1
|
Kispert A. Ureter development and associated congenital anomalies. Nat Rev Nephrol 2025; 21:366-382. [PMID: 40164775 DOI: 10.1038/s41581-025-00951-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2025] [Indexed: 04/02/2025]
Abstract
Malformations of the ureter are among the most common birth defects in humans. Although some of these anomalies are asymptomatic, others are clinically relevant, causing perinatal lethality or progressing to kidney failure in childhood. The genetic causes and developmental aetiology of ureteral anomalies are difficult to study in humans; however, embryological and genetic analyses in the mouse have provided insights into the complex developmental programmes that govern ureter formation from simple tissue primordia, and the pathological consequences that result from disruption of these programmes. Abnormalities in the formation of the nephric duct and ureteric bud lead to changes in the number of ureters (and kidneys), whereas the formation of ectopic ureteric buds, failure of the nephric duct to target the cloaca or failure of the distal ureter to mature underlie vesicoureteral reflux, ureter ectopia, ureterocoele and subsequent hydroureter. Alterations in ureter specification, early growth or cyto-differentiation programmes have now also been associated with various forms of perinatal hydroureter and hydronephrosis as a consequence of functional obstruction. The characterization of cellular processes and molecular drivers of ureterogenesis in the mouse may not only aid understanding of the aetiology of human ureteral anomalies, improve prognostication and benefit the development of therapeutic strategies, but may also prove important for efforts to generate a bioartificial organ.
Collapse
Affiliation(s)
- Andreas Kispert
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
2
|
Fogarty A, Jia S, Wilbourne J, DuPuis C, Zhao F. Crucial roles of mesenchymal Gata2 in murine epididymal development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645498. [PMID: 40196482 PMCID: PMC11974812 DOI: 10.1101/2025.03.26.645498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Androgens drive the morphogenesis and differentiation of the Wolffian duct (WD) into the epididymis, an essential organ for male reproduction, by binding to the androgen receptor (AR). However, it remains unclear whether other transcriptional programs operate beyond the central androgen/AR signaling in promoting WD development. We discovered that mesenchyme-specific deletion of the transcription factor Gata2 resulted in defective epididymal coiling in the corpus and caudal regions. The defective coiling in the absence of mesenchymal Gata2 did not result from androgen signaling deficiency, as there were no abnormalities in testicular morphology, androgen production, or AR/Ar expression, and dihydrotestosterone supplementation did not restore epididymal coiling in cultured WDs. Instead, Gata2 deletion reduced the expression of the mesenchyme-derived factor Inhba and epithelial proliferation, both of which play critical roles in epididymal coiling. The epididymal defect persisted into adulthood, with the uncoiled corpus and caudal epididymis exhibiting abnormal epithelial morphology and lumen environments, resulting in an unfavorable environment for sperm storage. Our results demonstrate the androgen-independent role of mesenchymal GATA2 in promoting epididymal development through Inhba induction and highlight the importance of proper fetal development in male reproduction.
Collapse
Affiliation(s)
- Allyssa Fogarty
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
- Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Shuai Jia
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jillian Wilbourne
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Claire DuPuis
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Fei Zhao
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
- Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
3
|
Tian X, Yao W, Tan J, Hu Z, Liu J. gdf11 is required for pronephros/cloaca development through targeting TGF-β signaling. Sci Rep 2025; 15:8052. [PMID: 40055488 PMCID: PMC11889093 DOI: 10.1038/s41598-025-92571-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/28/2025] [Indexed: 05/13/2025] Open
Abstract
The kidney is a vital organ responsible for removing toxins, producing urine, and regulating homeostasis. Developmental defects in the kidney lead to various congenital abnormalities that impair renal function. Gdf11, a member of the transforming growth factor β family, is associated with numerous renal abnormalities. In the early developmental stage, the pronephric duct and hindgut open into the cloaca, and Gdf11 shows significant expression in the hindgut of mice. However, the molecular and cellular roles of gdf11 in kidney and cloaca organogenesis remain unclear. Our study revealed that pronephros and cloaca formation were significantly disrupted upon gdf11 deletion or knockdown in zebrafish. Additionally, we found that the TGF-β pathway acts downstream of Gdf11 in promoting pronephros and cloaca development. Treatment with a TGF-β small molecule activator partially rescued the pronephros and cloaca developmental defects observed in gdf11-/- mutants. In summary, our findings provide strong evidence of a critical link between pronephros/cloaca formation and TGF-β signaling mediated by Gdf11. Our study also provides new insight into diseases related to renal and cloaca development.
Collapse
Affiliation(s)
- Xinning Tian
- Department of Cardiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Wantao Yao
- Department of Cardiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Jin Tan
- Department of Cardiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Zhangle Hu
- Department of Cardiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| | - Jingwen Liu
- Department of Cardiology, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- School of Basic Medicine, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
4
|
Strosahl J, Ye K, Pazdro R. Novel insights into the pleiotropic health effects of growth differentiation factor 11 gained from genome-wide association studies in population biobanks. BMC Genomics 2024; 25:837. [PMID: 39237910 PMCID: PMC11378601 DOI: 10.1186/s12864-024-10710-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 08/14/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor-β (TGF-β) superfamily that has gained considerable attention over the last decade for its observed ability to reverse age-related deterioration of multiple tissues, including the heart. Yet as many researchers have struggled to confirm the cardioprotective and anti-aging effects of GDF11, the topic has grown increasingly controversial, and the field has reached an impasse. We postulated that a clearer understanding of GDF11 could be gained by investigating its health effects at the population level. METHODS AND RESULTS We employed a comprehensive strategy to interrogate results from genome-wide association studies in population Biobanks. Interestingly, phenome-wide association studies (PheWAS) of GDF11 tissue-specific cis-eQTLs revealed associations with asthma, immune function, lung function, and thyroid phenotypes. Furthermore, PheWAS of GDF11 genetic variants confirmed these results, revealing similar associations with asthma, immune function, lung function, and thyroid health. To complement these findings, we mined results from transcriptome-wide association studies, which uncovered associations between predicted tissue-specific GDF11 expression and the same health effects identified from PheWAS analyses. CONCLUSIONS In this study, we report novel relationships between GDF11 and disease, namely asthma and hypothyroidism, in contrast to its formerly assumed role as a rejuvenating factor in basic aging and cardiovascular health. We propose that these associations are mediated through the involvement of GDF11 in inflammatory signaling pathways. Taken together, these findings provide new insights into the health effects of GDF11 at the population level and warrant future studies investigating the role of GDF11 in these specific health conditions.
Collapse
Affiliation(s)
- Jessica Strosahl
- Department of Nutritional Sciences, University of Georgia, 305 Sanford Drive, Athens, GA, 30602, USA
| | - Kaixiong Ye
- Department of Genetics, University of Georgia, Athens, GA, 30602, USA
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
| | - Robert Pazdro
- Department of Nutritional Sciences, University of Georgia, 305 Sanford Drive, Athens, GA, 30602, USA.
| |
Collapse
|
5
|
Gao G, Zhou Z. Isthmin-1: A critical regulator of branching morphogenesis and metanephric mesenchyme condensation during early kidney development. Bioessays 2024; 46:e2300189. [PMID: 38161234 DOI: 10.1002/bies.202300189] [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: 10/02/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
Isthmin-1 (Ism1) was first described to be syn-expressed with Fgf8 in Xenopus. However, its biological role has not been elucidated until recent years. Despite of accumulated evidence that Ism1 participates in angiogenesis, tumor invasion, macrophage apoptosis, and glucose metabolism, the cognate receptors for Ism1 remain largely unknown. Ism1 deficiency in mice results in renal agenesis (RA) with a transient loss of Gdnf transcription and impaired mesenchyme condensation at E11.5. Ism1 binds to and activates Integrin α8β1 to positively regulate Gdnf/Ret signaling, thus promoting mesenchyme condensation and ureteric epithelium branching morphogenesis. Here, we propose the hypothesis underlying the mechanism by which Ism1 regulates branching morphogenesis during early kidney development.
Collapse
Affiliation(s)
- Ge Gao
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhongjun Zhou
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Reproductive Medical Center, The University of Hong Kong - Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
6
|
Kraler S, Balbi C, Vdovenko D, Lapikova-Bryhinska T, Camici GG, Liberale L, Bonetti N, Canestro CD, Burger F, Roth A, Carbone F, Vassalli G, Mach F, Bhasin S, Wenzl FA, Muller O, Räber L, Matter CM, Montecucco F, Lüscher TF, Akhmedov A. Circulating GDF11 exacerbates myocardial injury in mice and associates with increased infarct size in humans. Cardiovasc Res 2023; 119:2729-2742. [PMID: 37742057 PMCID: PMC10757585 DOI: 10.1093/cvr/cvad153] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/18/2023] [Accepted: 09/04/2023] [Indexed: 09/25/2023] Open
Abstract
AIMS The heart rejuvenating effects of circulating growth differentiation factor 11 (GDF11), a transforming growth factor-β superfamily member that shares 90% homology with myostatin (MSTN), remains controversial. Here, we aimed to probe the role of GDF11 in acute myocardial infarction (MI), a frequent cause of heart failure and premature death during ageing. METHODS AND RESULTS In contrast to endogenous Mstn, myocardial Gdf11 declined during the course of ageing and was particularly reduced following ischaemia/reperfusion (I/R) injury, suggesting a therapeutic potential of GDF11 signalling in MI. Unexpectedly, boosting systemic Gdf11 by recombinant GDF11 delivery (0.1 mg/kg body weight over 30 days) prior to myocardial I/R augmented myocardial infarct size in C57BL/6 mice irrespective of their age, predominantly by accelerating pro-apoptotic signalling. While intrinsic cardioprotective signalling pathways remained unaffected by high circulating GDF11, targeted transcriptomics and immunomapping studies focusing on GDF11-associated downstream targets revealed attenuated Nkx2-5 expression confined to CD105-expressing cells, with pro-apoptotic activity, as assessed by caspase-3 levels, being particularly pronounced in adjacent cells, suggesting an indirect effect. By harnessing a highly specific and validated liquid chromatography-tandem mass spectrometry-based assay, we show that in prospectively recruited patients with MI circulating GDF11 but not MSTN levels incline with age. Moreover, GDF11 levels were particularly elevated in those at high risk for adverse outcomes following the acute event, with circulating GDF11 emerging as an independent predictor of myocardial infarct size, as estimated by standardized peak creatine kinase-MB levels. CONCLUSION Our data challenge the initially reported heart rejuvenating effects of circulating GDF11 and suggest that high levels of systemic GDF11 exacerbate myocardial injury in mice and humans alike. Persistently high GDF11 levels during ageing may contribute to the age-dependent loss of cardioprotective mechanisms and thus poor outcomes of elderly patients following acute MI.
