1
|
Schneider RF, Gunter HM, Salewski I, Woltering JM, Meyer A. Growth dynamics and molecular bases of evolutionary novel jaw extensions in halfbeaks and needlefishes (Beloniformes). Mol Ecol 2023; 32:5798-5811. [PMID: 37750351 DOI: 10.1111/mec.17143] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/17/2023] [Accepted: 09/05/2023] [Indexed: 09/27/2023]
Abstract
Evolutionary novelties-derived traits without clear homology found in the ancestors of a lineage-may promote ecological specialization and facilitate adaptive radiations. Examples for such novelties include the wings of bats, pharyngeal jaws of cichlids and flowers of angiosperms. Belonoid fishes (flying fishes, halfbeaks and needlefishes) feature an astonishing diversity of extremely elongated jaw phenotypes with undetermined evolutionary origins. We investigate the development of elongated jaws in a halfbeak (Dermogenys pusilla) and a needlefish (Xenentodon cancila) using morphometrics, transcriptomics and in situ hybridization. We confirm that these fishes' elongated jaws are composed of distinct base and novel 'extension' portions. These extensions are morphologically unique to belonoids, and we describe the growth dynamics of both bases and extensions throughout early development in both studied species. From transcriptomic profiling, we deduce that jaw extension outgrowth is guided by populations of multipotent cells originating from the anterior tip of the dentary. These cells are shielded from differentiation, but proliferate and migrate anteriorly during the extension's allometric growth phase. Cells left behind at the tip leave the shielded zone and undergo differentiation into osteoblast-like cells, which deposit extracellular matrix with both bone and cartilage characteristics that mineralizes and thereby provides rigidity. Such bone has characteristics akin to histological observations on the elongated 'kype' process on lower jaws of male salmon, which may hint at common conserved regulatory underpinnings. Future studies will evaluate the molecular pathways that govern the anterior migration and proliferation of these multipotent cells underlying the belonoids' evolutionary novel jaw extensions.
Collapse
Affiliation(s)
- Ralf F Schneider
- Lehrstuhl für Zoologie und Evolutionsbiologie, Department of Biology, University of Konstanz, Konstanz, Germany
- Department of Marine Ecology, GEOMAR, Kiel, Germany
| | - Helen M Gunter
- Lehrstuhl für Zoologie und Evolutionsbiologie, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Inken Salewski
- Lehrstuhl für Zoologie und Evolutionsbiologie, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Joost M Woltering
- Lehrstuhl für Zoologie und Evolutionsbiologie, Department of Biology, University of Konstanz, Konstanz, Germany
| | - Axel Meyer
- Lehrstuhl für Zoologie und Evolutionsbiologie, Department of Biology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
2
|
Deacon E, Li A, Boivin F, Dvorkin-Gheva A, Cunanan J, Bridgewater D. β-Catenin in the kidney stroma modulates pathways and genes to regulate kidney development. Dev Dyn 2023; 252:1224-1239. [PMID: 37227110 DOI: 10.1002/dvdy.603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Kidney development is regulated by cellular interactions between the ureteric epithelium, mesenchyme, and stroma. Previous studies demonstrate essential roles for stromal β-catenin in kidney development. However, how stromal β-catenin regulates kidney development is not known. We hypothesize that stromal β-catenin modulates pathways and genes that facilitate communications with neighboring cell populations to regulate kidney development. RESULTS We isolated purified stromal cells with wild type, deficient, and overexpressed β-catenin by fluorescence-activated cell sorting and conducted RNA Sequencing. A Gene Ontology network analysis demonstrated that stromal β-catenin modulates key kidney developmental processes, including branching morphogenesis, nephrogenesis and vascular formation. Specific stromal β-catenin candidate target genes that may mediate these effects included secreted, cell-surface and transcriptional factors that regulate branching morphogenesis and nephrogenesis (Wnts, Bmp, Fgfr, Tcf/Lef) and secreted vascular guidance cues (Angpt1, VEGF, Sema3a). We validated established β-catenin targets including Lef1 and novel candidate β-catenin targets including Sema3e which have unknown roles in kidney development. CONCLUSIONS These studies advance our understanding of gene and biological pathway dysregulation in the context of stromal β-catenin misexpression during kidney development. Our findings suggest that during normal kidney development, stromal β-catenin may regulate secreted and cell-surface proteins to communicate with adjacent cell populations.
Collapse
Affiliation(s)
- Erin Deacon
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Anna Li
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Felix Boivin
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Anna Dvorkin-Gheva
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Joanna Cunanan
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Darren Bridgewater
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
3
|
Schnell J, Achieng M, Lindström NO. Principles of human and mouse nephron development. Nat Rev Nephrol 2022; 18:628-642. [PMID: 35869368 DOI: 10.1038/s41581-022-00598-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2022] [Indexed: 12/17/2022]
Abstract
The mechanisms underlying kidney development in mice and humans is an area of intense study. Insights into kidney organogenesis have the potential to guide our understanding of the origin of congenital anomalies and enable the assembly of genetic diagnostic tools. A number of studies have delineated signalling nodes that regulate positional identities and cell fates of nephron progenitor and precursor cells, whereas cross-species comparisons have markedly enhanced our understanding of conserved and divergent features of mammalian kidney organogenesis. Greater insights into the complex cellular movements that occur as the proximal-distal axis is established have challenged our understanding of nephron patterning and provided important clues to the elaborate developmental context in which human kidney diseases can arise. Studies of kidney development in vivo have also facilitated efforts to recapitulate nephrogenesis in kidney organoids in vitro, by providing a detailed blueprint of signalling events, cell movements and patterning mechanisms that are required for the formation of correctly patterned nephrons and maturation of physiologically functional apparatus that are responsible for maintaining human health.
Collapse
Affiliation(s)
- Jack Schnell
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at University of Southern California, Los Angeles, CA, USA
| | - MaryAnne Achieng
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at University of Southern California, Los Angeles, CA, USA
| | - Nils Olof Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Klumpe HE, Langley MA, Linton JM, Su CJ, Antebi YE, Elowitz MB. The context-dependent, combinatorial logic of BMP signaling. Cell Syst 2022; 13:388-407.e10. [PMID: 35421361 PMCID: PMC9127470 DOI: 10.1016/j.cels.2022.03.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/23/2021] [Accepted: 03/18/2022] [Indexed: 12/12/2022]
Abstract
Cell-cell communication systems typically comprise families of ligand and receptor variants that function together in combinations. Pathway activation depends on the complex way in which ligands are presented extracellularly and receptors are expressed by the signal-receiving cell. To understand the combinatorial logic of such a system, we systematically measured pairwise bone morphogenetic protein (BMP) ligand interactions in cells with varying receptor expression. Ligands could be classified into equivalence groups based on their profile of positive and negative synergies with other ligands. These groups varied with receptor expression, explaining how ligands can functionally replace each other in one context but not another. Context-dependent combinatorial interactions could be explained by a biochemical model based on the competitive formation of alternative signaling complexes with distinct activities. Together, these results provide insights into the roles of BMP combinations in developmental and therapeutic contexts and establish a framework for analyzing other combinatorial, context-dependent signaling systems.
Collapse
Affiliation(s)
- Heidi E Klumpe
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA; Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Matthew A Langley
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - James M Linton
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Christina J Su
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Yaron E Antebi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Michael B Elowitz
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Department of Applied Physics, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
5
|
Harding P, Cunha DL, Moosajee M. Animal and cellular models of microphthalmia. THERAPEUTIC ADVANCES IN RARE DISEASE 2021; 2:2633004021997447. [PMID: 37181112 PMCID: PMC10032472 DOI: 10.1177/2633004021997447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/02/2021] [Indexed: 05/16/2023]
Abstract
Microphthalmia is a rare developmental eye disorder affecting 1 in 7000 births. It is defined as a small (axial length ⩾2 standard deviations below the age-adjusted mean) underdeveloped eye, caused by disruption of ocular development through genetic or environmental factors in the first trimester of pregnancy. Clinical phenotypic heterogeneity exists amongst patients with varying levels of severity, and associated ocular and systemic features. Up to 11% of blind children are reported to have microphthalmia, yet currently no treatments are available. By identifying the aetiology of microphthalmia and understanding how the mechanisms of eye development are disrupted, we can gain a better understanding of the pathogenesis. Animal models, mainly mouse, zebrafish and Xenopus, have provided extensive information on the genetic regulation of oculogenesis, and how perturbation of these pathways leads to microphthalmia. However, differences exist between species, hence cellular models, such as patient-derived induced pluripotent stem cell (iPSC) optic vesicles, are now being used to provide greater insights into the human disease process. Progress in 3D cellular modelling techniques has enhanced the ability of researchers to study interactions of different cell types during eye development. Through improved molecular knowledge of microphthalmia, preventative or postnatal therapies may be developed, together with establishing genotype-phenotype correlations in order to provide patients with the appropriate prognosis, multidisciplinary care and informed genetic counselling. This review summarises some key discoveries from animal and cellular models of microphthalmia and discusses how innovative new models can be used to further our understanding in the future. Plain language summary Animal and Cellular Models of the Eye Disorder, Microphthalmia (Small Eye) Microphthalmia, meaning a small, underdeveloped eye, is a rare disorder that children are born with. Genetic changes or variations in the environment during the first 3 months of pregnancy can disrupt early development of the eye, resulting in microphthalmia. Up to 11% of blind children have microphthalmia, yet currently no treatments are available. By understanding the genes necessary for eye development, we can determine how disruption by genetic changes or environmental factors can cause this condition. This helps us understand why microphthalmia occurs, and ensure patients are provided with the appropriate clinical care and genetic counselling advice. Additionally, by understanding the causes of microphthalmia, researchers can develop treatments to prevent or reduce the severity of this condition. Animal models, particularly mice, zebrafish and frogs, which can also develop small eyes due to the same genetic/environmental changes, have helped us understand the genes which are important for eye development and can cause birth eye defects when disrupted. Studying a patient's own cells grown in the laboratory can further help researchers understand how changes in genes affect their function. Both animal and cellular models can be used to develop and test new drugs, which could provide treatment options for patients living with microphthalmia. This review summarises the key discoveries from animal and cellular models of microphthalmia and discusses how innovative new models can be used to further our understanding in the future.
