: 10.1242/jcs.210781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 04/19/2018] [Indexed: 12/12/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) have been shown to regulate various developmental processes. However, the function of heparan sulfate (HS) during the development of mammalian stomach has not been characterized yet. Here, we investigate the role of epithelial HS in embryonic stomach by examining mice deficient in the glycosyltransferase gene Ext1 We show that HS exhibits a specific and dynamic expression pattern in mouse embryonic stomach. Depletion of the epithelial HS leads to stomach hypoplasia, with phenotypic differences in the gastric mucosa between the forestomach and hindstomach. In the posterior stomach, HS depletion disrupts glandular stomach patterning and cytodifferentiation via attenuation of Fgf signaling activity. Inhibition of Fgf signaling in vitro recapitulates the patterning defect. Ligand and carbohydrate engagement assay (LACE) reveals a diminished assembly of Fgf10 and Fgfr2b in the mutant. In the anterior stomach, loss of epithelial HS leads to stratification and differentiation defects of the multilayered squamous epithelium, along with reduced Hh and Bmp signaling activity. Our data demonstrate that epithelial HS plays multiple roles in regulating mammalian stomach morphogenesis in a regional-specific manner.
Collapse
Affiliation(s)
- Meina Huang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200438, China.,State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hua He
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tatyana Belenkaya
- Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Xinhua Lin
- State Key Laboratory of Genetic Engineering, Institute of Genetics, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200438, China .,Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
35
|
Retinoic acid signaling maintains epithelial and mesenchymal progenitors in the developing mouse ureter. Sci Rep 2017; 7:14803. [PMID: 29093497 PMCID: PMC5665985 DOI: 10.1038/s41598-017-14790-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 10/16/2017] [Indexed: 12/27/2022] Open
Abstract
The differentiated cell types of the mature ureter arise from the distal ureteric bud epithelium and its surrounding mesenchyme. Uncommitted epithelial cells first become intermediate cells from which both basal and superficial cells develop. Mesenchymal progenitors give rise to separated layers of adventitial fibrocytes, smooth muscle cells and lamina propria fibrocytes. How progenitor expansion and differentiation are balanced is poorly understood. Here, we addressed the role of retinoic acid (RA) signaling in these programs. Using expression analysis of components and target genes, we show that pathway activity is restricted to the mesenchymal and epithelial progenitor pools. Inhibition of RA signaling in ureter explant cultures resulted in tissue hypoplasia with a relative expansion of smooth muscle cells at the expense of lamina propria fibroblasts in the mesenchyme, and of superficial cells at the expense of intermediate cells in the ureteric epithelium. Administration of RA led to a slight reduction of smooth muscle cells, and almost completely prevented differentiation of intermediate cells into basal and superficial cells. We identified cellular programs and transcriptional targets of RA signaling that may account for this activity. We conclude that RA signaling is required and sufficient to maintain mesenchymal and epithelial progenitors in early ureter development.
Collapse
|