Collapse
Affiliation(s)
- Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, Zurich CH-8952, Switzerland
| | - Carolina Balbi
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, Zurich CH-8952, Switzerland
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Institute, EOC, Lugano, Switzerland
- Laboratories for Translational Research, EOC, Bellinzona, Switzerland
| | - Daria Vdovenko
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, Zurich CH-8952, Switzerland
| | | | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, Zurich CH-8952, Switzerland
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genova—Italian Cardiovascular Network, Genoa, Italy
| | - Nicole Bonetti
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, Zurich CH-8952, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Candela Diaz Canestro
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, Zurich CH-8952, Switzerland
| | - Fabienne Burger
- Division of Cardiology, Foundation for Medical Research, University of Geneva, Geneva, Switzerland
| | - Aline Roth
- Division of Cardiology, Foundation for Medical Research, University of Geneva, Geneva, Switzerland
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genova—Italian Cardiovascular Network, Genoa, Italy
| | - Giuseppe Vassalli
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, Zurich CH-8952, Switzerland
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Institute, EOC, Lugano, Switzerland
- Laboratories for Translational Research, EOC, Bellinzona, Switzerland
| | - François Mach
- Division of Cardiology, Foundation for Medical Research, University of Geneva, Geneva, Switzerland
| | - Shalender Bhasin
- Research Program in Men's Health: Aging and Metabolism, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, USA
| | - Florian A Wenzl
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, Zurich CH-8952, Switzerland
| | - Olivier Muller
- Department of Cardiology, University Hospital of Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Lorenz Räber
- Department of Cardiology, Inselspital Bern, Bern, Switzerland
| | - Christian M Matter
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, Zurich CH-8952, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genova—Italian Cardiovascular Network, Genoa, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, Zurich CH-8952, Switzerland
- Royal Brompton and Harefield Hospitals and Imperial College and Kings College, London, UK
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, Zurich CH-8952, Switzerland
| |
Collapse
|
7
|
Shao Y, Wang Y, Xu J, Yuan Y, Xing D. Growth differentiation factor 11: A new hope for the treatment of cardiovascular diseases. Cytokine Growth Factor Rev 2023; 71-72:82-93. [PMID: 37414617 DOI: 10.1016/j.cytogfr.2023.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/08/2023]
Abstract
Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor-β superfamily that has garnered significant attention due to its anti-cardiac aging properties. Many studies have revealed that GDF11 plays an indispensable role in the onset of cardiovascular diseases (CVDs). Consequently, it has emerged as a potential target and novel therapeutic agent for CVD treatment. However, currently, no literature reviews comprehensively summarize the research on GDF11 in the context of CVDs. Therefore, herein, we comprehensively described GDF11's structure, function, and signaling in various tissues. Furthermore, we focused on the latest findings concerning its involvement in CVD development and its potential for clinical translation as a CVD treatment. We aim to provide a theoretical basis for the prospects and future research directions of the GDF11 application regarding CVDs.
Collapse
Affiliation(s)
- Yingchun Shao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Yanhong Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Jiazhen Xu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Yang Yuan
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
8
|
Gao G, Li X, Jiang Z, Osorio L, Tang YL, Yu X, Jin G, Zhou Z. Isthmin-1 (Ism1) modulates renal branching morphogenesis and mesenchyme condensation during early kidney development. Nat Commun 2023; 14:2378. [PMID: 37185772 PMCID: PMC10130008 DOI: 10.1038/s41467-023-37992-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
The outgrowth of epithelial bud followed by reiterated bifurcations during renal development is driven by the ligand-receptor interactions between the epithelium and the surrounding mesenchyme. Here, by exploring ligand-receptor interactions in E10.5 and E11.5 kidneys by single cell RNA-seq, we find that Isthmin1 (Ism1), a secreted protein, resembles Gdnf expression and modulates kidney branching morphogenesis. Mice deficient for Ism1 exhibit defective ureteric bud bifurcation and impaired metanephric mesenchyme condensation in E11.5 embryos, attributable to the compromised Gdnf/Ret signaling, ultimately leading to renal agenesis and hypoplasia/dysplasia. By HRP-induced proximity labelling, we further identify integrin α8β1 as a receptor of Ism1 in E11.5 kidney and demonstrate that Ism1 promoted cell-cell adhesion through interacting with Integrin α8β1, the receptor whose activation is responsible for Gdnf expression and mesenchyme condensation. Taken together, our work reveals Ism1 as a critical regulator of cell-cell interaction that modulates Gdnf/Ret signaling during early kidney development.
Collapse
Affiliation(s)
- Ge Gao
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
- School of Biomedical Sciences, LKS Faculty of medicine, The University of Hong Kong, Hong Kong, China
| | - Xiaoping Li
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, 510630, Guangdong, China
| | - Zhixin Jiang
- School of Biomedical Sciences, LKS Faculty of medicine, The University of Hong Kong, Hong Kong, China
| | - Liliana Osorio
- School of Biomedical Sciences, LKS Faculty of medicine, The University of Hong Kong, Hong Kong, China
| | - Ying Lam Tang
- School of Biomedical Sciences, LKS Faculty of medicine, The University of Hong Kong, Hong Kong, China
| | - Xueqing Yu
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
| | - Guoxiang Jin
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China
| | - Zhongjun Zhou
- Guangdong Cardiovascular Institute, Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China.
- School of Biomedical Sciences, LKS Faculty of medicine, The University of Hong Kong, Hong Kong, China.
- Reproductive Medical Center, The University of Hong Kong - Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
9
|
Lian J, Walker RG, D'Amico A, Vujic A, Mills MJ, Messemer KA, Mendello KR, Goldstein JM, Leacock KA, Epp S, Stimpfl EV, Thompson TB, Wagers AJ, Lee RT. Functional substitutions of amino acids that differ between GDF11 and GDF8 impact skeletal development and skeletal muscle. Life Sci Alliance 2023; 6:e202201662. [PMID: 36631218 PMCID: PMC9834663 DOI: 10.26508/lsa.202201662] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 01/13/2023] Open
Abstract
Growth differentiation factor 11 (GDF11) and GDF8 (MSTN) are closely related TGF-β family proteins that interact with nearly identical signaling receptors and antagonists. However, GDF11 appears to activate SMAD2/3 more potently than GDF8 in vitro and in vivo. The ligands possess divergent structural properties, whereby substituting unique GDF11 amino acids into GDF8 enhanced the activity of the resulting chimeric GDF8. We investigated potentially distinct endogenous activities of GDF11 and GDF8 in vivo by genetically modifying their mature signaling domains. Full recoding of GDF8 to that of GDF11 yielded mice lacking GDF8, with GDF11 levels ∼50-fold higher than normal, and exhibiting modestly decreased muscle mass, with no apparent negative impacts on health or survival. Substitution of two specific amino acids in the fingertip region of GDF11 with the corresponding GDF8 residues resulted in prenatal axial skeletal transformations, consistent with Gdf11-deficient mice, without apparent perturbation of skeletal or cardiac muscle development or homeostasis. These experiments uncover distinctive features between the GDF11 and GDF8 mature domains in vivo and identify a specific requirement for GDF11 in early-stage skeletal development.
Collapse
Affiliation(s)
- John Lian
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Ryan G Walker
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Andrea D'Amico
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Ana Vujic
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Melanie J Mills
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Kathleen A Messemer
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Kourtney R Mendello
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Jill M Goldstein
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Krystynne A Leacock
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Soraya Epp
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Emma V Stimpfl
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Thomas B Thompson
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, OH, USA
| | - Amy J Wagers
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Joslin Diabetes Center, Boston, MA, USA
- Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, USA
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| |
Collapse
|
10
|
Lee SJ, Lehar A, Rydzik R, Youngstrom DW, Bhasin S, Liu Y, Germain-Lee EL. Functional replacement of myostatin with GDF-11 in the germline of mice. Skelet Muscle 2022; 12:7. [PMID: 35287700 PMCID: PMC8922734 DOI: 10.1186/s13395-022-00290-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/04/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Myostatin (MSTN) is a transforming growth factor-ß superfamily member that acts as a major regulator of skeletal muscle mass. GDF-11, which is highly related to MSTN, plays multiple roles during embryonic development, including regulating development of the axial skeleton, kidneys, nervous system, and pancreas. As MSTN and GDF-11 share a high degree of amino acid sequence identity, behave virtually identically in cell culture assays, and utilize similar regulatory and signaling components, a critical question is whether their distinct biological functions result from inherent differences in their abilities to interact with specific regulatory and signaling components or whether their distinct biological functions mainly reflect their differing temporal and spatial patterns of expression. METHODS We generated and characterized mice in which we precisely replaced in the germline the portion of the Mstn gene encoding the mature C-terminal peptide with the corresponding region of Gdf11. RESULTS In mice homozygous for the knock-in allele, all of the circulating MSTN protein was replaced with GDF-11, resulting in ~ 30-40-fold increased levels of circulating GDF-11. Male mice homozygous for the knock-in allele had slightly decreased muscle weights, slightly increased weight gain in response to a high-fat diet, slightly increased plasma cholesterol and HDL levels, and significantly decreased bone density and bone mass, whereas female mice were mostly unaffected. CONCLUSIONS GDF-11 appears to be capable of nearly completely functionally replacing MSTN in the control of muscle mass. The developmental and physiological consequences of replacing MSTN with GDF-11 are strikingly limited.
Collapse
Affiliation(s)
- Se-Jin Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA. .,Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, USA.
| | - Adam Lehar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Renata Rydzik
- Department of Orthopaedic Surgery, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Daniel W Youngstrom
- Department of Orthopaedic Surgery, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Shalender Bhasin
- Brigham Research Assay Core Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yewei Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Emily L Germain-Lee
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, USA.,Department of Reconstructive Sciences, Center for Regenerative Medicine and Skeletal Development, University of Connecticut School of Dental Medicine, Farmington, CT, USA.,Division of Endocrinology & Diabetes and Center for Rare Bone Disorders, Connecticut Children's, Farmington, CT, USA
| |
Collapse
|
11
|
Mohammadi B, Esmaeilizadeh Z, Rajabibazl M, Ghaderian SMH, Omrani MD, Fazeli Z. Preconditioning of human adipose tissue-derived mesenchymal stem cells with HEK293-coditioned media can influence on the expression of BMP2, BMP6 and BMP11: Potential application in the treatment of renal lesions. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
12
|
Esmaeilizadeh Z, Mohammadi B, Rajabibazl M, Ghaderian SMH, Omrani MD, Fazeli Z. Expression Analysis of GDNF/RET Signaling Pathway in Human AD-MSCs Grown in HEK 293 Conditioned Medium (HEK293-CM). Cell Biochem Biophys 2020; 78:531-539. [PMID: 32803668 DOI: 10.1007/s12013-020-00936-z] [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: 10/01/2019] [Accepted: 08/05/2020] [Indexed: 10/23/2022]
Abstract
Mesenchymal stem cells have been considered as the suitable source for the repair of kidney lesions. The study and identification of novel approaches could improve the efficiency of these cells in the recovery of kidney. In the present study, the effect of HEK 293 conditioned medium (HEK293-CM) was evaluated on the expression of GDNF/RET signaling pathway and their downstream genes in the human adipose-derived mesenchymal stem cells (AD-MSCs). For this purpose, the human AD-MSCs were cultured in the medium containing HEK293-CM. After the RNA extraction and cDNA synthesis, the expression level of GFRA1, GDNF, SPRY1, ETV4, ETV5, and CRLF1 genes were determined by SYBR Green Real time PCR. The obtained results indicated that the GDNF and GFRA1 expression enhanced in the AD-MSCs following treatment with 10% HEK293-CM-5%FBS as compared to the untreated AD-MSCs. These results were consistent with the decreased expression of SPRY1. The significant increased expression of ETV4, ETV5, and CRLF1 genes also showed that HEK293-CM activated the GDNF/RET signaling pathway in the AD-MSCs (P < 0.05). The obtained data suggested that the treatment with HEK293-CM activated the GDNF/RET signaling pathway in the human AD-MSCs.