Collapse
Affiliation(s)
| | | | - Mariya Moosajee
- UCL Institute of Ophthalmology, 11-43 Bath
Street, London, EC1V 9EL, UK
- Moorfields Eye Hospital NHS Foundation Trust,
London, UK
- Great Ormond Street Hospital for Children NHS
Foundation Trust, London, UK
- The Francis Crick Institute, London, UK
| |
Collapse
|
6
|
Molecular detection of maturation stages in the developing kidney. Dev Biol 2020; 470:62-73. [PMID: 33197428 DOI: 10.1016/j.ydbio.2020.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/31/2020] [Accepted: 11/03/2020] [Indexed: 12/21/2022]
Abstract
Recent advances in stem cell biology have enabled the generation of kidney organoids in vitro, and further maturation of these organoids is observed after experimental transplantation. However, the current organoids remain immature and their precise maturation stages are difficult to determine because of limited information on developmental stage-dependent gene expressions in the kidney in vivo. To establish relevant molecular coordinates, we performed single-cell RNA sequencing (scRNA-seq) on developing kidneys at different stages in the mouse. By selecting genes that exhibited upregulation at birth compared with embryonic day 15.5 as well as cell lineage-specific expression, we generated gene lists correlated with developmental stages in individual cell lineages. Application of these lists to transplanted embryonic kidneys revealed that most cell types, other than the collecting ducts, exhibited similar maturation to kidneys at the neonatal stage in vivo, revealing non-synchronous maturation across the cell lineages. Thus, our scRNA-seq data can serve as useful molecular coordinates to assess the maturation of developing kidneys and eventually of kidney organoids.
Collapse
|
7
|
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
|
8
|
Kim AD, Lake BB, Chen S, Wu Y, Guo J, Parvez RK, Tran T, Thornton ME, Grubbs B, McMahon JA, Zhang K, McMahon AP. Cellular Recruitment by Podocyte-Derived Pro-migratory Factors in Assembly of the Human Renal Filter. iScience 2019; 20:402-414. [PMID: 31622881 PMCID: PMC6817668 DOI: 10.1016/j.isci.2019.09.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/21/2019] [Accepted: 09/23/2019] [Indexed: 12/20/2022] Open
Abstract
Analysis of kidney disease-causing genes and pathology resulting from systemic diseases highlight the importance of the kidney's filtering system, the renal corpuscles. To elucidate the developmental processes that establish the renal corpuscle, we performed single-nucleus droplet-based sequencing of the human fetal kidney. This enabled the identification of nephron, interstitial, and vascular cell types that together generate the renal corpuscles. Trajectory analysis identified transient developmental gene expression, predicting precursors or mature podocytes express FBLN2, BMP4, or NTN4, in conjunction with recruitment, differentiation, and modeling of vascular and mesangial cell types into a functional filter. In vitro studies provide evidence that these factors exhibit angiogenic or mesangial recruiting and inductive properties consistent with a key organizing role for podocyte precursors in kidney development. Together these studies define a spatiotemporal developmental program for the primary filtration unit of the human kidney and provide novel insights into cell interactions regulating co-assembly of constituent cell types.
Collapse
Affiliation(s)
- Albert D Kim
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Blue B Lake
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Song Chen
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Yan Wu
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Riana K Parvez
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Tracy Tran
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Matthew E Thornton
- Maternal Fetal Medicine Division, University of Southern California, Los Angeles, CA, USA
| | - Brendan Grubbs
- Maternal Fetal Medicine Division, University of Southern California, Los Angeles, CA, USA
| | - Jill A McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Kun Zhang
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA.
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
9
|
Kim HS, Neugebauer J, McKnite A, Tilak A, Christian JL. BMP7 functions predominantly as a heterodimer with BMP2 or BMP4 during mammalian embryogenesis. eLife 2019; 8:48872. [PMID: 31566563 PMCID: PMC6785266 DOI: 10.7554/elife.48872] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/28/2019] [Indexed: 12/15/2022] Open
Abstract
BMP7/BMP2 or BMP7/BMP4 heterodimers are more active than homodimers in vitro, but it is not known whether these heterodimers signal in vivo. To test this, we generated knock in mice carrying a mutation (Bmp7R-GFlag) that prevents proteolytic activation of the dimerized BMP7 precursor protein. This mutation eliminates the function of BMP7 homodimers and all other BMPs that normally heterodimerize with BMP7. While Bmp7 null homozygotes are live born, Bmp7R-GFlag homozygotes are embryonic lethal and have broadly reduced BMP activity. Furthermore, compound heterozygotes carrying the Bmp7R-G allele together with a null allele of Bmp2 or Bmp4 die during embryogenesis with defects in ventral body wall closure and/or the heart. Co-immunoprecipitation assays confirm that endogenous BMP4/7 heterodimers exist. Thus, BMP7 functions predominantly as a heterodimer with BMP2 or BMP4 during mammalian development, which may explain why mutations in either Bmp4 or Bmp7 lead to a similar spectrum of congenital defects in humans.
Collapse
Affiliation(s)
- Hyung-Seok Kim
- Department of Neurobiology and Anatomy and Internal Medicine, Division of Hematology and Hematologic Malignancies, School of Medicine, University of Utah, Salt Lake City, United States
| | - Judith Neugebauer
- Department of Neurobiology and Anatomy and Internal Medicine, Division of Hematology and Hematologic Malignancies, School of Medicine, University of Utah, Salt Lake City, United States
| | - Autumn McKnite
- Department of Neurobiology and Anatomy and Internal Medicine, Division of Hematology and Hematologic Malignancies, School of Medicine, University of Utah, Salt Lake City, United States
| | - Anup Tilak
- Department of Cell and Developmental Biology, School of Medicine, Oregon Health and Sciences University, Portland, United States
| | - Jan L Christian
- Department of Neurobiology and Anatomy and Internal Medicine, Division of Hematology and Hematologic Malignancies, School of Medicine, University of Utah, Salt Lake City, United States
| |
Collapse
|
10
|
Bradford STJ, Ranghini EJ, Grimley E, Lee PH, Dressler GR. High-throughput screens for agonists of bone morphogenetic protein (BMP) signaling identify potent benzoxazole compounds. J Biol Chem 2019; 294:3125-3136. [PMID: 30602563 DOI: 10.1074/jbc.ra118.006817] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/27/2018] [Indexed: 12/31/2022] Open
Abstract
Bone morphogenetic protein (BMP) signaling is critical in renal development and disease. In animal models of chronic kidney disease (CKD), re-activation of BMP signaling is reported to be protective by promoting renal repair and regeneration. Clinical use of recombinant BMPs, however, requires harmful doses to achieve efficacy and is costly because of BMPs' complex synthesis. Therefore, alternative strategies are needed to harness the beneficial effects of BMP signaling in CKD. Key aspects of the BMP signaling pathway can be regulated by both extracellular and intracellular molecules. In particular, secreted proteins like noggin and chordin inhibit BMP activity, whereas kielin/chordin-like proteins (KCP) enhance it and attenuate kidney fibrosis or CKD. Clinical development of KCP, however, is precluded by its size and complexity. Therefore, we propose an alternative strategy to enhance BMP signaling by using small molecules, which are simpler to synthesize and more cost-effective. To address our objective, here we developed a small-molecule high-throughput screen (HTS) with human renal cells having an integrated luciferase construct highly responsive to BMPs. We demonstrate the activity of a potent benzoxazole compound, sb4, that rapidly stimulated BMP signaling in these cells. Activation of BMP signaling by sb4 increased the phosphorylation of key second messengers (SMAD-1/5/9) and also increased expression of direct target genes (inhibitors of DNA binding, Id1 and Id3) in canonical BMP signaling. Our results underscore the feasibility of utilizing HTS to identify compounds that mimic key downstream events of BMP signaling in renal cells and have yielded a lead BMP agonist.
Collapse
Affiliation(s)
- Shayna T J Bradford
- From the Department of Pathology and.,the Molecular and Cellular Pathology Graduate Program, School of Medicine, and
| | | | - Edward Grimley
- From the Department of Pathology and.,the Molecular and Cellular Pathology Graduate Program, School of Medicine, and
| | - Pil H Lee
- the Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109
| | | |
Collapse
|
11
|
Stangenberg S, Nguyen LT, Chan YL, Zaky A, Pollock CA, Chen H, Saad S. Maternal L-carnitine supplementation ameliorates renal underdevelopment and epigenetic changes in male mice offspring due to maternal smoking. Clin Exp Pharmacol Physiol 2018; 46:183-193. [PMID: 30290012 DOI: 10.1111/1440-1681.13038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 09/18/2018] [Accepted: 09/27/2018] [Indexed: 01/02/2023]
Abstract
OBJECTIVES Epidemiological and animal studies showed that L-carnitine (LC) supplementation can ameliorate oxidative stress-induced tissues damage. We have previously shown that maternal cigarette smoke exposure (SE) can increase renal oxidative stress in newborn offspring with postnatal kidney underdevelopment and renal dysfunction in adulthood, which were normalised by LC administration in the SE dams during pregnancy. Exposure to an adverse intrauterine environment may lead to alteration in the epigenome, a mechanism by which adverse prenatal conditions increase the susceptibility to chronic disease later in life. The current study aimed to determine whether maternal SE induces epigenetic changes in the offspring's kidney are associated with renal underdevelopment, and the protective effect of maternal LC supplementation. METHOD Female Balb/c mice (7 weeks) were exposed to cigarette smoke (SE) or air (Sham) for 6 weeks prior to mating, during gestation and lactation. A subgroup of the SE dams received LC via drinking water (SE + LC, 1.5 mmol/L) throughout gestation and lactation. Male offspring were studied at postnatal day (P)1, P20, and 13 weeks. RESULTS Maternal SE altered the expression of renal development markers glial cell line-derived neurotrophic factor and fibroblast growth factor 2, which were associated with increased renal global DNA methylation and DNA methyltransferase 1 mRNA expression at birth. These disorders were reversed by maternal LC administration. CONCLUSION The effect of maternal SE on renal underdevelopment involves global epigenetic alterations from birth, which can be prevented by maternal LC supplementation.
Collapse
Affiliation(s)
- Stefanie Stangenberg
- Renal Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Long The Nguyen
- Renal Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Yik Lung Chan
- RCMB, Woolcock Institute of Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Amgad Zaky
- Renal Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Carol A Pollock
- Renal Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Hui Chen
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Sonia Saad
- Renal Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, New South Wales, Australia.,Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
12
|
Vigolo E, Markó L, Hinze C, Müller DN, Schmidt-Ullrich R, Schmidt-Ott KM. Canonical BMP signaling in tubular cells mediates recovery after acute kidney injury. Kidney Int 2018; 95:108-122. [PMID: 30447934 DOI: 10.1016/j.kint.2018.08.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/03/2018] [Accepted: 08/16/2018] [Indexed: 10/27/2022]
Abstract
Bone morphogenetic protein (BMP) signaling has been shown to modulate the development of renal fibrosis in animal models of kidney injury, but the downstream mediators are incompletely understood. In wild-type mice, canonical BMP signaling mediated by SMAD1/5/8 transcription factors was constitutively active in healthy renal tubules, transiently down-regulated after ischemia reperfusion injury (IRI), and reactivated during successful tubular regeneration. We then induced IRI in mice with a tubular-specific BMP receptor 1A (BMPR1A) deletion. These mice failed to reactivate SMAD1/5/8 signaling in the post-ischemic phase and developed renal fibrosis after injury. Using unbiased genomic analyses, we identified three genes encoding inhibitor of DNA-binding (ID) proteins (Id1, Id2, and Id4) as key targets of BMPR1A-SMAD1/5/8 signaling. BMPR1A-deficient mice failed to re-induce these targets following IRI. Instead, BMPR1A-deficiency resulted in activation of pro-fibrotic signaling proteins that are normally repressed by ID proteins, namely, p38 mitogen-activated protein kinase and cell cycle inhibitor p27. These data indicate that the post-ischemic activation of canonical BMP signaling acts endogenously to repress pro-fibrotic signaling in tubular cells and may help to prevent the progression of acute kidney injury to chronic kidney disease.
Collapse
Affiliation(s)
- Emilia Vigolo
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Lajos Markó
- Experimental and Clinical Research Center, a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany; Berlin Institute of Health, Berlin, Germany
| | - Christian Hinze
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Berlin Institute of Health, Berlin, Germany; Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dominik N Müller
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Experimental and Clinical Research Center, a joint cooperation between the Max Delbrück Center for Molecular Medicine and the Charité Medical Faculty, Berlin, Germany; Berlin Institute of Health, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Ruth Schmidt-Ullrich
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Kai M Schmidt-Ott
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; Berlin Institute of Health, Berlin, Germany; Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
13
|
van der Ven AT, Vivante A, Hildebrandt F. Novel Insights into the Pathogenesis of Monogenic Congenital Anomalies of the Kidney and Urinary Tract. J Am Soc Nephrol 2017; 29:36-50. [PMID: 29079659 DOI: 10.1681/asn.2017050561] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Congenital anomalies of the kidneys and urinary tract (CAKUT) comprise a large spectrum of congenital malformations ranging from severe manifestations, such as renal agenesis, to potentially milder conditions, such as vesicoureteral reflux. CAKUT causes approximately 40% of ESRD that manifests within the first three decades of life. Several lines of evidence indicate that CAKUT is often caused by recessive or dominant mutations in single (monogenic) genes. To date, approximately 40 monogenic genes are known to cause CAKUT if mutated, explaining 5%-20% of patients. However, hundreds of different monogenic CAKUT genes probably exist. The discovery of novel CAKUT-causing genes remains challenging because of this pronounced heterogeneity, variable expressivity, and incomplete penetrance. We here give an overview of known genetic causes for human CAKUT and shed light on distinct renal morphogenetic pathways that were identified as relevant for CAKUT in mice and humans.
Collapse
Affiliation(s)
- Amelie T van der Ven
- Divison of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Asaf Vivante
- Divison of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Friedhelm Hildebrandt
- Divison of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
14
|
Tsujimura T, Idei M, Yoshikawa M, Takase O, Hishikawa K. Roles and regulation of bone morphogenetic protein-7 in kidney development and diseases. World J Stem Cells 2016; 8:288-296. [PMID: 27679685 PMCID: PMC5031890 DOI: 10.4252/wjsc.v8.i9.288] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/12/2016] [Accepted: 07/22/2016] [Indexed: 02/07/2023] Open
Abstract
The gene encoding bone morphogenetic protein-7 (BMP7) is expressed in the developing kidney in embryos and also in the mature organ in adults. During kidney development, expression of BMP7 is essential to determine the final number of nephrons in and proper size of the organ. The secreted BMP7 acts on the nephron progenitor cells to exert its dual functions: To maintain and expand the progenitor population and to provide them with competence to respond to differentiation cues, each relying on distinct signaling pathways. Intriguingly, in the adult organ, BMP7 has been implicated in protection against and regeneration from injury. Exogenous administration of recombinant BMP7 to animal models of kidney diseases has shown promising effects in counteracting inflammation, apoptosis and fibrosis evoked upon injury. Although the expression pattern of BMP7 has been well described, the mechanisms by which it is regulated have remained elusive and the processes by which the secretion sites of BMP7 impinge upon its functions in kidney development and diseases have not yet been assessed. Understanding the regulatory mechanisms will pave the way towards gaining better insight into the roles of BMP7, and to achieving desired control of the gene expression as a therapeutic strategy for kidney diseases.
Collapse
|
15
|
Manson SR, Austin PF, Guo Q, Moore KH. BMP-7 Signaling and its Critical Roles in Kidney Development, the Responses to Renal Injury, and Chronic Kidney Disease. VITAMINS AND HORMONES 2016; 99:91-144. [PMID: 26279374 DOI: 10.1016/bs.vh.2015.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD) is a significant health problem that most commonly results from congenital abnormalities in children and chronic renal injury in adults. The therapeutic potential of BMP-7 was first recognized nearly two decades ago with studies demonstrating its requirement for kidney development and ability to inhibit the pathogenesis of renal injury in models of CKD. Since this time, our understanding of CKD has advanced considerably and treatment strategies have evolved with the identification of many additional signaling pathways, cell types, and pathologic processes that contribute to disease progression. The purpose of this review is to revisit the seminal studies that initially established the importance of BMP-7, highlight recent advances in BMP-7 research, and then integrate this knowledge with current research paradigms. We will provide an overview of the evolutionarily conserved roles of BMP proteins and the features that allow BMP signaling pathways to function as critical signaling nodes for controlling biological processes, including those related to CKD. We will discuss the multifaceted functions of BMP-7 during kidney development and the potential for alterations in BMP-7 signaling to result in congenital abnormalities and pediatric kidney disease. We will summarize the renal protective effects of recombinant BMP-7 in experimental models of CKD and then propose a model to describe the potential physiological role of endogenous BMP-7 in the innate repair mechanisms of the kidneys that respond to renal injury. Finally, we will highlight emerging clinical approaches for applying our knowledge of BMP-7 toward improving the treatment of patients with CKD.
Collapse
Affiliation(s)
- Scott R Manson
- Department of Surgery, Division of Urology, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA.
| | - Paul F Austin
- Department of Surgery, Division of Urology, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA
| | - Qiusha Guo
- Department of Surgery, Division of Urology, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA
| | - Katelynn H Moore
- Department of Surgery, Division of Urology, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA
| |
Collapse
|
16
|
Brown AC, Muthukrishnan SD, Oxburgh L. A synthetic niche for nephron progenitor cells. Dev Cell 2015; 34:229-41. [PMID: 26190145 DOI: 10.1016/j.devcel.2015.06.021] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 05/16/2015] [Accepted: 06/19/2015] [Indexed: 01/06/2023]
Abstract
FGF, BMP, and WNT balance embryonic nephron progenitor cell (NPC) renewal and differentiation. By modulating these pathways, we have created an in vitro niche in which NPCs from embryonic kidneys or derived from human embryonic stem cells can be propagated. NPC cultures expanded up to one billion-fold in this environment can be induced to form tubules expressing nephron differentiation markers. Single-cell culture reveals phenotypic variability within the early CITED1-expressing NPC compartment, indicating that it is a mixture of cells with varying progenitor potential. Furthermore, we find that the developmental age of NPCs does not correlate with propagation capacity, indicating that cessation of nephrogenesis is related to factors other than an intrinsic clock. This in vitro nephron progenitor niche will have important applications for expansion of cells for engraftment and will facilitate investigation of mechanisms that determine the balance between renewal and differentiation in these cells.
Collapse
Affiliation(s)
- Aaron C Brown
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074, USA
| | - Sree Deepthi Muthukrishnan
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074, USA
| | - Leif Oxburgh
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074, USA.
| |
Collapse
|
17
|
Jiang M, Ku WY, Zhou Z, Dellon ES, Falk GW, Nakagawa H, Wang ML, Liu K, Wang J, Katzka DA, Peters JH, Lan X, Que J. BMP-driven NRF2 activation in esophageal basal cell differentiation and eosinophilic esophagitis. J Clin Invest 2015; 125:1557-68. [PMID: 25774506 DOI: 10.1172/jci78850] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 01/29/2015] [Indexed: 12/17/2022] Open
Abstract
Tissue homeostasis requires balanced self-renewal and differentiation of stem/progenitor cells, especially in tissues that are constantly replenished like the esophagus. Disruption of this balance is associated with pathological conditions, including eosinophilic esophagitis (EoE), in which basal progenitor cells become hyperplastic upon proinflammatory stimulation. However, how basal cells respond to the inflammatory environment at the molecular level remains undetermined. We previously reported that the bone morphogenetic protein (BMP) signaling pathway is critical for epithelial morphogenesis in the embryonic esophagus. Here, we address how this pathway regulates tissue homeostasis and EoE development in the adult esophagus. BMP signaling was specifically activated in differentiated squamous epithelium, but not in basal progenitor cells, which express the BMP antagonist follistatin. Previous reports indicate that increased BMP activity promotes Barrett's intestinal differentiation; however, in mice, basal progenitor cell-specific expression of constitutively active BMP promoted squamous differentiation. Moreover, BMP activation increased intracellular ROS levels, initiating an NRF2-mediated oxidative response during basal progenitor cell differentiation. In both a mouse EoE model and human biopsies, reduced squamous differentiation was associated with high levels of follistatin and disrupted BMP/NRF2 pathways. We therefore propose a model in which normal squamous differentiation of basal progenitor cells is mediated by BMP-driven NRF2 activation and basal cell hyperplasia is promoted by disruption of BMP signaling in EoE.