Collapse
Affiliation(s)
- Zahra Esmaeilizadeh
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahar Mohammadi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Rajabibazl
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mir Davood Omrani
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Fazeli
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
Frohlich J, Vinciguerra M. Candidate rejuvenating factor GDF11 and tissue fibrosis: friend or foe? GeroScience 2020; 42:1475-1498. [PMID: 33025411 PMCID: PMC7732895 DOI: 10.1007/s11357-020-00279-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Growth differentiation factor 11 (GDF11 or bone morphogenetic protein 11, BMP11) belongs to the transforming growth factor-β superfamily and is closely related to other family member-myostatin (also known as GDF8). GDF11 was firstly identified in 2004 due to its ability to rejuvenate the function of multiple organs in old mice. However, in the past few years, the heralded rejuvenating effects of GDF11 have been seriously questioned by many studies that do not support the idea that restoring levels of GDF11 in aging improves overall organ structure and function. Moreover, with increasing controversies, several other studies described the involvement of GDF11 in fibrotic processes in various organ setups. This review paper focuses on the GDF11 and its pro- or anti-fibrotic actions in major organs and tissues, with the goal to summarize our knowledge on its emerging role in regulating the progression of fibrosis in different pathological conditions, and to guide upcoming research efforts.
Collapse
Affiliation(s)
- Jan Frohlich
- International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic
| | - Manlio Vinciguerra
- International Clinical Research Center, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic.
- Institute for Liver and Digestive Health, Division of Medicine, University College London (UCL), London, UK.
| |
Collapse
|
14
|
Kang JS, Yang YR. Circulating plasma factors involved in rejuvenation. Aging (Albany NY) 2020; 12:23394-23408. [PMID: 33197235 PMCID: PMC7746393 DOI: 10.18632/aging.103933] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 07/30/2020] [Indexed: 12/17/2022]
Abstract
Aging is defined as a time-dependent functional decline that occurs in many physiological systems. This decline is the primary risk factor for prominent human pathologies such as cancer, metabolic disorders, cardiovascular disorders, and neurodegenerative diseases. Aging and age-related diseases have multiple causes. Parabiosis experiments, in which the circulatory systems of young and old mice were surgically joined, revealed that young plasma counteracts aging and rejuvenates organs in old mice, suggesting the existence of rejuvenating factors that become less abundant with aging. Diverse approaches have identified a large number of plasma proteins whose levels differ significantly between young and old mice, as well as numerous rejuvenating factors that reverse aged-related impairments in multiple tissues. These observations suggest that increasing the levels of key rejuvenating factors could promote restorative biological processes or inhibit pathological degeneration. Inspired by such findings, several companies have begun selling “young blood transfusions,” and others have tested young plasma as a treatment for Alzheimer’s disease. Here, we summarize the current findings regarding rejuvenating factors.
Collapse
Affiliation(s)
- Jae Sook Kang
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Yong Ryoul Yang
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| |
Collapse
|
15
|
Similar sequences but dissimilar biological functions of GDF11 and myostatin. Exp Mol Med 2020; 52:1673-1693. [PMID: 33077875 PMCID: PMC8080601 DOI: 10.1038/s12276-020-00516-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 12/27/2022] Open
Abstract
Growth differentiation factor 11 (GDF11) and myostatin (MSTN) are closely related TGFβ family members that are often believed to serve similar functions due to their high homology. However, genetic studies in animals provide clear evidence that they perform distinct roles. While the loss of Mstn leads to hypermuscularity, the deletion of Gdf11 results in abnormal skeletal patterning and organ development. The perinatal lethality of Gdf11-null mice, which contrasts with the long-term viability of Mstn-null mice, has led most research to focus on utilizing recombinant GDF11 proteins to investigate the postnatal functions of GDF11. However, the reported outcomes of the exogenous application of recombinant GDF11 proteins are controversial partly because of the different sources and qualities of recombinant GDF11 used and because recombinant GDF11 and MSTN proteins are nearly indistinguishable due to their similar structural and biochemical properties. Here, we analyze the similarities and differences between GDF11 and MSTN from an evolutionary point of view and summarize the current understanding of the biological processing, signaling, and physiological functions of GDF11 and MSTN. Finally, we discuss the potential use of recombinant GDF11 as a therapeutic option for a wide range of medical conditions and the possible adverse effects of GDF11 inhibition mediated by MSTN inhibitors.
Collapse
|
16
|
Hart CG, Karimi-Abdolrezaee S. Bone morphogenetic proteins: New insights into their roles and mechanisms in CNS development, pathology and repair. Exp Neurol 2020; 334:113455. [PMID: 32877654 DOI: 10.1016/j.expneurol.2020.113455] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/18/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023]
Abstract
Bone morphogenetic proteins (BMPs) are a highly conserved and diverse family of proteins that play essential roles in various stages of development including the formation and patterning of the central nervous system (CNS). Bioavailability and function of BMPs are regulated by input from a plethora of transcription factors and signaling pathways. Intriguingly, recent literature has uncovered novel roles for BMPs in regulating homeostatic and pathological responses in the adult CNS. Basal levels of BMP ligands and receptors are widely expressed in the adult brain and spinal cord with differential expression patterns across CNS regions, cell types and subcellular locations. Recent evidence indicates that several BMP isoforms are transiently or chronically upregulated in the aged or pathological CNS. Genetic knockout and pharmacological studies have elucidated that BMPs regulate several aspects of CNS injury and repair including cell survival and differentiation, reactive astrogliosis and glial scar formation, axon regeneration, and myelin preservation and repair. Several BMP isoforms can be upregulated in the injured or diseased CNS simultaneously yet exert complementary or opposing effects on the endogenous cell responses after injury. Emerging studies also show that dysregulation of BMPs is associated with various CNS pathologies. Interestingly, modulation of BMPs can lead to beneficial or detrimental effects on CNS injury and repair mechanisms in a ligand, temporally or spatially specific manner, which reflect the complexity of BMP signaling. Given the significance of BMPs in neurodevelopment, a better understanding of their role in the context of injury may provide new therapeutic targets for the pathologic CNS. This review will provide a timely overview on the foundation and recent advancements in knowledge regarding the role and mechanisms of BMP signaling in the developing and adult CNS, and their implications in pathological responses and repair processes after injury or diseases.
Collapse
Affiliation(s)
- Christopher G Hart
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
17
|
Martens H, Hennies I, Getwan M, Christians A, Weiss AC, Brand F, Gjerstad AC, Christians A, Gucev Z, Geffers R, Seeman T, Kispert A, Tasic V, Bjerre A, Lienkamp SS, Haffner D, Weber RG. Rare heterozygous GDF6 variants in patients with renal anomalies. Eur J Hum Genet 2020; 28:1681-1693. [PMID: 32737436 PMCID: PMC7784874 DOI: 10.1038/s41431-020-0678-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/05/2020] [Accepted: 06/15/2020] [Indexed: 01/22/2023] Open
Abstract
Although over 50 genes are known to cause renal malformation if mutated, the underlying genetic basis, most easily identified in syndromic cases, remains unsolved in most patients. In search of novel causative genes, whole-exome sequencing in a patient with renal, i.e., crossed fused renal ectopia, and extrarenal, i.e., skeletal, eye, and ear, malformations yielded a rare heterozygous variant in the GDF6 gene encoding growth differentiation factor 6, a member of the BMP family of ligands. Previously, GDF6 variants were reported to cause pleiotropic defects including skeletal, e.g., vertebral, carpal, tarsal fusions, and ocular, e.g., microphthalmia and coloboma, phenotypes. To assess the role of GDF6 in the pathogenesis of renal malformation, we performed targeted sequencing in 193 further patients identifying rare GDF6 variants in two cases with kidney hypodysplasia and extrarenal manifestations. During development, gdf6 was expressed in the pronephric tubule of Xenopus laevis, and Gdf6 expression was observed in the ureteric tree of the murine kidney by RNA in situ hybridization. CRISPR/Cas9-derived knockout of Gdf6 attenuated migration of murine IMCD3 cells, an effect rescued by expression of wild-type but not mutant GDF6, indicating affected variant function regarding a fundamental developmental process. Knockdown of gdf6 in Xenopus laevis resulted in impaired pronephros development. Altogether, we identified rare heterozygous GDF6 variants in 1.6% of all renal anomaly patients and 5.4% of renal anomaly patients additionally manifesting skeletal, ocular, or auricular abnormalities, adding renal hypodysplasia and fusion to the phenotype spectrum of GDF6 variant carriers and suggesting an involvement of GDF6 in nephrogenesis.
Collapse
Affiliation(s)
- Helge Martens
- Department of Human Genetics, Hannover Medical School, 30625, Hannover, Germany
| | - Imke Hennies
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, 30625, Hannover, Germany
| | - Maike Getwan
- Department of Medicine, Renal Division, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany.,Institute of Anatomy and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057, Zurich, Switzerland
| | - Anne Christians
- Department of Human Genetics, Hannover Medical School, 30625, Hannover, Germany
| | - Anna-Carina Weiss
- Institute of Molecular Biology, Hannover Medical School, 30625, Hannover, Germany
| | - Frank Brand
- Department of Human Genetics, Hannover Medical School, 30625, Hannover, Germany
| | - Ann Christin Gjerstad
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, 0424, Oslo, Norway
| | - Arne Christians
- Department of Neuropathology, Institute of Pathology, Hannover Medical School, 30625, Hannover, Germany
| | - Zoran Gucev
- Medical Faculty Skopje, University Children's Hospital, 1000, Skopje, North Macedonia
| | - Robert Geffers
- Genome Analytics Research Group, Helmholtz Centre for Infection Research, 38124, Braunschweig, Germany
| | - Tomáš Seeman
- Department of Paediatrics and Transplantation Center, University Hospital Motol, Second Faculty of Medicine, Charles University, 150 06, Prague, Czech Republic
| | - Andreas Kispert
- Institute of Molecular Biology, Hannover Medical School, 30625, Hannover, Germany
| | - Velibor Tasic
- Medical Faculty Skopje, University Children's Hospital, 1000, Skopje, North Macedonia
| | - Anna Bjerre
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, 0424, Oslo, Norway
| | - Soeren S Lienkamp
- Department of Medicine, Renal Division, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79110, Freiburg, Germany.,Institute of Anatomy and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057, Zurich, Switzerland
| | - Dieter Haffner
- Department of Pediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, 30625, Hannover, Germany
| | - Ruthild G Weber
- Department of Human Genetics, Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
18
|
Li L, Wei X, Wang D, Lv Z, Geng X, Li P, Lu J, Wang K, Wang X, Sun J, Cao X, Wei L. Positive Effects of a Young Systemic Environment and High Growth Differentiation Factor 11 Levels on Chondrocyte Proliferation and Cartilage Matrix Synthesis in Old Mice. Arthritis Rheumatol 2020; 72:1123-1133. [PMID: 32067417 DOI: 10.1002/art.41230] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/06/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To investigate the effects of a young systemic environment and growth differentiation factor 11 (GDF-11) on aging cartilage. METHODS A heterochronic parabiosis model (2-month-old mouse and 12-month-old mouse [Y/O]), an isochronic parabiosis model (12-month-old mouse and 12-month-old mouse [O/O]), and 12-month-old mice alone (O) were evaluated. Knee joints and chondrocytes from old mice were examined by radiography, histology, cell proliferation assays, immunohistochemistry, Western blotting, and quantitative reverse transcriptase-polymerase chain reaction 16 weeks after parabiosis surgery. GDF-11 was injected into 12-month-old mouse joints daily for 16 weeks. Cartilage degeneration, cell proliferation, and osteoarthritis-related gene expression were evaluated. RESULTS Osteoarthritis Research Society International scores in old mice were significantly lower in the Y/O group than in the O/O and O groups (both P < 0.05). The percentage of 5-ethynyl-2'-deoxyuridine-positive chondrocytes in old mice was significantly higher in the Y/O group than in the other groups (P < 0.05). Type II collagen (CII) and SOX9 messenger RNA levels differed in cartilage from old mice in the Y/O group compared to the O/O and O groups (both P < 0.05). RUNX-2, CX, and matrix metalloproteinase 13 levels were significantly lower in cartilage from old mice in the Y/O group compared to the O/O and O groups (both P < 0.05). Similar results were obtained for protein expression levels and after GDF-11 treatment in vitro and in vivo. Phosphorylated Smad2/3 (pSmad2/3) levels were higher in the recombinant GDF-11-treated group than in the control group. CONCLUSION A young systemic environment promotes chondrocyte proliferation and cartilage matrix synthesis in old mice. GDF-11, a "young factor," contributes to these effects through the up-regulation of pSmad2/3.
Collapse
Affiliation(s)
- Lu Li
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaochun Wei
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Dongming Wang
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhi Lv
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiang Geng
- Shanxi Health Vocational College, Jinzhong, China
| | - Pengcui Li
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jiangong Lu
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Kaihang Wang
- Subsidiary High School of Taiyuan Normal University, Taiyuan, China
| | - Xiaohu Wang
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jian Sun
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaoming Cao
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Lei Wei
- Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
19
|
May RD, Frauchiger DA, Albers CE, Tekari A, Benneker LM, Klenke FM, Hofstetter W, Gantenbein B. Application of Cytokines of the Bone Morphogenetic Protein (BMP) Family in Spinal Fusion - Effects on the Bone, Intervertebral Disc and Mesenchymal Stromal Cells. Curr Stem Cell Res Ther 2020; 14:618-643. [PMID: 31455201 PMCID: PMC7040507 DOI: 10.2174/1574888x14666190628103528] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Abstract
Low back pain is a prevalent socio-economic burden and is often associated with damaged or degenerated intervertebral discs (IVDs). When conservative therapy fails, removal of the IVD (discectomy), followed by intersomatic spinal fusion, is currently the standard practice in clinics. The remaining space is filled with an intersomatic device (cage) and with bone substitutes to achieve disc height compensation and bone fusion. As a complication, in up to 30% of cases, spinal non-fusions result in a painful pseudoarthrosis. Bone morphogenetic proteins (BMPs) have been clinically applied with varied outcomes. Several members of the BMP family, such as BMP2, BMP4, BMP6, BMP7, and BMP9, are known to induce osteogenesis. Questions remain on why hyper-physiological doses of BMPs do not show beneficial effects in certain patients. In this respect, BMP antagonists secreted by mesenchymal cells, which might interfere with or block the action of BMPs, have drawn research attention as possible targets for the enhancement of spinal fusion or the prevention of non-unions. Examples of these antagonists are noggin, gremlin1 and 2, chordin, follistatin, BMP3, and twisted gastrulation. In this review, we discuss current evidence of the osteogenic effects of several members of the BMP family on osteoblasts, IVD cells, and mesenchymal stromal cells. We consider in vitro and in vivo studies performed in human, mouse, rat, and rabbit related to BMP and BMP antagonists in the last two decades. We give insights into the effects that BMP have on the ossification of the spine. Furthermore, the benefits, pitfalls, and possible safety concerns using these cytokines for the improvement of spinal fusion are discussed.
Collapse
Affiliation(s)
- Rahel Deborah May
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | | | - Christoph Emmanuel Albers
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| | - Adel Tekari
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Lorin Michael Benneker
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| | - Frank Michael Klenke
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| | - Willy Hofstetter
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Benjamin Gantenbein
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| |
Collapse
|
20
|
GDF11 promotes osteogenesis as opposed to MSTN, and follistatin, a MSTN/GDF11 inhibitor, increases muscle mass but weakens bone. Proc Natl Acad Sci U S A 2020; 117:4910-4920. [PMID: 32071240 PMCID: PMC7060712 DOI: 10.1073/pnas.1916034117] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
MSTN, a member of the TGF-β family, has been widely shown to suppress muscle growth, leading to an intense effort being directed at targeting MSTN to treat patients with muscle loss. GDF11 is another TGF-β family member closely related to MSTN, but its postnatal function is less clear. Using conditional knockout techniques, we show that GDF11 enhances bone mass in contrast to MSTN, emphasizing that MSTN/GDF11 inhibitors, such as FST, can induce adverse effects on bone through GDF11 inhibition. Because most MSTN inhibitors also inhibit GDF11 due to the high sequence similarity between MSTN and GDF11, our findings suggest that their opposing roles must be carefully considered when developing MSTN inhibitors for clinical applications. Growth and differentiation factor 11 (GDF11) and myostatin (MSTN) are closely related transforming growth factor β (TGF-β) family members, but their biological functions are quite distinct. While MSTN has been widely shown to inhibit muscle growth, GDF11 regulates skeletal patterning and organ development during embryogenesis. Postnatal functions of GDF11, however, remain less clear and controversial. Due to the perinatal lethality of Gdf11 null mice, previous studies used recombinant GDF11 protein to prove its postnatal function. However, recombinant GDF11 and MSTN proteins share nearly identical biochemical properties, and most GDF11-binding molecules have also been shown to bind MSTN, generating the possibility that the effects mediated by recombinant GDF11 protein actually reproduce the endogenous functions of MSTN. To clarify the endogenous functions of GDF11, here, we focus on genetic studies and show that Gdf11 null mice, despite significantly down-regulating Mstn expression, exhibit reduced bone mass through impaired osteoblast (OB) and chondrocyte (CH) maturations and increased osteoclastogenesis, while the opposite is observed in Mstn null mice that display enhanced bone mass. Mechanistically, Mstn deletion up-regulates Gdf11 expression, which activates bone morphogenetic protein (BMP) signaling pathway to enhance osteogenesis. Also, mice overexpressing follistatin (FST), a MSTN/GDF11 inhibitor, exhibit increased muscle mass accompanied by bone fractures, unlike Mstn null mice that display increased muscle mass without fractures, indicating that inhibition of GDF11 impairs bone strength. Together, our findings suggest that GDF11 promotes osteogenesis in contrast to MSTN, and these opposing roles of GDF11 and MSTN must be considered to avoid the detrimental effect of GDF11 inhibition when developing MSTN/GDF11 inhibitors for therapeutic purposes.
Collapse
|
21
|
Liu W, Zhou L, Xue H, Li H, Yuan Q. Growth differentiation factor 11 impairs titanium implant healing in the femur and leads to mandibular bone loss. J Periodontol 2020; 91:1203-1212. [PMID: 31983062 DOI: 10.1002/jper.19-0247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 12/24/2019] [Accepted: 12/25/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND Growth differentiation factor 11 (GDF11), a secreted member of the transforming growth factor-β superfamily, has recently been suggested as an anti-aging factor that declines with age in the bloodstream, and restoration of the youthful level by administration of its recombinant protein could reverse age-related dysfunctions. However, its effects on titanium implant osseointegration and mandibular bone during aging remain unknown. METHODS Two-month-old and 18-month-old C57BL male mice were given daily intraperitoneal injections of recombinant GDF11 (rGDF11) or vehicle for 6 weeks. Experimental titanium implants were inserted into femurs on the fourth week. Inhibition of GDF11 function was achieved by GDF11 antibody. Implant-bearing femurs were subjected to histomorphometric analysis and biomechanical evaluation. Mandibles were scanned with micro-CT and decalcified for histological measurements. RESULTS In both young adult and aged mice, supraphysiologic GDF11 leads to a significantly decreased bone-to-implant contact ratio (BIC) and peri-implant bone volume/total volume (BV/TV) at the histologic level and reduced resistance at the biomechanical level, indicating weakened implant fixation. Moreover, rGDF11 administration resulted in less trabecular bone volume and thinner cortical thickness in the mandible, which was further compromised in the old animals. In contrast, inhibition of GDF11 improved peri-implant bone healing in old mice. CONCLUSIONS Rather than functioning as a rejuvenating factor, exogenous GDF11 negatively affects not only titanium implant healing but also mandibular bone in both young and old mice. In contrast, neutralization of endogenous GDF11 has positive effects on implant fixation in aged mice.
Collapse
Affiliation(s)
- Weiqing Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Liyan Zhou
- Dept. of Implant, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hanxiao Xue
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Hanshi Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
22
|
Kuure S, Sariola H. Mouse Models of Congenital Kidney Anomalies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:109-136. [PMID: 32304071 DOI: 10.1007/978-981-15-2389-2_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are common birth defects, which cause the majority of chronic kidney diseases in children. CAKUT covers a wide range of malformations that derive from deficiencies in embryonic kidney and lower urinary tract development, including renal aplasia, hypodysplasia, hypoplasia, ectopia, and different forms of ureter abnormalities. The majority of the genetic causes of CAKUT remain unknown. Research on mutant mice has identified multiple genes that critically regulate renal differentiation. The data generated from this research have served as an excellent resource to identify the genetic bases of human kidney defects and have led to significantly improved diagnostics. Furthermore, genetic data from human CAKUT studies have also revealed novel genes regulating kidney differentiation.
Collapse
Affiliation(s)
- Satu Kuure
- GM-Unit, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland. .,Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. .,Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Hannu Sariola
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Paediatric Pathology, HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
23
|
Goldstein JM, Valido A, Lewandowski JP, Walker RG, Mills MJ, Messemer KA, Besseling P, Lee KH, Wattrus SJ, Cho M, Lee RT, Wagers AJ. Variation in zygotic CRISPR/Cas9 gene editing outcomes generates novel reporter and deletion alleles at the Gdf11 locus. Sci Rep 2019; 9:18613. [PMID: 31819086 PMCID: PMC6901511 DOI: 10.1038/s41598-019-54766-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 11/19/2019] [Indexed: 01/20/2023] Open
Abstract
Recent advances in CRISPR/Cas gene editing technology have significantly expanded the possibilities and accelerated the pace of creating genetically engineered animal models. However, CRISPR/Cas-based strategies designed to precisely edit the genome can often yield unintended outcomes. Here, we report the use of zygotic CRISPR/Cas9 injections to generate a knock-in GFP reporter mouse at the Gdf11 locus. Phenotypic and genomic characterization of founder animals from these injections revealed a subset that contained the correct targeting event and exhibited GFP expression that, within the hematopoietic system, was restricted predominantly to lymphoid cells. Yet, in another subset of founder mice, we detected aberrant integration events at the target site that dramatically and inaccurately shifted hematopoietic GFP expression from the lymphoid to the myeloid lineage. Additionally, we recovered multiple Gdf11 deletion alleles that modified the C-terminus of the GDF11 protein. When bred to homozygosity, most of these alleles recapitulated skeletal phenotypes reported previously for Gdf11 knockout mice, suggesting that these represent null alleles. However, we also recovered one Gdf11 deletion allele that encodes a novel GDF11 variant protein ("GDF11-WE") predicted to contain two additional amino acids (tryptophan (W) and glutamic acid (E)) at the C-terminus of the mature ligand. Unlike the other Gdf11 deletion alleles recovered in this study, homozygosity for the Gdf11WE allele did not phenocopy Gdf11 knockout skeletal phenotypes. Further investigation using in vivo and in vitro approaches demonstrated that GDF11-WE retains substantial physiological function, indicating that GDF11 can tolerate at least some modifications of its C-terminus and providing unexpected insights into its biochemical activities. Altogether, our study confirms that one-step zygotic injections of CRISPR/Cas gene editing complexes provide a quick and powerful tool to generate gene-modified mouse models. Moreover, our findings underscore the critical importance of thorough characterization and validation of any modified alleles generated by CRISPR, as unintended on-target effects that fail to be detected by simple PCR screening can produce substantially altered phenotypic readouts.