Collapse
|
18
|
WT1 controls antagonistic FGF and BMP-pSMAD pathways in early renal progenitors. Nat Commun 2014; 5:4444. [DOI: 10.1038/ncomms5444] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 06/18/2014] [Indexed: 01/28/2023] Open
|
19
|
Bone morphogenetic protein signaling in nephron progenitor cells. Pediatr Nephrol 2014; 29:531-6. [PMID: 23954916 PMCID: PMC3944211 DOI: 10.1007/s00467-013-2589-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 07/19/2013] [Accepted: 07/24/2013] [Indexed: 02/01/2023]
Abstract
Bone morphogenetic protein (BMP) signaling plays an essential role in many aspects of kidney development, and is a major determinant of outcome in kidney injury. BMP treatment is also an essential component of protocols for differentiation of nephron progenitors from pluripotent stem cells. This review discusses the role of BMP signaling to nephron progenitor cells in each of these contexts.
Collapse
|
20
|
Martínez-Fernández ML, Bermejo-Sánchez E, Fernández B, MacDonald A, Fernández-Toral J, Martínez-Frías ML. Haploinsufficiency ofBMP4gene may be the underlying cause of Frías syndrome. Am J Med Genet A 2013; 164A:338-45. [DOI: 10.1002/ajmg.a.36224] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 08/07/2013] [Indexed: 11/05/2022]
Affiliation(s)
- María Luisa Martínez-Fernández
- CIBER de Enfermedades Raras (CIBERER) (U724); Instituto de Salud Carlos III, Ministerio de Economía y Competitividad; Madrid Spain
- Spanish Collaborative Study of Congenital Malformations (ECEMC); CIAC (Research Center on Congenital Anomalies), Instituto de Salud Carlos III; Madrid Spain
| | - Eva Bermejo-Sánchez
- CIBER de Enfermedades Raras (CIBERER) (U724); Instituto de Salud Carlos III, Ministerio de Economía y Competitividad; Madrid Spain
- Spanish Collaborative Study of Congenital Malformations (ECEMC); CIAC (Research Center on Congenital Anomalies), Instituto de Salud Carlos III; Madrid Spain
- Instituto de Investigación de Enfermedades Raras (IIER); Instituto de Salud Carlos III, Ministerio de Economía y Competitividad; Madrid Spain
| | - Belén Fernández
- Servicio de Neonatología; Hospital Universitario Central de Asturias; Oviedo (Asturias) Spain
| | - Alexandra MacDonald
- Spanish Collaborative Study of Congenital Malformations (ECEMC); CIAC (Research Center on Congenital Anomalies), Instituto de Salud Carlos III; Madrid Spain
| | - Joaquín Fernández-Toral
- Servicio de Genética, Hospital Universitario Central de Asturias, Facultad de Medicina; Universidad de Oviedo; Oviedo (Asturias) Spain
| | - María Luisa Martínez-Frías
- CIBER de Enfermedades Raras (CIBERER) (U724); Instituto de Salud Carlos III, Ministerio de Economía y Competitividad; Madrid Spain
- Spanish Collaborative Study of Congenital Malformations (ECEMC); CIAC (Research Center on Congenital Anomalies), Instituto de Salud Carlos III; Madrid Spain
- Departamento de Farmacología, Facultad de Medicina; Universidad Complutense de Madrid; Madrid Spain
| |
Collapse
|
21
|
Kreidberg JA. WT1 and kidney progenitor cells. Organogenesis 2012; 6:61-70. [PMID: 20885852 DOI: 10.4161/org.6.2.11928] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Accepted: 03/29/2010] [Indexed: 12/31/2022] Open
Abstract
Kidney development has been studied over the past sixty years as a model of embryonic induction during organogenesis. Wilms' tumor-1 (WT1), that encodes a transcription factor and RNA-binding protein, was one of the first tumor suppressor genes identified, and was soon thereafter shown to be associated with syndromic forms of childhood kidney disease and gonadal dysgenesis. Kidney agenesis, resulting from a null mutation in the WT1 gene, was one of the first examples of organ agenesis resulting from a gene targeting experiment. Thus, the study of the WT1 gene and its encoded proteins has been at the forefront of developmental biology, tumor biology and the molecular basis for disease. WT1 is now known to have an important role in kidney progenitor cells during development. This review will discuss recent advances in our understanding of kidney progenitor cells, and the recent identification of WT1 target genes in these cells.
Collapse
Affiliation(s)
- Jordan A Kreidberg
- Department of Medicine, Children's Hospital Boston, Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
22
|
Xu K, Wu X, Shapiro E, Huang H, Zhang L, Hickling D, Deng Y, Lee P, Li J, Lepor H, Grishina I. Bmp7 functions via a polarity mechanism to promote cloacal septation. PLoS One 2012; 7:e29372. [PMID: 22253716 PMCID: PMC3258230 DOI: 10.1371/journal.pone.0029372] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 11/27/2011] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND During normal development in human and other placental mammals, the embryonic cloacal cavity separates along the axial longitudinal plane to give rise to the urethral system, ventrally, and the rectum, dorsally. Defects in cloacal development are very common and present clinically as a rectourethral fistula in about 1 in 5,000 live human births. Yet, the cellular mechanisms of cloacal septation remain poorly understood. METHODOLOGY/PRINCIPAL FINDINGS We previously detected Bone morphogenetic protein 7 (Bmp7) expression in the urorectal mesenchyme (URM), and have shown that loss of Bmp7 function results in the arrest of cloacal septation. Here, we present evidence that cloacal partitioning is driven by Bmp7 signaling in the cloacal endoderm. We performed TUNEL and immunofluorescent analysis on cloacal sections from Bmp7 null and control littermate embryos. We found that loss of Bmp7 results in a dramatic decrease in the endoderm survival and a delay in differentiation. We used immunological methods to show that Bmp7 functions by activating the c-Jun N-terminal kinase (JNK) pathway. We carried out confocal and 3D imaging analysis of mitotic chromosome bundles to show that during normal septation cells in the cloacal endoderm divide predominantly in the apical-basal direction. Loss of Bmp7/JNK signaling results in randomization of mitotic angles in the cloacal endoderm. We also conducted immunohistochemical analysis of human fetal sections to show that BMP/phospho-SMAD and JNK pathways function in the human cloacal region similar as in the mouse. CONCLUSION/SIGNIFICANCE Our results strongly indicate that Bmp7/JNK signaling regulates remodeling of the cloacal endoderm resulting in a topological separation of the urinary and digestive systems. Our study points to the importance of Bmp and JNK signaling in cloacal development and rectourethral malformations.
Collapse
Affiliation(s)
- Kun Xu
- Department of Urology, School of Medicine, New York University, New York, New York, United States of America
- Department of Toxicology, Jilin University, Changchun City, China
| | - Xinyu Wu
- Department of Urology, School of Medicine, New York University, New York, New York, United States of America
- Department of Pathology, School of Medicine, New York University, New York, New York, United States of America
| | - Ellen Shapiro
- Department of Urology, School of Medicine, New York University, New York, New York, United States of America
| | - Honging Huang
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Lixia Zhang
- Department of Urology, School of Medicine, New York University, New York, New York, United States of America
| | - Duane Hickling
- Department of Urology, School of Medicine, New York University, New York, New York, United States of America
| | - Yan Deng
- Microscopy Core, School of Medicine, New York University, New York, New York, United States of America
| | - Peng Lee
- Department of Urology, School of Medicine, New York University, New York, New York, United States of America
- Department of Pathology, School of Medicine, New York University, New York, New York, United States of America
| | - Juan Li
- Department of Toxicology, Jilin University, Changchun City, China
| | - Herbert Lepor
- Department of Urology, School of Medicine, New York University, New York, New York, United States of America
| | - Irina Grishina
- Department of Urology, School of Medicine, New York University, New York, New York, United States of America
| |
Collapse
|
23
|
Lopez-Rios J. Sensing BMP pathway activity by immune detection of phosphorylated R-Smad proteins in mouse embryonic kidney. Methods Mol Biol 2012; 886:267-73. [PMID: 22639269 DOI: 10.1007/978-1-61779-851-1_24] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
At the onset of mammalian kidney development, the ureteric bud invades the surrounding metanephric mesenchyme, and genetic studies in the mouse have shown that BMP pathway activity has to be antagonized in the vicinity of the epithelium, a task performed by the secreted BMP antagonist Grem1. Here, we describe a short protocol that allows for detection of the pattern of BMP canonical signal transduction by using antibodies that specifically recognize the phosphorylated forms of R-Smad proteins (Smad1, Smad5, and Smad8), which provides a way to monitor overall pathway activity in the mammalian embryonic kidney.
Collapse
Affiliation(s)
- Javier Lopez-Rios
- Developmental Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
24
|
Tetralogy of Fallot Associated with Dysplastic Kidneys, Cloacal Anomalies, and Female Pseudohermaphroditism: A Systemic Anomaly of Septation? Case Rep Obstet Gynecol 2012; 2012:502919. [PMID: 22811944 PMCID: PMC3395183 DOI: 10.1155/2012/502919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 05/09/2012] [Indexed: 11/17/2022] Open
Abstract
A 20-week fetus was diagnosed with tetralogy of Fallot and multicystic kidneys. The postmortem study showed missing müllerian structures with small streak ovaries, external male genitalia, and an abnormal cloacal septation (imperforate anus with a sigmoid colon opening in the bladder). As the observed anomalies were related with septation, a mechanism related with the activation of specific growth factors, we discuss the possibility of a disorder in the function of the bone morphogenetic proteins as a common cause for the widespread anomalies found in this fetus.