Collapse
Affiliation(s)
- Jill M Goldstein
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
- Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, 02215, USA
| | - Austin Valido
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Jordan P Lewandowski
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Ryan G Walker
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Melanie J Mills
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Kathleen A Messemer
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
- Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, 02215, USA
| | - Paul Besseling
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Kyu Ha Lee
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Samuel J Wattrus
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Miook Cho
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
- Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, 02215, USA
| | - Richard T Lee
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA
| | - Amy J Wagers
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA.
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
- Paul F. Glenn Center for the Biology of Aging, Harvard Medical School, Boston, MA, 02215, USA.
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, 02215, USA.
| |
Collapse
|
24
|
Activin Is Superior to BMP7 for Efficient Maintenance of Human iPSC-Derived Nephron Progenitors. Stem Cell Reports 2019; 13:322-337. [PMID: 31378669 PMCID: PMC6700502 DOI: 10.1016/j.stemcr.2019.07.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 07/01/2019] [Accepted: 07/01/2019] [Indexed: 12/29/2022] Open
Abstract
Kidney formation is regulated by the balance between maintenance and differentiation of nephron progenitor cells (NPCs). Now that directed differentiation of NPCs from human induced pluripotent stem cells (iPSCs) can be achieved, maintenance and propagation of NPCs in vitro should be beneficial for regenerative medicine. Although WNT and FGF signals were previously shown to be essential for NPC propagation, the requirement for BMP/TGFβ signaling remains controversial. Here we reveal that activin has superior effects to BMP7 on maintenance efficiency of human iPSC-derived NPCs. Activin expanded ITGA8+/PDGFRA-/SIX2-GFP+ NPCs by 5-fold per week at 80%-90% efficiency, and the propagated cells possessed robust capacity for nephron formation both in vitro and in vivo. The expanded cells also maintained their nephron-forming potential after freezing. Furthermore, the protocol was applicable to multiple non-GFP-tagged iPSC lines. Thus, our activin-based protocol will be applicable to a variety of research fields including disease modeling and drug screening.
Collapse
|
25
|
Liu X, Zhang Q, Fan C, Tian J, Liu X, Li G. GDF11 upregulation independently predicts shorter overall-survival of uveal melanoma. PLoS One 2019; 14:e0214073. [PMID: 30883611 PMCID: PMC6422293 DOI: 10.1371/journal.pone.0214073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 03/06/2019] [Indexed: 01/10/2023] Open
Abstract
Growth differentiation factor 11 (GDF11), is a member of the transforming growth factor-beta (TGF-β) superfamily and bone morphogenetic protein (BMP) subfamily. In this study, we aimed to assess the expression profile of GDF11, its prognostic value in terms of OS, as well as the potential mechanisms leading to its dysregulation in uveal melanoma. A retrospective study was conducted using our primary data and genetic, clinicopathological and overall survival (OS) data from the Cancer Genome Atlas-Uveal Melanoma (TCGA-UVM). Results showed that GDF11 expression was significantly higher in tumor tissues compared with that in adjacent normal tissues. High GDF11 expression was associated with uveal melanoma in advanced stages (IV), epithelioid cell dominant subtype, as well as extrascleral extension. Univariate analysis showed that older age, epithelioid cell dominant, with extrascleral extension and increased GDF11 expression were associated with unfavorable OS. Multivariate analysis confirmed that GDF11 expression was an independent prognostic indicator of unfavorable OS (HR: 1.704, 95%CI: 1.143–2.540, p = 0.009), after adjustment of age, histological subtypes and extrascleral extension. Among the 80 cases of uveal melanoma, only 3 cases had low-level copy gain (+1) and 2 cases had heterozygous loss (-1). No somatic mutations, including SNPs and small INDELs were observed in GDF11 DNA. The methylation of these four CpG sites had weakly (cg22950598 and cg23689080), moderately (cg09890930), or strongly (cg05511733) negative correlation with GDF11 expression. In addition, the patients with high methylation of these four sites had significantly better OS compared to the group with low methylation. Based on these findings, we infer that methylation modulated GDF11 expression might be a valuable prognostic biomarker regarding OS in uveal melanoma.
Collapse
Affiliation(s)
- Xun Liu
- Department of Ophthalmology, Weifang People's Hospital, Weifang, China
| | - Qinghai Zhang
- Department of ICU, Weifang People's Hospital, Weifang, China
| | - Chuanfeng Fan
- Department of Ophthalmology, Taian Aier Eye Hospital, Taian, China
| | - Jie Tian
- Department of Ophthalmology, Weifang People's Hospital, Weifang, China
| | - Xinchang Liu
- Department of Ophthalmology, Weifang People's Hospital, Weifang, China
| | - Guofeng Li
- Department of Ophthalmology, Weifang People's Hospital, Weifang, China
- * E-mail:
| |
Collapse
|
26
|
Zhang H, Bagherie-Lachidan M, Badouel C, Enderle L, Peidis P, Bremner R, Kuure S, Jain S, McNeill H. FAT4 Fine-Tunes Kidney Development by Regulating RET Signaling. Dev Cell 2019; 48:780-792.e4. [PMID: 30853441 DOI: 10.1016/j.devcel.2019.02.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 12/06/2018] [Accepted: 02/01/2019] [Indexed: 12/27/2022]
Abstract
FAT4 mutations lead to several human diseases that disrupt the normal development of the kidney. However, the underlying mechanism remains elusive. In studying the duplex kidney phenotypes observed upon deletion of Fat4 in mice, we have uncovered an interaction between the atypical cadherin FAT4 and RET, a tyrosine kinase receptor essential for kidney development. Analysis of kidney development in Fat4-/- kidneys revealed abnormal ureteric budding and excessive RET signaling. Removal of one copy of the RET ligand Gdnf rescues Fat4-/- kidney development, supporting the proposal that loss of Fat4 hyperactivates RET signaling. Conditional knockout analyses revealed a non-autonomous role for Fat4 in regulating RET signaling. Mechanistically, we found that FAT4 interacts with RET through extracellular cadherin repeats. Importantly, expression of FAT4 perturbs the assembly of the RET-GFRA1-GDNF complex, reducing RET signaling. Thus, FAT4 interacts with RET to fine-tune RET signaling, establishing a juxtacrine mechanism controlling kidney development.
Collapse
Affiliation(s)
- Hongtao Zhang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Mazdak Bagherie-Lachidan
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Caroline Badouel
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 Route de Narbonne, Toulouse 31062, France
| | - Leonie Enderle
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Philippos Peidis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Rod Bremner
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Departments of Ophthalmology and Visual Science, and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Satu Kuure
- GM-unit at Laboratory Animal Centre, HiLIFE and Medicum, University of Helsinki, Helsinki 00014, Finland
| | - Sanjay Jain
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Helen McNeill
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
27
|
Impact of next generation sequencing on our understanding of CAKUT. Semin Cell Dev Biol 2018; 91:104-110. [PMID: 30172048 DOI: 10.1016/j.semcdb.2018.08.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 08/16/2018] [Accepted: 08/28/2018] [Indexed: 12/29/2022]
Abstract
Congenital abnormalities of the kidney and urinary tract (CAKUT) form the leading cause of pediatric end-stage renal disease. Knowledge on the molecular mechanisms that underlie CAKUT leads to the improvement of DNA diagnostics and counseling regarding prognosis and recurrence risk estimation for CAKUT patients and their relatives. Implementation of next generation sequencing in research and diagnostic settings has led to the identification of the molecular basis of many developmental diseases. In this review, we summarize the efforts on next generation sequencing in CAKUT research and we discuss how next generation sequencing added to our understanding of CAKUT genetics. Although next generation sequencing has certainly proven to be a game changer in the field of disease gene identification and novel CAKUT-causing gene variants have been identified, most CAKUT cases still remain unsolved. Occurring with genetic and phenotypic heterogeneity along with incomplete penetrance, the identification of CAKUT etiology poses many challenges. We see great potential for combined -omics approaches that include next generation sequencing in the identification of CAKUT-specific biomarkers, which is necessary to optimize the care for CAKUT patients.
Collapse
|
28
|
Davidson AJ, Lewis P, Przepiorski A, Sander V. Turning mesoderm into kidney. Semin Cell Dev Biol 2018; 91:86-93. [PMID: 30172050 DOI: 10.1016/j.semcdb.2018.08.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 08/24/2018] [Accepted: 08/28/2018] [Indexed: 02/07/2023]
Abstract
The intermediate mesoderm is located between the somites and the lateral plate mesoderm and gives rise to renal progenitors that contribute to the three mammalian kidney types (pronephros, mesonephros and metanephros). In this review, focusing largely on murine kidney development, we examine how the intermediate mesoderm forms during gastrulation/axis elongation and how it progressively gives rise to distinct renal progenitors along the rostro-caudal axis. We highlight some of the potential signalling cues and core transcription factor circuits that direct these processes, up to the point of early metanephric kidney formation.
Collapse
Affiliation(s)
- Alan J Davidson
- Department of Molecular Medicine & Pathology, School of Medical Sciences, Faculty of Medical & Health Sciences, The University of Auckland, Private Bag 921019, Auckland 1142, New Zealand.
| | - Paula Lewis
- Department of Molecular Medicine & Pathology, School of Medical Sciences, Faculty of Medical & Health Sciences, The University of Auckland, Private Bag 921019, Auckland 1142, New Zealand
| | - Aneta Przepiorski
- Department of Molecular Medicine & Pathology, School of Medical Sciences, Faculty of Medical & Health Sciences, The University of Auckland, Private Bag 921019, Auckland 1142, New Zealand
| | - Veronika Sander
- Department of Molecular Medicine & Pathology, School of Medical Sciences, Faculty of Medical & Health Sciences, The University of Auckland, Private Bag 921019, Auckland 1142, New Zealand
| |
Collapse
|
29
|
Pons M, Koniaris LG, Moe SM, Gutierrez JC, Esquela-Kerscher A, Zimmers TA. GDF11 induces kidney fibrosis, renal cell epithelial-to-mesenchymal transition, and kidney dysfunction and failure. Surgery 2018; 164:262-273. [PMID: 29731246 DOI: 10.1016/j.surg.2018.03.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 03/04/2018] [Accepted: 03/05/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND GDF11 modulates embryonic patterning and kidney organogenesis. Herein, we sought to define GDF11 function in the adult kidney and in renal diseases. METHODS In vitro renal cell lines, genetic, and murine in vivo renal injury models were examined. RESULTS Among tissues tested, Gdf11 was highest in normal adult mouse kidney. Expression was increased acutely after 5/6 nephrectomy, ischemia-reperfusion injury, kanamycin toxicity, or unilateral ureteric obstruction. Systemic, high-dose GDF11 administration in adult mice led to renal failure, with accompanying kidney atrophy, interstitial fibrosis, epithelial-to-mesenchymal transition of renal tubular cells, and eventually death. These effects were associated with phosphorylation of SMAD2 and could be blocked by follistatin. In contrast, Gdf11 heterozygous mice showed reduced renal Gdf11 expression, renal fibrosis, and expression of fibrosis-associated genes both at baseline and after unilateral ureteric obstruction compared with wild-type littermates. The kidney-specific consequences of GDF11 dose modulation are direct effects on kidney cells. GDF11 induced proliferation and activation of NRK49f renal fibroblasts and also promoted epithelial-to-mesenchymal transition of IMCD-3 tubular epithelial cells in a SMAD3-dependent manner. CONCLUSION Taken together, these data suggest that GDF11 and its downstream signals are critical in vivo mediators of renal injury. These effects are through direct actions of GDF11 on renal tubular cells and fibroblasts. Thus, regulation of GDF11 presents a therapeutic target for diseases involving renal fibrosis and impaired tubular function.