Collapse
|
25
|
Larman BW, Karolak MJ, Lindner V, Oxburgh L. Distinct bone morphogenetic proteins activate indistinguishable transcriptional responses in nephron epithelia including Notch target genes. Cell Signal 2012; 24:257-64. [PMID: 21945409 PMCID: PMC3205934 DOI: 10.1016/j.cellsig.2011.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 08/22/2011] [Accepted: 09/06/2011] [Indexed: 10/17/2022]
Abstract
Endogenous Bone Morphogenetic Protein (BMP) signaling plays a significant role in the kidney's recovery from acute injury and exogenous administration of BMP7 has therapeutic potential in numerous rodent models of renal injury and disease. However, in the healthy kidney endogenous BMP7 ligand is vigorously counteracted by extracellular antagonists such as USAG1 and CHRDL1. Little is known about the degree of BMP signaling and the ligands driving it in the healthy adult kidney. In this study we characterize basal BMP signaling in the healthy tubular nephron, and show that BMP2 is expressed in proximal nephron epithelial cells. Comparative gene profiling of proximal tubule cell responses to BMP2 and BMP7 does not reveal any qualitative difference, suggesting that identical BMP gene targets may be activated in healthy and injured organs. Interestingly, our gene profiling analysis shows that BMP signaling activates a number of Notch regulated transcription factors, including HEY1. As in other biological systems, HEY1 functions as a negative feedback regulator of BMP2 expression in the proximal tubule. In summary, this work reveals endogenous BMP signaling patterns in the healthy human and mouse kidneys, and identifies novel gene targets, some of which are involved in the complex regulation of BMP signaling in the adult kidney.
Collapse
Affiliation(s)
- Barry W Larman
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME 04074, United States
| | | | | | | |
Collapse
|
26
|
Chi L, Saarela U, Railo A, Prunskaite-Hyyryläinen R, Skovorodkin I, Anthony S, Katsu K, Liu Y, Shan J, Salgueiro AM, Belo JA, Davies J, Yokouchi Y, Vainio SJ. A secreted BMP antagonist, Cer1, fine tunes the spatial organization of the ureteric bud tree during mouse kidney development. PLoS One 2011; 6:e27676. [PMID: 22114682 PMCID: PMC3219680 DOI: 10.1371/journal.pone.0027676] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 10/21/2011] [Indexed: 01/02/2023] Open
Abstract
The epithelial ureteric bud is critical for mammalian kidney development as it generates the ureter and the collecting duct system that induces nephrogenesis in dicrete locations in the kidney mesenchyme during its emergence. We show that a secreted Bmp antagonist Cerberus homologue (Cer1) fine tunes the organization of the ureteric tree during organogenesis in the mouse embryo. Both enhanced ureteric expression of Cer1 and Cer1 knock out enlarge kidney size, and these changes are associated with an altered three-dimensional structure of the ureteric tree as revealed by optical projection tomography. Enhanced Cer1 expression changes the ureteric bud branching programme so that more trifid and lateral branches rather than bifid ones develop, as seen in time-lapse organ culture. These changes may be the reasons for the modified spatial arrangement of the ureteric tree in the kidneys of Cer1+ embryos. Cer1 gain of function is associated with moderately elevated expression of Gdnf and Wnt11, which is also induced in the case of Cer1 deficiency, where Bmp4 expression is reduced, indicating the dependence of Bmp expression on Cer1. Cer1 binds at least Bmp2/4 and antagonizes Bmp signalling in cell culture. In line with this, supplementation of Bmp4 restored the ureteric bud tip number, which was reduced by Cer1+ to bring it closer to the normal, consistent with models suggesting that Bmp signalling inhibits ureteric bud development. Genetic reduction of Wnt11 inhibited the Cer1-stimulated kidney development, but Cer1 did not influence Wnt11 signalling in cell culture, although it did inhibit the Wnt3a-induced canonical Top Flash reporter to some extent. We conclude that Cer1 fine tunes the spatial organization of the ureteric tree by coordinating the activities of the growth-promoting ureteric bud signals Gndf and Wnt11 via Bmp-mediated antagonism and to some degree via the canonical Wnt signalling involved in branching.
Collapse
Affiliation(s)
- Lijun Chi
- Laboratory of Developmental Biology, Department of Medical Biochemistry and Molecular Biology, Center for Cell Matrix Research, Institute of Biomedicine Oulu, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Ulla Saarela
- Laboratory of Developmental Biology, Department of Medical Biochemistry and Molecular Biology, Center for Cell Matrix Research, Institute of Biomedicine Oulu, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Antti Railo
- Laboratory of Developmental Biology, Department of Medical Biochemistry and Molecular Biology, Center for Cell Matrix Research, Institute of Biomedicine Oulu, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Renata Prunskaite-Hyyryläinen
- Laboratory of Developmental Biology, Department of Medical Biochemistry and Molecular Biology, Center for Cell Matrix Research, Institute of Biomedicine Oulu, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Ilya Skovorodkin
- Laboratory of Developmental Biology, Department of Medical Biochemistry and Molecular Biology, Center for Cell Matrix Research, Institute of Biomedicine Oulu, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Shelagh Anthony
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Kenjiro Katsu
- Division of Pattern Formation, Department of Organogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Yu Liu
- Texas A&M Health Science Center, Center for Development and Diseases, Institute of Biosciences and Technology, Houston, Texas, United States of America
| | - Jingdong Shan
- Laboratory of Developmental Biology, Department of Medical Biochemistry and Molecular Biology, Center for Cell Matrix Research, Institute of Biomedicine Oulu, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Ana Marisa Salgueiro
- Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Regenerative Medicine Program, Algarve, Portugal
- IBB-Institute for Biotechnology and Bioengineering, Centro de Biomedicina Molecular e Estrutural, Universidade do Algarve, Faro, Portugal
| | - José António Belo
- Departamento de Ciências Biomédicas e Medicina, Universidade do Algarve, Regenerative Medicine Program, Algarve, Portugal
- IBB-Institute for Biotechnology and Bioengineering, Centro de Biomedicina Molecular e Estrutural, Universidade do Algarve, Faro, Portugal
| | - Jamie Davies
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Yuji Yokouchi
- Division of Pattern Formation, Department of Organogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Seppo J. Vainio
- Laboratory of Developmental Biology, Department of Medical Biochemistry and Molecular Biology, Center for Cell Matrix Research, Institute of Biomedicine Oulu, Biocenter Oulu, University of Oulu, Oulu, Finland
- * E-mail:
| |
Collapse
|
27
|
Oxburgh L, Brown AC, Fetting J, Hill B. BMP signaling in the nephron progenitor niche. Pediatr Nephrol 2011; 26:1491-7. [PMID: 21373777 PMCID: PMC3319359 DOI: 10.1007/s00467-011-1819-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 01/04/2011] [Accepted: 01/25/2011] [Indexed: 12/22/2022]
Abstract
Bone morphogenic proteins (BMPs) play diverse roles in embryonic kidney development, regulating essential aspects of both ureteric bud and nephron development. In this review, we provide an overview of reported expression patterns and functions of BMP signaling components within the nephrogenic zone or nephron progenitor niche of the developing kidney. Reported in situ hybridization results are relatively challenging to interpret and sometimes conflicting. Comparing these with high-resolution microarray gene expression data available in Gudmap, we propose a consensus gene expression pattern indicating that essential components of both the Smad-mediated pathway and the Smad-independent MAPK pathways are expressed in the nephron progenitor cell compartment and may be activated by BMPs, but that cortical interstitium may only be able to respond to BMPs through mitogen activated protein kinase (MAPK) signaling. Localization of phosphorylated Smad transcription factors and studies of a BMP reporter mouse strain however indicate limited transcriptional responsiveness to Smad-mediated signaling in cap mesenchyme. An overview of genetic inactivation, organ culture, and primary cell studies indicates that BMP signaling may elicit two important biological outcomes in the nephrogenic zone: survival of the cap mesenchyme, and the physical segregation of interstitial and progenitor cell compartments. Ongoing studies using a novel primary cell system that establishes the nephrogenic zone ex vivo are pursuing the concept that the balance between Smad-mediated and Smad-independent responses to BMP ligand may underlie these distinct outcomes.
Collapse
Affiliation(s)
- Leif Oxburgh
- Department of Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074, USA.
| | | | | | | |
Collapse
|
28
|
Oxburgh L. Control of the bone morphogenetic protein 7 gene in developmental and adult life. Curr Genomics 2011; 10:223-30. [PMID: 19949543 PMCID: PMC2709933 DOI: 10.2174/138920209788488490] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Revised: 03/20/2009] [Accepted: 03/20/2009] [Indexed: 11/24/2022] Open
Abstract
The TGFβ superfamily growth factor BMP7 performs essential biological functions in embryonic development and regeneration of injured tissue in the adult. BMP7 activity is regulated at numerous levels in the signaling pathway by the expression of extracellular antagonists, decoy receptors and inhibitory cell signaling components. Additionally, expression of the BMP7 gene is tightly controlled both during embryonic development and adult life. In this review, the current status of work on regulation of BMP7 at the genomic level is discussed. In situ hybridization and reporter gene studies have conclusively defined patterns of BMP7 expression in many tissues. Additionally, both in vivo and cell culture studies have defined some of the mechanistic bases for this regulation. In addition to transcriptional activation mediated by binding of activating transcription factors, there is also strong evidence for repression through recruitment of histone modifying enzymes to specific genetic elements. This review summarizes our current understanding of BMP7 gene regulation in embryonic development and adult tissues.