Collapse
Affiliation(s)
- Marianne Pons
- Department of Surgery, Indiana University School of Medicine, Indianapolis
| | | | - Sharon M Moe
- Department of Medicine, Indiana University School of Medicine, Indianapolis; Roudebush Veterans Administration Medical Center, Indianapolis, IN
| | | | - Aurora Esquela-Kerscher
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk
| | - Teresa A Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis; Departments of Anatomy and Cell Biology, Biochemistry and Molecular Biology and Otolaryngology-Head & Neck Surgery, Indiana University School of Medicine, Indianapolis; IU Simon Cancer Center, Indianapolis, IN
| |
Collapse
|
30
|
Jin Q, Qiao C, Li J, Li J, Xiao X. Neonatal Systemic AAV-Mediated Gene Delivery of GDF11 Inhibits Skeletal Muscle Growth. Mol Ther 2018; 26:1109-1117. [PMID: 29503194 DOI: 10.1016/j.ymthe.2018.01.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 01/10/2018] [Accepted: 01/16/2018] [Indexed: 12/19/2022] Open
Abstract
Growth and differentiation factor 11 (GDF11; BMP11) is a circulating cytokine in the transforming growth factor beta (TGF-β) superfamily. Treatment with recombinant GDF11 (rGDF11) protein has previously been shown to reverse skeletal muscle dysfunction in aged mice. However, the actions of GDF11 in skeletal muscle are still not fully understood. Because GDF11 activates the TGF-β-SMAD2/3 pathway, we hypothesized that GDF11 overexpression would inhibit skeletal muscle growth. To test this hypothesis, we generated recombinant adeno-associated virus serotype 9 (AAV9) vectors harboring the gene for either human GDF11 (AAV9-GDF11) or human IgG1 Fc-fused GDF11 propeptide (AAV9-GDF11Pro-Fc-1) to study the effects of GDF11 overexpression or blockade on skeletal muscle growth and function in vivo. After intravenous administration of AAV9-GDF11 into neonatal C57BL/6J mice, we observed sustained limb muscle growth inhibition along with reductions in forelimb grip strength and treadmill running endurance at 16 weeks. Conversely, treatment with AAV9-GDF11Pro-Fc-1 led to increased limb muscle mass and forelimb grip strength after 28 weeks, although a difference in the total body mass/muscle mass ratio was not observed between treatment and control groups. In sum, our results suggest GDF11 overexpression has an inhibitory effect on skeletal muscle growth.
Collapse
Affiliation(s)
- Quan Jin
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chunping Qiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jianbin Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Juan Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xiao Xiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
31
|
Role of growth differentiation factor 11 in development, physiology and disease. Oncotarget 2017; 8:81604-81616. [PMID: 29113418 PMCID: PMC5655313 DOI: 10.18632/oncotarget.20258] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 07/28/2017] [Indexed: 12/31/2022] Open
Abstract
Growth differentiation factor (GDF11) is a member of TGF-β/BMP superfamily that activates Smad and non-Smad signaling pathways and regulates expression of its target nuclear genes. Since its discovery in 1999, studies have shown the involvement of GDF11 in normal physiological processes, such as embryonic development and erythropoiesis, as well as in the pathophysiology of aging, cardiovascular disease, diabetes mellitus, and cancer. In addition, there are contradictory reports regarding the role of GDF11 in aging, cardiovascular disease, diabetes mellitus, osteogenesis, skeletal muscle development, and neurogenesis. In this review, we describe the GDF11 signaling pathway and its potential role in development, physiology and disease.
Collapse
|
32
|
Plotnikov EY, Pavlenko TA, Pevzner IB, Zorova LD, Manskikh VN, Silachev DN, Sukhikh GT, Zorov DB. The role of oxidative stress in acute renal injury of newborn rats exposed to hypoxia and endotoxin. FEBS J 2017; 284:3069-3078. [DOI: 10.1111/febs.14177] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/05/2017] [Accepted: 07/13/2017] [Indexed: 01/24/2023]
Affiliation(s)
- Egor Y. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology; M.V. Lomonosov Moscow State University; Russia
- V. I. Kulakov Research Center of Obstetrics, Gynecology and Perinatology; Ministry of Health of the Russian Federation; Moscow Russia
| | - Tatiana A. Pavlenko
- A.N. Belozersky Institute of Physico-Chemical Biology; M.V. Lomonosov Moscow State University; Russia
- Russian Cardiology Research and Production Center; Moscow Russia
| | - Irina B. Pevzner
- A.N. Belozersky Institute of Physico-Chemical Biology; M.V. Lomonosov Moscow State University; Russia
- V. I. Kulakov Research Center of Obstetrics, Gynecology and Perinatology; Ministry of Health of the Russian Federation; Moscow Russia
| | - Ljubava D. Zorova
- A.N. Belozersky Institute of Physico-Chemical Biology; M.V. Lomonosov Moscow State University; Russia
- V. I. Kulakov Research Center of Obstetrics, Gynecology and Perinatology; Ministry of Health of the Russian Federation; Moscow Russia
- International Laser Center; M.V. Lomonosov Moscow State University; Russia
| | - Vasily N. Manskikh
- A.N. Belozersky Institute of Physico-Chemical Biology; M.V. Lomonosov Moscow State University; Russia
| | - Denis N. Silachev
- A.N. Belozersky Institute of Physico-Chemical Biology; M.V. Lomonosov Moscow State University; Russia
- V. I. Kulakov Research Center of Obstetrics, Gynecology and Perinatology; Ministry of Health of the Russian Federation; Moscow Russia
| | - Gennady T. Sukhikh
- V. I. Kulakov Research Center of Obstetrics, Gynecology and Perinatology; Ministry of Health of the Russian Federation; Moscow Russia
| | - Dmitry B. Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology; M.V. Lomonosov Moscow State University; Russia
- V. I. Kulakov Research Center of Obstetrics, Gynecology and Perinatology; Ministry of Health of the Russian Federation; Moscow Russia
| |
Collapse
|
33
|
Abstract
In addition to its roles in embryonic development, Growth and Differentiation Factor 11 (GDF 11) has recently drawn much interest about its roles in other processes, such as aging. GDF 11 has been shown to play pivotal roles in the rescue of the proliferative and regenerative capabilities of skeletal muscle, neural stem cells and cardiomyocytes. We would be remiss not to point that some controversy exists regarding the role of GDF 11 in biological processes and whether it will serve as a therapeutic agent. The latest studies have shown that the level of circulating GDF 11 correlates with the outcomes of patients with cardiovascular diseases, cancer and uremia. Based on these studies, GDF 11 is a promising candidate to serve as a novel biomarker of diseases. This brief review gives a detailed and concise view of the regulation and functions of GDF 11 and its roles in development, neurogenesis and erythropoiesis as well as the prospect of using this protein as an indicator of cardiac health and aging.
Collapse
Affiliation(s)
- A Jamaiyar
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA; School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - W Wan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA; Department of Cardiology, Renmin Hospital of Wuhan University, Hubei, China
| | - D M Janota
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - M K Enrick
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - W M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - L Yin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA.
| |
Collapse
|
34
|
Qin X, Kuang H, Chen L, Wei S, Yu D, Liang F. Coexpression of growth differentiation factor 11 and reactive oxygen species in metastatic oral cancer and its role in inducing the epithelial to mesenchymal transition. Oral Surg Oral Med Oral Pathol Oral Radiol 2017; 123:697-706. [PMID: 28478937 DOI: 10.1016/j.oooo.2017.03.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 02/05/2017] [Accepted: 03/07/2017] [Indexed: 12/20/2022]
Abstract
OBJECTIVES The aim of this study was to investigate growth differentiation factor 11 (GDF11) and reactive oxygen species (ROS) expression in metastatic oral cancer and explored their roles in inducing epithelial to mesenchymal transition (EMT). STUDY DESIGN The expression of GDF11, ROS, and EMT-related markers was evaluated in primary tumor tissues from patients with oral squamous cell carcinoma (OSCC). SCC-9 cells, a human tongue carcinoma cell line, were treated with recombinant GDF11. Induction of EMT, expression of EMT-related markers, and the effect of ROS on EMT in SCC-9 cells were analyzed. RESULTS Overexpression of GDF11 and ROS was observed in patients with metastatic oral cancer. Downregulated expression of E-cadherin and upregulated expression of vimentin, δ-EF1, SIP-1, MMP-2, and MMP-9 were observed in patients with metastatic oral cancer, relative to the expression of these factors in patients with nonmetastatic oral cancer. With recombinant GDF11 treatment, obvious spindle-shaped cells appeared, and gene expressions of EMT-related markers were altered in SCC-9 cells. Treatment with the powerful antioxidant N-acetylcysteine inhibited GDF11-induced EMT and cell migration. CONCLUSIONS GDF11 induces EMT and cell migration with ROS involvement in SCC-9 cells. Overexpression of GDF11 and ROS is associated with metastatic oral cancer. GDF11 and ROS may participate in metastasis of oral cancer through EMT.