Collapse
Affiliation(s)
- Leif Oxburgh
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074, USA
| |
Collapse
|
29
|
Jenkins RH, Fraser DJ. BMP-6 emerges as a potential major regulator of fibrosis in the kidney. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:964-5. [PMID: 21356347 DOI: 10.1016/j.ajpath.2010.12.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 12/14/2010] [Indexed: 02/06/2023]
Affiliation(s)
- Robert H Jenkins
- Institute of Nephrology, School of Medicine, Cardiff University, Heath Park, Cardiff, United Kingdom
| | | |
Collapse
|
30
|
Dendooven A, van Oostrom O, van der Giezen DM, Leeuwis JW, Snijckers C, Joles JA, Robertson EJ, Verhaar MC, Nguyen TQ, Goldschmeding R. Loss of endogenous bone morphogenetic protein-6 aggravates renal fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1069-79. [PMID: 21356359 DOI: 10.1016/j.ajpath.2010.12.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 11/03/2010] [Accepted: 12/07/2010] [Indexed: 02/03/2023]
Abstract
Bone morphogenetic protein-6 (BMP-6) suppresses inflammatory genes in renal proximal tubular cells and regulates iron metabolism by inducing hepcidin. In diabetic patients, an increase of myofibroblast progenitor cells (MFPCs), also known as fibrocytes, was found to be associated with decreased BMP-6 expression. We hypothesized that loss of endogenous BMP-6 would aggravate renal injury and fibrosis. Wild type (WT) and BMP-6 null mice underwent unilateral ureteral obstruction. In WT mice, ureteral obstruction down-regulated BMP-6. Obstructed kidneys of BMP-6 null mice showed more casts (1.5-fold), epithelial necrosis (1.4-fold), and brush border loss (1.3-fold). This was associated with more inflammation (1.8-fold more CD45(+) cells) and more pronounced overexpression of profibrotic genes for αSMA (2.0-fold), collagen I (6.8-fold), fibronectin (4.3-fold), CTGF (1.8-fold), and PAI-1 (3.8-fold), despite similar BMP-7 expression. Also, 1.3-fold more MFPCs were obtained from BMP-6 null than from WT mononuclear cell cultures, but in vivo only very few MFPCs were observed in obstructed kidneys, irrespective of BMP-6 genotype. The obstructed kidneys of BMP-6 null mice showed 2.2-fold more iron deposition, in association with 3.3-fold higher expression of the oxidative stress marker HO-1. Thus, ureteral obstruction leads to down-regulation of BMP-6 expression, and BMP-6 deficiency aggravates tubulointerstitial damage and fibrosis independent of BMP-7. This process appears to involve loss of both direct anti-inflammatory and antifibrotic action and indirect suppressive effects on renal iron deposition, oxidative stress, and MFPCs.
Collapse
Affiliation(s)
- Amélie Dendooven
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Gonçalves A, Zeller R. Genetic analysis reveals an unexpected role of BMP7 in initiation of ureteric bud outgrowth in mouse embryos. PLoS One 2011; 6:e19370. [PMID: 21552539 PMCID: PMC3084290 DOI: 10.1371/journal.pone.0019370] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 04/04/2011] [Indexed: 11/19/2022] Open
Abstract
Background Genetic analysis in the mouse revealed that GREMLIN1 (GREM1)-mediated antagonism of BMP4 is essential for ureteric epithelial branching as the disruption of ureteric bud outgrowth and renal agenesis in Grem1-deficient embryos is restored by additional inactivation of one Bmp4 allele. Another BMP ligand, BMP7, was shown to control the proliferative expansion of nephrogenic progenitors and its requirement for nephrogenesis can be genetically substituted by Bmp4. Therefore, we investigated whether BMP7 in turn also participates in inhibiting ureteric bud outgrowth during the initiation of metanephric kidney development. Methodology/Principal Findings Genetic inactivation of one Bmp7 allele in Grem1-deficient mouse embryos does not alleviate the bilateral renal agenesis, while complete inactivation of Bmp7 restores ureteric bud outgrowth and branching. In mouse embryos lacking both Grem1 and Bmp7, GDNF/WNT11 feedback signaling and the expression of the Etv4 target gene, which regulates formation of the invading ureteric bud tip, are restored. In contrast to the restoration of ureteric bud outgrowth and branching, nephrogenesis remains aberrant as revealed by the premature loss of Six2 expressing nephrogenic progenitor cells. Therefore, very few nephrons develop in kidneys lacking both Grem1 and Bmp7 and the resulting dysplastic phenotype is indistinguishable from the one of Bmp7-deficient mouse embryos. Conclusions/Significance Our study reveals an unexpected inhibitory role of BMP7 during the onset of ureteric bud outgrowth. As BMP4, BMP7 and GREM1 are expressed in distinct mesenchymal and epithelial domains, the localized antagonistic interactions of GREM1 with BMPs could restrict and guide ureteric bud outgrowth and branching. The robustness and likely significant redundancy of the underlying signaling system is evidenced by the fact that global reduction of Bmp4 or inactivation of Bmp7 are both able to restore ureteric bud outgrowth and epithelial branching in Grem1-deficient mouse embryos.
Collapse
Affiliation(s)
- Alexandre Gonçalves
- Developmental Genetics, Department of Biomedicine, University of Basel Medical Faculty, Basel, Switzerland
| | - Rolf Zeller
- Developmental Genetics, Department of Biomedicine, University of Basel Medical Faculty, Basel, Switzerland
- * E-mail:
| |
Collapse
|
32
|
Abstract
The germ cell lineage is our lifelong reservoir of reproductive stem cells and our mechanism for transmitting genes to future generations. These highly specialised cells are specified early during development and then migrate to the embryonic gonads where sex differentiation occurs. Germ cell sex differentiation is directed by the somatic gonadal environment and is characterised by two distinct cell cycle states that are maintained until after birth. In the mouse, XY germ cells in a testis cease mitotic proliferation and enter G(1)/G(0) arrest from 12.5 dpc, while XX germ cells in an ovary enter prophase I of meiosis from 13.5 dpc. This chapter discusses the factors known to control proliferation and survival of germ cells during their journey of specification to sex differentiation during development.
Collapse
Affiliation(s)
- Cassy M Spiller
- Division of Molecular Genetics and Development, Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | | |
Collapse
|
33
|
Tsuji K, Cox K, Gamer L, Graf D, Economides A, Rosen V. Conditional deletion of BMP7 from the limb skeleton does not affect bone formation or fracture repair. J Orthop Res 2010; 28:384-9. [PMID: 19780203 PMCID: PMC2862367 DOI: 10.1002/jor.20996] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
While the osteoinductive activity of recombinant bone morphogenetic protein 7 (BMP7) is well established, evaluation of the role of endogenous BMP7 in bone formation and fracture healing has been hampered by perinatal lethality in BMP7 knockout mice. Here we employ conditional deletion of BMP7 from the embryonic limb prior to the onset of skeletogenesis to create limb bones lacking BMP7. We find that the absence of locally produced BMP7 has no effect on postnatal limb growth, articular cartilage formation, maintenance of bone mass, or fracture healing. Our data suggest that other BMPs present in adult bone are sufficient to compensate for the absence of BMP7.
Collapse
Affiliation(s)
- Kunikazu Tsuji
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, Massachusetts 02115
| | - Karen Cox
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, Massachusetts 02115
| | - Laura Gamer
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, Massachusetts 02115
| | - Daniel Graf
- Institute of Immunology, Biomedical Sciences Research Center, Athens, Greece
| | | | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Boston, Massachusetts 02115
| |
Collapse
|
34
|
Matluk N, Rochira JA, Karaczyn A, Adams T, Verdi JM. A role for NRAGE in NF-kappaB activation through the non-canonical BMP pathway. BMC Biol 2010; 8:7. [PMID: 20100315 PMCID: PMC2829509 DOI: 10.1186/1741-7007-8-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Accepted: 01/25/2010] [Indexed: 11/15/2022] Open
Abstract
Background Previous studies have linked neurotrophin receptor-interacting MAGE protein to the bone morphogenic protein signaling pathway and its effect on p38 mediated apoptosis of neural progenitor cells via the XIAP-Tak1-Tab1 complex. Its effect on NF-κB has yet to be explored. Results Herein we report that NRAGE, via the same XIAP-Tak1-Tab1 complex, is required for the phosphorylation of IKK -α/β and subsequent transcriptional activation of the p65 subunit of NF-κB. Ablation of endogenous NRAGE by siRNA inhibited NF-κB pathway activation, while ablation of Tak1 and Tab1 by morpholino inhibited overexpression of NRAGE from activating NF-κB. Finally, cytokine profiling of an NRAGE over-expressing stable line revealed the expression of macrophage migration inhibitory factor. Conclusion Modulation of NRAGE expression revealed novel roles in regulating NF-κB activity in the non-canonical bone morphogenic protein signaling pathway. The expression of macrophage migration inhibitory factor by bone morphogenic protein -4 reveals novel crosstalk between an immune cytokine and a developmental pathway.
Collapse
Affiliation(s)
- Nicholas Matluk
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074, USA
| | | | | | | | | |
Collapse
|
35
|
Yun S, Saijoh Y, Hirokawa KE, Kopinke D, Murtaugh LC, Monuki ES, Levine EM. Lhx2 links the intrinsic and extrinsic factors that control optic cup formation. Development 2009; 136:3895-906. [PMID: 19906857 DOI: 10.1242/dev.041202] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
A crucial step in eye organogenesis is the transition of the optic vesicle into the optic cup. Several transcription factors and extracellular signals mediate this transition, but whether a single factor links them into a common genetic network is unclear. Here, we provide evidence that the LIM homeobox gene Lhx2, which is expressed in the optic neuroepithelium, fulfils such a role. In Lhx2(-/-) mouse embryos, eye field specification and optic vesicle morphogenesis occur, but development arrests prior to optic cup formation in both the optic neuroepithelium and lens ectoderm. This is accompanied by failure to maintain or initiate the expression patterns of optic-vesicle-patterning and lens-inducing determinants. Of the signaling pathways examined, only BMP signaling is noticeably altered and Bmp4 and Bmp7 mRNAs are undetectable. Lhx2(-/-) optic vesicles and lens ectoderm upregulate Pax2, Fgf15 and Sox2 in response to BMP treatments, and Lhx2 genetic mosaics reveal that transcription factors, including Vsx2 and Mitf, require Lhx2 cell-autonomously for their expression. Our data indicate that Lhx2 is required for optic vesicle patterning and lens formation in part by regulating BMP signaling in an autocrine manner in the optic neuroepithelium and in a paracrine manner in the lens ectoderm. We propose a model in which Lhx2 is a central link in a genetic network that coordinates the multiple pathways leading to optic cup formation.
Collapse
Affiliation(s)
- Sanghee Yun
- Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Abnormalities of kidney and urinary tract development are the most common cause of end-stage kidney failure in childhood in the United States. Over the past 20 years, the advent of mutant and transgenic mice and the manipulation of gene expression in other animal models has resulted in major advances in identification of the cellular and molecular mechanisms that direct kidney morphogenesis, providing insights into the pathophysiology of renal and urologic anomalies. This review focuses on the molecular mechanisms that define kidney progenitor cell populations, induce nephron formation within the metanephric mesenchyme, initiate and organize ureteric bud branching, and participate in terminal differentiation of the nephron. Highlighted are common signaling pathways that function at multiple stages during kidney development, including signaling via Wnts, bone morphogenic proteins, fibroblast growth factor, sonic hedgehog, RET/glial cell-derived neurotrophic factor, and notch pathways. Also emphasized are the roles of transcription factors Odd1, Eya1, Pax2, Lim1, and WT-1 in directing renal development. Areas requiring future investigation include the factors that modulate signaling pathways to provide temporal and site-specific effects. The evolution of our understanding of the cellular and molecular mechanisms of kidney development may provide methods for improved diagnosis of renal anomalies and, hopefully, targets for intervention for this common cause of childhood end-stage kidney disease.
Collapse
Affiliation(s)
- Kimberly J Reidy
- Department of Pediatrics/Division of Pediatric Nephrology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, 3415 Bainbridge Ave, Bronx, NY 10467, USA
| | | |
Collapse
|
37
|
Batchelder CA, Lee CCI, Matsell DG, Yoder MC, Tarantal AF. Renal ontogeny in the rhesus monkey (Macaca mulatta) and directed differentiation of human embryonic stem cells towards kidney precursors. Differentiation 2009; 78:45-56. [PMID: 19500897 PMCID: PMC2744219 DOI: 10.1016/j.diff.2009.05.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2008] [Revised: 03/16/2009] [Accepted: 05/03/2009] [Indexed: 01/14/2023]
Abstract
The development of the metanephric kidney was studied immunohistochemically across gestation in monkeys to identify markers of cell specification, and to aid in developing experimental paradigms for renal precursor differentiation from human embryonic stem cells (hESC). PAX2, an important kidney developmental marker, was expressed at the tips of the ureteric bud, in the surrounding condensing mesenchyme, and in the renal vesicle. Vimentin, a mesenchymal and renal marker, was strongly expressed in the metanephric blastema then found to be limited to the glomerulus and interstitial cells of the medulla and cortex. A model of gene expression based on human and nonhuman primate renal ontogeny was developed and incorporated into studies of hESC differentiation. Spontaneous hESC differentiation revealed markers of metanephric mesenchyme (OSR1, PAX2, SIX2, WT1) that increased over time, followed by upregulation of kidney precursor markers (EYA1, LIM1, CD24). Directed hESC differentiation was also evaluated with the addition of retinoic acid, Activin-A, and BMP-4 or BMP-7, and using different culture substrate conditions. Of the culture substrates studied, gelatin most closely recapitulated the anticipated directed developmental pattern of renal gene expression. No differences were found when BMP-4 and BMP-7 were compared with baseline conditions. PAX2 and Vimentin immunoreactivity in differentiating hESC was also similar to the renal precursor patterns reported for human fetal kidneys and findings described in rhesus monkeys. The results of these studies are as follows: (1) provide additional data to support that rhesus monkey kidney development parallels that of humans, and (2) provide a useful model for hESC directed differentiation towards renal precursors.
Collapse
Affiliation(s)
- Cynthia A. Batchelder
- Center of Excellence in Translational Human Stem Cell Research, University of California, Davis, CA, USA
- California National Primate Research Center, University of California, Davis, CA, USA
| | - C. Chang I. Lee
- Center of Excellence in Translational Human Stem Cell Research, University of California, Davis, CA, USA
- California National Primate Research Center, University of California, Davis, CA, USA
| | - Douglas G. Matsell
- Center of Excellence in Translational Human Stem Cell Research, University of California, Davis, CA, USA
- Child and Family Research Institute, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mervin C. Yoder
- Center of Excellence in Translational Human Stem Cell Research, University of California, Davis, CA, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Alice F. Tarantal
- Center of Excellence in Translational Human Stem Cell Research, University of California, Davis, CA, USA
- California National Primate Research Center, University of California, Davis, CA, USA
- Department of Pediatrics and Cell Biology and Human Anatomy, University of California, Davis, CA, USA
| |
Collapse
|
38
|
Bessa PC, Casal M, Reis RL. Bone morphogenetic proteins in tissue engineering: the road from the laboratory to the clinic, part I (basic concepts). J Tissue Eng Regen Med 2008; 2:1-13. [PMID: 18293427 DOI: 10.1002/term.63] [Citation(s) in RCA: 233] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Discovered in 1965, bone morphogenetic proteins (BMPs) are a group of cytokines from the transforming growth factor-beta (TGFbeta) superfamily with significant roles in bone and cartilage formation. BMPs are used as powerful osteoinductive components of diverse tissue-engineering products for the healing of bone. Several BMPs with different physiological roles have been identified in humans. The purpose of this review is to cover the biological function of the main members of BMP family, the latest research on BMPs signalling pathways and advances in the production of recombinant BMPs for tissue engineering purposes.
Collapse
Affiliation(s)
- P C Bessa
- 3Bs Research Group, Biomaterials, Biodegradables and Biomimetics, Department of Polymer Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
| | | | | |
Collapse
|
39
|
Blank U, Seto ML, Adams DC, Wojchowski DM, Karolak MJ, Oxburgh L. An in vivo reporter of BMP signaling in organogenesis reveals targets in the developing kidney. BMC DEVELOPMENTAL BIOLOGY 2008; 8:86. [PMID: 18801194 PMCID: PMC2561030 DOI: 10.1186/1471-213x-8-86] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2008] [Accepted: 09/18/2008] [Indexed: 12/22/2022]
Abstract
Background Bone morphogenetic proteins (BMPs) regulate essential processes during organogenesis, and a functional understanding of these secreted proteins depends on identification of their target cells. In this study, we generate a transgenic reporter for organogenesis studies that we use to define BMP pathway activation in the developing kidney. Results Mouse strains reporting on BMP pathway activation were generated by transgenically expressing β-galactosidase under the control of BMP responsive elements from Id1. Reporter expression corresponds well with immunoassays for pathway activation in all organs studied, validating the model. Using these reporters we have generated a detailed map of cellular targets of BMP signaling in the developing kidney. We find that SMAD dependent BMP signaling is active in collecting duct trunks, but not tips. Furthermore, glomerular endothelial cells, and proximal nephron tubules from the renal vesicle stage onward show pathway activation. Surprisingly, little activation is detected in the nephrogenic zone of the kidney, and in organ culture BMP treatment fails to activate SMAD dependent BMP signaling in nephron progenitor cells. In contrast, signaling is efficiently induced in collecting duct tips. Conclusion Transgenic reporters driven by control elements from BMP responsive genes such as Id1 offer significant advantages in sensitivity and consistency over immunostaining for studies of BMP pathway activation. They also provide opportunities for analysis of BMP signaling in organ and primary cell cultures subjected to experimental manipulation. Using such a reporter, we made the surprising finding that SMAD dependent BMP signaling is inactive in nephron progenitors, and that these cells are refractory to activation by applied growth factors. Furthermore, we find that the BMP pathway is not normally active in collecting duct tips, but that it can be ectopically activated by BMP treatment, offering a possible explanation for the inhibitory effects of BMP treatment on collecting duct growth and branching.
Collapse
Affiliation(s)
- Ulrika Blank
- Department of Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Abdel-Hakeem AK, Henry TQ, Magee TR, Desai M, Ross MG, Mansano RZ, Torday JS, Nast CC. Mechanisms of impaired nephrogenesis with fetal growth restriction: altered renal transcription and growth factor expression. Am J Obstet Gynecol 2008; 199:252.e1-7. [PMID: 18639218 PMCID: PMC2932650 DOI: 10.1016/j.ajog.2008.05.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2008] [Revised: 03/01/2008] [Accepted: 05/24/2008] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Maternal food restriction during pregnancy results in growth-restricted newborns and reduced glomerular number, contributing to programmed offspring hypertension. We investigated whether reduced nephrogenesis may be programmed by dysregulation of factors controlling ureteric bud branching and mesenchyme to epithelial transformation. STUDY DESIGN At 10 to 20 days' gestation, Sprague Dawley pregnant rats (n = 6/group) received ad libitum food; food-restricted rats were 50% food restricted. At embryonic day 20, messenger ribonucleic acid (mRNA) and protein expression of Wilms' tumor 1 gene product (WT1), paired box transcription factor (Pax)-2, fibroblast growth factor (FGF)-2, glial cell line-derived neurotrophic factor (GDNF), cRET, wingless-type mouse mammary tumor virus integration site (WNT)4, WNT11, bone morphogenetic protein (BMP)-4, BMP7, and FGF7 were determined by real-time polymerase chain reaction and Western blotting. RESULTS Maternal food restriction resulted in up-regulated mRNA expression for WT1, FGF2, and BMP7, whereas Pax2, GDNF, FGF7, BMP4, WNT4, and WNT11 mRNAs were down-regulated. Protein expression was concordant for WT1, GDNF, Pax2, FGF7, BMP4, and WNT4. CONCLUSION Maternal food restriction altered gene expression of fetal renal transcription and growth factors and likely contributes to development of offspring hypertension.
Collapse
Affiliation(s)
- Ahmed K Abdel-Hakeem
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Archdeacon P, Detwiler RK. Bone morphogenetic protein 7 (BMP7): a critical role in kidney development and a putative modulator of kidney injury. Adv Chronic Kidney Dis 2008; 15:314-20. [PMID: 18565482 DOI: 10.1053/j.ackd.2008.04.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Bone morphogenetic protein 7 (BMP7), a member of the tumor growth factor beta superfamily, appears to have a role in both kidney development and response to kidney injury. Signals through its pathways permit both epithelial differentiation during embryogenesis and preservation of normal kidney architecture after stress, leading to the hypothesis that BMP7 may exert its kidney-protective effects in adult animals by preventing or reversing epithelial-to-mesenchymal transformation. This review attempts to synthesize the data supporting those conclusions and suggest some future areas of research.