Collapse
Affiliation(s)
- Xiangming Qin
- Graduate School, Guangxi Medical University, Nanning, Guangxi, China
| | - Hai Kuang
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Lei Chen
- Graduate School, Guangxi Medical University, Nanning, Guangxi, China
| | - Shanliang Wei
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Dahai Yu
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Feixin Liang
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
35
|
Short KM, Smyth IM. The contribution of branching morphogenesis to kidney development and disease. Nat Rev Nephrol 2016; 12:754-767. [DOI: 10.1038/nrneph.2016.157] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
36
|
Zhang Y, Li Q, Liu D, Huang Q, Cai G, Cui S, Sun X, Chen X. GDF11 improves tubular regeneration after acute kidney injury in elderly mice. Sci Rep 2016; 6:34624. [PMID: 27703192 PMCID: PMC5050408 DOI: 10.1038/srep34624] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/09/2016] [Indexed: 01/27/2023] Open
Abstract
The GDF11 expression pattern and its effect on organ regeneration after acute injury in the elderly population are highly controversial topics. In our study, GDF11/8 expression increased after kidney ischemia–reperfusion injury (IRI), and the relatively lower level of GDF11/8 in the kidneys of aged mice was associated with a loss of proliferative capacity and a decline in renal repair, compared to young mice. In vivo, GDF11 supplementation in aged mice increased vimentin and Pax2 expression in the kidneys as well as the percentage of 5-ethynyl-2′-deoxyuridine (EdU)-positive proximal tubular epithelial cells. GDF11 improved the renal repair, recovery of renal function, and survival of elderly mice at 72 h after IRI. Moreover, the addition of recombinant GDF11 to primary renal epithelial cells increased proliferation, migration, and dedifferentiation by upregulating the ERK1/2 pathway in vitro. Our study indicates that GDF11/8 in the kidney decreases with age and that GDF11 can increase tubular cell dedifferentiation and proliferation as well as improve tubular regeneration after acute kidney injury (AKI) in old mice.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, 100853, China.,Medical College, Nankai University, Tianjin, 300071, China
| | - Qinggang Li
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, 100853, China
| | - Dong Liu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, 100853, China.,Department of Nephrology, Chinese PLA Air Force General Hospital, Beijing, 100142, China
| | - Qi Huang
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, 100853, China
| | - Guangyan Cai
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, 100853, China
| | - Shaoyuan Cui
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, 100853, China
| | - Xuefeng Sun
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, 100853, China
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing, 100853, China
| |
Collapse
|
37
|
Liu W, Zhou L, Zhou C, Zhang S, Jing J, Xie L, Sun N, Duan X, Jing W, Liang X, Zhao H, Ye L, Chen Q, Yuan Q. GDF11 decreases bone mass by stimulating osteoclastogenesis and inhibiting osteoblast differentiation. Nat Commun 2016; 7:12794. [PMID: 27653144 PMCID: PMC5036163 DOI: 10.1038/ncomms12794] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 08/02/2016] [Indexed: 02/05/2023] Open
Abstract
Osteoporosis is an age-related disease that affects millions of people. Growth differentiation factor 11 (GDF11) is a secreted member of the transforming growth factor beta (TGF-β) superfamily. Deletion of Gdf11 has been shown to result in a skeletal anterior-posterior patterning disorder. Here we show a role for GDF11 in bone remodelling. GDF11 treatment leads to bone loss in both young and aged mice. GDF11 inhibits osteoblast differentiation and also stimulates RANKL-induced osteoclastogenesis through Smad2/3 and c-Fos-dependent induction of Nfatc1. Injection of GDF11 impairs bone regeneration in mice and blocking GDF11 function prevents oestrogen-deficiency-induced bone loss and ameliorates age-related osteoporosis. Our data demonstrate that GDF11 is a previously unrecognized regulator of bone remodelling and suggest that GDF11 is a potential target for treatment of osteoporosis.
Collapse
Affiliation(s)
- Weiqing Liu
- State Key Laboratory of Oral diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Liyan Zhou
- State Key Laboratory of Oral diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chenchen Zhou
- State Key Laboratory of Oral diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shiwen Zhang
- State Key Laboratory of Oral diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Junjun Jing
- State Key Laboratory of Oral diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Liang Xie
- State Key Laboratory of Oral diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ningyuan Sun
- State Key Laboratory of Oral diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xiaobo Duan
- State Key Laboratory of Oral diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Wei Jing
- State Key Laboratory of Oral diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xing Liang
- State Key Laboratory of Oral diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hu Zhao
- State Key Laboratory of Oral diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ling Ye
- State Key Laboratory of Oral diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Qianming Chen
- State Key Laboratory of Oral diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Quan Yuan
- State Key Laboratory of Oral diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
38
|
Walker RG, Poggioli T, Katsimpardi L, Buchanan SM, Oh J, Wattrus S, Heidecker B, Fong YW, Rubin LL, Ganz P, Thompson TB, Wagers AJ, Lee RT. Biochemistry and Biology of GDF11 and Myostatin: Similarities, Differences, and Questions for Future Investigation. Circ Res 2016; 118:1125-41; discussion 1142. [PMID: 27034275 DOI: 10.1161/circresaha.116.308391] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 03/07/2016] [Indexed: 02/06/2023]
Abstract
Growth differentiation factor 11 (GDF11) and myostatin (or GDF8) are closely related members of the transforming growth factor β superfamily and are often perceived to serve similar or overlapping roles. Yet, despite commonalities in protein sequence, receptor utilization and signaling, accumulating evidence suggests that these 2 ligands can have distinct functions in many situations. GDF11 is essential for mammalian development and has been suggested to regulate aging of multiple tissues, whereas myostatin is a well-described negative regulator of postnatal skeletal and cardiac muscle mass and modulates metabolic processes. In this review, we discuss the biochemical regulation of GDF11 and myostatin and their functions in the heart, skeletal muscle, and brain. We also highlight recent clinical findings with respect to a potential role for GDF11 and/or myostatin in humans with heart disease. Finally, we address key outstanding questions related to GDF11 and myostatin dynamics and signaling during development, growth, and aging.
Collapse
Affiliation(s)
- Ryan G Walker
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Tommaso Poggioli
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Lida Katsimpardi
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Sean M Buchanan
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Juhyun Oh
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Sam Wattrus
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Bettina Heidecker
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Yick W Fong
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Lee L Rubin
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Peter Ganz
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Thomas B Thompson
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Amy J Wagers
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.).
| | - Richard T Lee
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.).
| |
Collapse
|
39
|
Yamagishi SI, Matsui T, Kurokawa Y, Fukami K. Serum Levels of Growth Differentiation Factor 11 Are Independently Associated with Low Hemoglobin Values in Hemodialysis Patients. Biores Open Access 2016; 5:155-8. [PMID: 27298756 PMCID: PMC4900214 DOI: 10.1089/biores.2016.0015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Circulating levels of growth differentiation factor 11 (GDF11) have been shown to decrease with age in several mammalian species, and supplementation of GDF11 by heterochronic parabiosis or systemic administration reverses age-related organ damage. However, there is some controversy about the pathophysiological role of GDF11 in aging-associated organ damage. Since aging process is accelerated in uremia, we compared serum levels of GDF11 in hemodialysis (HD) patients with those in age-matched healthy controls, and then determined the independent clinical correlates of GDF11 in HD subjects. Sixty-two maintenance HD patients (34 male and 28 female; mean age, 52.6 years; mean duration of HD, 7.7 months) were enrolled in the present study. Twenty-nine age-matched subjects were used as a control. GDF11 was measured by a commercially available enzyme-linked immunosorbent assay kit. Serum GDF11 levels in HD patients were significantly higher than those in controls (9.4 ± 5.1 pg/mL vs. 7.3 ± 5.9 pg/mL). A statistical significance was demonstrated between GDF11 and hemoglobin (inversely). Multiple stepwise regression analysis revealed that hemoglobin (p < 0.001) was a sole independent correlate of GDF11 levels in HD patients (R2 = 0.168). Our present study suggests that kinetics and regulation of circulating GDF11 may differ between normal physiological aging process and accelerated pathological aging conditions, such as uremia. Given that GDF11 has been shown to inhibit erythroid maturation in mice, elevation of GDF11 levels may be involved in erythropoietin-resistant anemia in HD patients.
Collapse
Affiliation(s)
- Sho-Ichi Yamagishi
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine , Kurume, Japan
| | - Takanori Matsui
- Department of Pathophysiology and Therapeutics of Diabetic Vascular Complications, Kurume University School of Medicine , Kurume, Japan
| | - Yuka Kurokawa
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine , Kurume, Japan
| | - Kei Fukami
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine , Kurume, Japan
| |
Collapse
|
40
|
Jurberg AD, Vasconcelos-Fontes L, Cotta-de-Almeida V. A Tale from TGF-β Superfamily for Thymus Ontogeny and Function. Front Immunol 2015; 6:442. [PMID: 26441956 PMCID: PMC4564722 DOI: 10.3389/fimmu.2015.00442] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 08/14/2015] [Indexed: 12/16/2022] Open
Abstract
Multiple signaling pathways control every aspect of cell behavior, organ formation, and tissue homeostasis throughout the lifespan of any individual. This review takes an ontogenetic view focused on the large superfamily of TGF-β/bone morphogenetic protein ligands to address thymus morphogenesis and function in T cell differentiation. Recent findings on a role of GDF11 for reversing aging-related phenotypes are also discussed.
Collapse
Affiliation(s)
- Arnon Dias Jurberg
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro , Brazil ; Graduate Program in Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro , Rio de Janeiro , Brazil
| | - Larissa Vasconcelos-Fontes
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro , Brazil
| | - Vinícius Cotta-de-Almeida
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro , Brazil
| |
Collapse
|
41
|
Nagalakshmi VK, Yu J. The ureteric bud epithelium: morphogenesis and roles in metanephric kidney patterning. Mol Reprod Dev 2015; 82:151-66. [PMID: 25783232 DOI: 10.1002/mrd.22462] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 01/12/2015] [Indexed: 01/03/2023]
Abstract
The mammalian metanephric kidney is composed of two epithelial components, the collecting duct system and the nephron epithelium, that differentiate from two different tissues -the ureteric bud epithelium and the nephron progenitors, respectively-of intermediate mesoderm origin. The collecting duct system is generated through reiterative ureteric bud branching morphogenesis, whereas the nephron epithelium is formed in a process termed nephrogenesis, which is initiated with the mesenchymal-epithelial transition of the nephron progenitors. Ureteric bud branching morphogenesis is regulated by nephron progenitors, and in return, the ureteric bud epithelium regulates nephrogenesis. The metanephric kidney is physiologically divided along the corticomedullary axis into subcompartments that are enriched with specific segments of these two epithelial structures. Here, we provide an overview of the major molecular and cellular processes underlying the morphogenesis and patterning of the ureteric bud epithelium and its roles in the cortico-medullary patterning of the metanephric kidney.
Collapse
Affiliation(s)
- Vidya K Nagalakshmi
- Department of Cell Biology and Division of Center of Immunity, Inflammation and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | | |
Collapse
|
42
|
Das UN. Molecular, Biochemical, and Physiological Basis of Beneficial Actions of Exercise. DIET AND EXERCISE IN COGNITIVE FUNCTION AND NEUROLOGICAL DISEASES 2015:183-204. [DOI: 10.1002/9781118840634.ch17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
43
|
Vanbekbergen N, Hendrickx M, Leyns L. Growth differentiation factor 11 is an encephalic regionalizing factor in neural differentiated mouse embryonic stem cells. BMC Res Notes 2014; 7:766. [PMID: 25352416 PMCID: PMC4228095 DOI: 10.1186/1756-0500-7-766] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 10/14/2014] [Indexed: 12/02/2022] Open
Abstract
Background The central nervous system has a complex structural organization and consists of different subdomains along the antero-posterior axis. However, questions remain about the molecular mechanisms leading to the regionalization of this organ. We used a previously developed methodology to identify the novel patterning role of GDF11, a TGF-β signaling factor. Findings Using an assay based on neural differentiated mouse embryonic stem cells, GDF11 is shown to induce diencephalic (posterior forebrain), mesencephalic (midbrain) and metencephalic (anterior hindbrain) fates at the expense of telencephalic (anterior forebrain) specification. GDF11 has not previously been implicated in the early patterning of the nervous system. In addition, inhibition of the TGF-β type I receptors Alk4, Alk5 and Alk7 by the pharmacological inhibitor SB431542 caused a strong anteriorization of the cells. Conclusions Our findings suggest that GDF11 is involved in the earliest steps of the brain patterning during neurogenesis in the vertebrate embryo and is shown to be a regionalizing factor of the regional fate in the developing brain. This regionalization is not a typical posteriorizing signal as seen with retinoic acid, FGF or BMP molecules. To our knowledge, this is the first time that GDF11 is implicated in the earliest steps of the patterning of the neural plate.
Collapse
Affiliation(s)
| | | | - Luc Leyns
- Department of Biology, Lab for Cell Genetics, Vrije Universiteit Brussel (VUB), 2 Pleinlaan, B-1050 Brussels, Belgium.
| |
Collapse
|
44
|
Wang RN, Green J, Wang Z, Deng Y, Qiao M, Peabody M, Zhang Q, Ye J, Yan Z, Denduluri S, Idowu O, Li M, Shen C, Hu A, Haydon RC, Kang R, Mok J, Lee MJ, Luu HL, Shi LL. Bone Morphogenetic Protein (BMP) signaling in development and human diseases. Genes Dis 2014; 1:87-105. [PMID: 25401122 PMCID: PMC4232216 DOI: 10.1016/j.gendis.2014.07.005] [Citation(s) in RCA: 733] [Impact Index Per Article: 66.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 07/15/2014] [Indexed: 02/06/2023] Open
Abstract
Bone Morphogenetic Proteins (BMPs) are a group of signaling molecules that belongs to the Transforming Growth Factor-β (TGF-β) superfamily of proteins. Initially discovered for their ability to induce bone formation, BMPs are now known to play crucial roles in all organ systems. BMPs are important in embryogenesis and development, and also in maintenance of adult tissue homeostasis. Mouse knockout models of various components of the BMP signaling pathway result in embryonic lethality or marked defects, highlighting the essential functions of BMPs. In this review, we first outline the basic aspects of BMP signaling and then focus on genetically manipulated mouse knockout models that have helped elucidate the role of BMPs in development. A significant portion of this review is devoted to the prominent human pathologies associated with dysregulated BMP signaling.
Collapse
Affiliation(s)
- Richard N. Wang
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jordan Green
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zhongliang Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Youlin Deng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Min Qiao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Michael Peabody
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Qian Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Jixing Ye
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- School of Bioengineering, Chongqing University, Chongqing, China
| | - Zhengjian Yan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Sahitya Denduluri
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Olumuyiwa Idowu
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Melissa Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Christine Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Alan Hu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Richard Kang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - James Mok
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue L. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Lewis L. Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
45
|
Blake J, Rosenblum ND. Renal branching morphogenesis: morphogenetic and signaling mechanisms. Semin Cell Dev Biol 2014; 36:2-12. [PMID: 25080023 DOI: 10.1016/j.semcdb.2014.07.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 07/14/2014] [Accepted: 07/17/2014] [Indexed: 12/28/2022]
Abstract
The human kidney is composed of an arborized network of collecting ducts, calyces and urinary pelvis that facilitate urine excretion and regulate urine composition. The renal collecting system is formed in utero, completed by the 34th week of gestation in humans, and dictates final nephron complement. The renal collecting system arises from the ureteric bud, a derivative of the intermediate-mesoderm derived nephric duct that responds to inductive signals from adjacent tissues via a process termed ureteric induction. The ureteric bud subsequently undergoes a series of iterative branching and remodeling events in a process called renal branching morphogenesis. Altered signaling that disrupts patterning of the nephric duct, ureteric induction, or renal branching morphogenesis leads to varied malformations of the renal collecting system collectively known as congenital anomalies of the kidney and urinary tract (CAKUT) and is the most frequently detected congenital renal aberration in infants. Here, we describe critical morphogenetic and cellular events that govern nephric duct specification, ureteric bud induction, renal branching morphogenesis, and cessation of renal branching morphogenesis. We also highlight salient molecular signaling pathways that govern these processes, and the investigative techniques used to interrogate them.
Collapse
Affiliation(s)
- Joshua Blake
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Canada; Department of Physiology, University of Toronto, Canada
| | - Norman D Rosenblum
- Division of Nephrology, Department of Paediatrics, The Hospital for Sick Children, Canada; Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Canada; Department of Physiology, University of Toronto, Canada.
| |
Collapse
|
46
|
Gene regulatory network of renal primordium development. Pediatr Nephrol 2014; 29:637-44. [PMID: 24104595 DOI: 10.1007/s00467-013-2635-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/06/2013] [Accepted: 09/06/2013] [Indexed: 12/23/2022]
Abstract
Animal development progresses through the stepwise deployment of gene regulatory networks (GRN) encoded in the genome. Comparative analyses in different species and organ systems have revealed that GRN blueprints are composed of subcircuits with stereotypical architectures that are often reused as modular units. In this review, we report the evidence for the GRN underlying renal primordium development. In vertebrates, renal development is initiated by the induction of a field of intermediate mesoderm cells competent to undergo lineage specification and nephric (Wolffian) duct formation. Definition of the renal field leads to the activation of a core regulatory subcircuit composed of the transcription factors Pax2/8, Gata3 and Lim1. These transcription factors turn on a second layer of transcriptional regulators while also activating effectors of tissue morphogenesis and cellular specialization. Elongation and connection of the nephric duct to the cloaca (bladder/urethra primordium) is followed by metanephric kidney induction through signals emanating from the metanephric mesenchyme. Central to this process is the activation and positioning of the glial cell line-derived neurotrophic factor (Gdnf)-Ret signaling pathway by network subcircuits located in the mesenchyme and epithelial tissues of the caudal trunk. Evidence shows that each step of the renal primordium developmental program is regulated by structured GRN subunits organized in a hierarchical manner. Understanding the structure and dynamics of the renal GRN will help us understand the intrinsic phenotypical variability of congenital anomalies of the kidney and urinary tract and guide our approaches to regenerative medicine.
Collapse
|
47
|
Costantini F. Genetic controls and cellular behaviors in branching morphogenesis of the renal collecting system. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2014; 1:693-713. [PMID: 22942910 DOI: 10.1002/wdev.52] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The mammalian kidney, which at maturity contains thousands of nephrons joined to a highly branched collecting duct (CD) system, is an important model system for studying the development of a complex organ. Furthermore, congenital anomalies of the kidney and urinary tract, often resulting from defects in ureteric bud branching morphogenesis, are relatively common human birth defects. Kidney development is initiated by interactions between the nephric duct and the metanephric mesenchyme, leading to the outgrowth and repeated branching of the ureteric bud epithelium, which gives rise to the entire renal CD system. Meanwhile, signals from the ureteric bud induce the mesenchyme cells to form the nephron epithelia. This review focuses on development of the CD system, with emphasis on the mouse as an experimental system. The major topics covered include the origin and development of the nephric duct, formation of the ureteric bud, branching morphogenesis of the ureteric bud, and elongation of the CDs. The signals, receptors, transcription factors, and other regulatory molecules implicated in these processes are discussed. In addition, our current knowledge of cellular behaviors that are controlled by these genes and underlie development of the collecting system is reviewed.
Collapse
Affiliation(s)
- Frank Costantini
- Department of Genetics and Development, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
48
|
Jeanplong F, Falconer SJ, Oldham JM, Maqbool NJ, Thomas M, Hennebry A, McMahon CD. Identification and expression of a novel transcript of the growth and differentiation factor-11 gene. Mol Cell Biochem 2013; 390:9-18. [PMID: 24378996 DOI: 10.1007/s11010-013-1949-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 12/19/2013] [Indexed: 10/25/2022]
Abstract
The growth and differentiation factor-11 (GDF-11) gene is thought to code for a single protein that plays a crucial role in regulating the development of multiple tissues. In this study, we aimed to investigate if the GDF-11 gene has another transcript and, if so, to characterise this transcript and determine its tissue-specific and developmental expression. We have identified a novel transcript of GDF-11 in mouse muscle, which contains the 3' region of intron 1, exon 2, exon 3 and 3'UTR, and has two transcription initiation sites and a single termination site. We named the novel transcript GDF-11ΔEx1 because it does not contain exon 1 of canonical GDF-11. The GDF-11ΔEx1 transcript was expressed in the skeletal muscles, heart, brain and kidney, but was undetectable in the liver and gut. The concentration of the GDF-11ΔEx1 transcript was increased in gastrocnemius muscles from three to 6 weeks of age, a period of accelerated muscle growth, steadily declined thereafter and was higher in male than female mice (P < 0.001 for age and sex). GDF-11ΔEx1 cDNA was predicted to code for a putative N-terminal-truncated propeptide and the canonical ligand for GDF-11. However, propeptide-specific antibodies could not identify proteins of the expected size in skeletal muscle. Interestingly, in silico analysis of the GDF-11ΔEx1 RNA predicted a secondary structure with the potential to coordinate multiple protein interactions as a molecular scaffold. Therefore, we postulate that GDF-11ΔEx1 may act as a long non-coding RNA to regulate the transcription of canonical GDF-11 and/or other genes in skeletal muscle and other tissues.
Collapse
Affiliation(s)
- Ferenc Jeanplong
- AgResearch, Ruakura Research Centre, Private Bag 3123, Hamilton, New Zealand,
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
In an intriguing new study, Loffredo et al report that joining the circulation of old mice with that of young mice reduces age-related cardiac hypertrophy. They also found that the growth factor growth/differentiation factor 11 is a circulating negative regulator of cardiac hypertrophy which suggests that raising growth/differentiation factor 11 levels may be useful to treat cardiac hypertrophy associated with aging.
Collapse
Affiliation(s)
- Alexandra C McPherron
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
50
|
Tee JB, Choi Y, Dnyanmote A, Decambre M, Ito C, Bush KT, Nigam SK. GDNF-independent ureteric budding: role of PI3K-independent activation of AKT and FOSB/JUN/AP-1 signaling. Biol Open 2013; 2:952-9. [PMID: 24143282 PMCID: PMC3773342 DOI: 10.1242/bio.20135595] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 06/20/2013] [Indexed: 01/19/2023] Open
Abstract
A significant fraction of mice deficient in either glial cell-derived neurotrophic factor (GDNF) or its co-receptors (Gfrα1, Ret), undergoes ureteric bud (UB) outgrowth leading to the formation of a rudimentary kidney. Previous studies using the isolated Wolffian duct (WD) culture indicate that activation of fibroblast growth factor (FGF) receptor signaling, together with suppression of BMP/Activin signaling, is critical for GDNF-independent WD budding (Maeshima et al., 2007). By expression analysis of embryonic kidney from Ret((-/-)) mice, we found the upregulation of several FGFs, including FGF7. To examine the intracellular pathways, we then analyzed GDNF-dependent and GDNF-independent budding in the isolated WD culture. In both conditions, Akt activation was found to be important; however, whereas this occurred through PI3-kinase in GDNF-dependent budding, in the case of GDNF-independent budding, Akt activation was apparently via a PI3-kinase independent mechanism. Jnk signaling and the AP-1 transcription factor complex were also implicated in GDNF-independent budding. FosB, a binding partner of c-Jun in the formation of AP-1, was the most highly upregulated gene in the ret knockout kidney (in which budding had still occurred), and we found that its siRNA-mediated knockdown in isolated WDs also blocked GDNF-independent budding. Taken together with the finding that inhibition of Jnk signaling does not block Akt activation/phosphorylation in GDNF-independent budding, the data support necessary roles for both FosB/Jun/AP-1 signaling and PI3-kinase-independent activation of Akt in GDNF-independent budding. A model is proposed for signaling events that involve Akt and JNK working to regulate GDNF-independent WD budding.
Collapse
Affiliation(s)
- James B Tee
- Department of Medicine, University of California , San Diego, La Jolla, CA 92093-0693 , USA ; Present address: Department of Pediatrics, University of Calgary and Alberta Children's Hospital, Calgary, AB T3B 6A8, Canada
| | | | | | | | | | | | | |
Collapse
|