Collapse
|
42
|
Ueda H, Miyazaki Y, Matsusaka T, Utsunomiya Y, Kawamura T, Hosoya T, Ichikawa I. Bmp in podocytes is essential for normal glomerular capillary formation. J Am Soc Nephrol 2008; 19:685-94. [PMID: 18272846 DOI: 10.1681/asn.2006090983] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Bone morphogenetic protein (BMP) 4 exerts multiple biological effects on kidney and ureter development. To examine the role of BMP4 in glomerular morphogenesis, we generated transgenic mice with altered BMP4 function in podocytes by conferring tissue-specificity with the nephrin (Nphs1) promoter. At birth, Tg(Nphs1-Nog) mice, which had loss of BMP4 function in podocytes, were found to have glomerular microaneurysms, collapsed glomerular capillary tufts, enlarged Bowman's capsules, and fewer normal proximal tubules. Conversely, Tg(Nphs1-Bmp4) mice, which had increased BMP4 function in podocytes, demonstrated defects in glomerular capillary formation, but podocytes were not appreciably affected. The Tg(Nphs1-Nog) and Tg(Nphs1-Bmp4) mice shared morphological characteristics with the previously reported podocyte-specific Vegf-A over-expressing and knockout mice, respectively. Consistent with the morphological similarity, in situ hybridization revealed an intense signal for podocyte expression of Vegf in Tg(Nphs1-Nog) mice, whereas the signal was markedly suppressed in Tg(Nphs1-Bmp4) mice. However, in vitro studies with metanephroi failed to demonstrate a direct interaction between BMP4 or Noggin and VEGF in podocytes. Instead, immunostaining showed that phosphorylated Smads, the mediators of BMP signaling, are present in endothelial and/or mesangial cells, but not in podocytes, within the developing glomeruli. Therefore, this study suggests that podocyte-derived BMP plays an important role in glomerular capillary formation, perhaps by acting on non-podocyte glomerular cells in a paracrine fashion.
Collapse
Affiliation(s)
- Hiroyuki Ueda
- Division of Kidney and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishishinbashi, Minato-ku, Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|
43
|
Nguyen TQ, Goldschmeding R. Bone Morphogenetic Protein-7 and Connective Tissue Growth Factor: Novel Targets for Treatment of Renal Fibrosis? Pharm Res 2008; 25:2416-26. [DOI: 10.1007/s11095-008-9548-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2007] [Accepted: 01/28/2008] [Indexed: 12/11/2022]
|
44
|
Hampton DW, Asher RA, Kondo T, Steeves JD, Ramer MS, Fawcett JW. A potential role for bone morphogenetic protein signalling in glial cell fate determination following adult central nervous system injury in vivo. Eur J Neurosci 2007; 26:3024-35. [PMID: 18028109 DOI: 10.1111/j.1460-9568.2007.05940.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Bone morphogenetic proteins (BMPs) and their endogenous inhibitors, including noggin, chordin and follistatin, have roles in pattern formation and fate specification of neuronal and glial cells during nervous system development. We have examined their influence on glial reactions in the injured central nervous system (CNS). We show that penetrating injuries to the brain and spinal cord resulted in the upregulation of BMP-2/4, BMP-7, and noggin, with the latter being expressed almost exclusively by reactive astrocytes at the injury site, and we show that astrocytes in vitro produce noggin. As BMPs have been shown to drive cultured NG2-positive oligodendrocyte precursors (OPCs) towards a multipotential phenotype (type II astrocytes), we investigated the effects of inhibiting noggin with a function-blocking antibody (noggin-FbAb). In vitro, BMP-driven conversion of OPCs to type 2 astrocytes was inhibited by noggin, an effect that was reversed by noggin-FbAb. Noggin-FbAb also increased the number of type 2 astrocytes generated from cultured OPCs exposed to an astrocyte feeder layer, consistent with astrocytes producing both BMPs and noggin. In knife cut injuries in vivo, noggin-FbAb treatment resulted in an increase in the number of NG2-positive cells and small GFAP-positive cells in the injury site, and the appearance of glial cells with the morphological and antigenic characteristics of type 2 astrocytes (as generated in vitro), with coexpression of both GFAP and NG2. This potential conversion of inhibitory OPCs to type 2 astrocyte-like cells in vivo suggests that endogenous BMPs, unmasked by noggin antagonism, might be exploited to manipulate cell fate following CNS trauma.
Collapse
Affiliation(s)
- David W Hampton
- ICORD, University of British Columbia, Vancouver, BC, Canada.
| | | | | | | | | | | |
Collapse
|
45
|
Adams D, Karolak M, Robertson E, Oxburgh L. Control of kidney, eye and limb expression of Bmp7 by an enhancer element highly conserved between species. Dev Biol 2007; 311:679-90. [PMID: 17936743 PMCID: PMC2394512 DOI: 10.1016/j.ydbio.2007.08.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2007] [Revised: 08/10/2007] [Accepted: 08/20/2007] [Indexed: 01/04/2023]
Abstract
Bmp7 is expressed in numerous tissues throughout development and is required for morphogenesis of the eye, hindlimb and kidney. In this study we show that the majority if not all of the cis-regulatory sequence governing expression at these anatomical sites during development is present in approximately 20 kb surrounding exon 1. In eye, limb and kidney, multiple distinct enhancer elements drive Bmp7 expression within each organ. In the eye, the elements driving expression in the pigmented epithelium and iris are spatially separated. In the kidney, Bmp7 expression in collecting ducts and nephron progenitors is driven by separate enhancer elements. Similarly, limb mesenchyme and apical ectodermal ridge expression are governed by separate elements. Although enhancers for pigmented epithelium, nephrogenic mesenchyme and apical ectodermal ridge are distributed across the approximately 20 kb region, an element of approximately 480 base pairs within intron 1 governs expression within the developing iris, collecting duct system of the kidney and limb mesenchyme. This element is remarkably conserved both in sequence and position in the Bmp7 locus between different vertebrates, ranging from Xenopus tropicalis to Homo sapiens, demonstrating that there is strong selective pressure for Bmp7 expression at these tissue sites. Furthermore, we show that the frog enhancer functions appropriately in transgenic mice. Interestingly, the intron 1 element cannot be found in the Bmp7 genes of vertebrates such as Danio rerio and Takifugu rubripes indicating that this modification of the Bmp7 gene might have arisen during the adaptation from aquatic to terrestrial life. Mutational analysis demonstrates that the enhancer activity of the intron 1 element is entirely dependent on the presence of a 10 base pair site within the intron 1 enhancer containing a predicted binding site for the FOXD3 transcription factor.
Collapse
Affiliation(s)
- Derek Adams
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, Maine 04074, USA
| | - Michele Karolak
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, Maine 04074, USA
| | - Elizabeth Robertson
- Wellcome Trust Center for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Leif Oxburgh
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, Maine 04074, USA
| |
Collapse
|
46
|
Huang Z, Nelson ER, Smith RL, Goodman SB. The sequential expression profiles of growth factors from osteoprogenitors [correction of osteroprogenitors] to osteoblasts in vitro. ACTA ACUST UNITED AC 2007; 13:2311-20. [PMID: 17523879 DOI: 10.1089/ten.2006.0423] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In this study, we delineate the sequential expression of selected growth factors associated with bone formation in vitro. Mineralization, osteocalcin, and alkaline phosphatase (ALP-2) were measured to monitor the differentiation and maturation of osteoprogenitor cells collected from C57BL mice. Bone-related growth factors, including transforming growth factor beta (TGF-beta), fibroblast growth factor 2 (FGF-2), platelet-derived growth factor (PDGF), insulinlike growth factor (IGF)-1, vascular endothelial growth factor (VEGF), bone morphogenetic protein (BMP)-2, and BMP-7, were selected. Enzyme-linked immunosorbent assay (ELISA) and reverse transcriptase polymerase chain reaction (RT-PCR) were used to measure growth factors at the protein and messenger ribonucleic acid (mRNA) level, respectively. The results found that ALP-2 expression increased progressively over time, whereas mineralization and osteocalcin did not become evident until culture day 14. VEGF and IGF-1 were upregulated early during proliferation. PDGF and TGF-beta mRNA expression was bimodal. FGF-2 and BMP-2 mRNAs were expressed only later in differentiation. FGF-2 mRNA signal levels were highest at day 14 and remained prominent through day 28 of culture. BMP-2 showed a similar profile as FGF-2. BMP-7 was not detectable using RT-PCR or ELISA. Strong correlations existed for the expression patterns between several early-response growth factors (VEGF, TGF-beta, and IGF-1) and were also evident for several late-response growth factors (BMP-2, PDGF, and FGF-2). Differential expression for grouped sets of growth factors occurs during the temporal acquisition of bone-specific markers as osteoprogenitor cell maturation proceeds in vitro.
Collapse
Affiliation(s)
- Zhinong Huang
- Department of Orthopedic Surgery, Stanford University Medical Center, Stanford, California 94305, USA
| | | | | | | |
Collapse
|
47
|
Abstract
Exogenous bone morphogenetic protein 4 (BMP4) inhibits ureteric branching morphogenesis and amplifies the already existing branching asymmetry in the developing mouse kidney in vitro. In the present study we examined ureteric branching morphogenesis in BMP4/lacZ heterozygous (BMP4(+/-)) mice in vitro under control conditions and in the presence of exogenous BMP4 using three-dimensional image analysis software. The relative expression of BMP4 mRNA was determined in BMP4(+/-) and wildtype urogenital ridges using real-time PCR. Embryonic day 12.5 (E12.5) BMP4(+/-) and wildtype mouse metanephroi were cultured for 48 h with or without 260 ng mL(-1) recombinant human BMP4 (rhBMP4) and were then wholemount immunostained in order to identify the ureteric epithelium, which was quantified in three dimensions. Despite a significant reduction in BMP4 mRNA in BMP4(+/-) mice, qualitative and quantitative studies identified no differences in ureteric branching morphogenesis between phenotypically normal BMP4(+/-) and wildtype metanephroi in either BMP4-treated or control cultures. Both BMP4(+/-) and wildtype metanephroi cultured in the presence of BMP4 showed a decrease in total ureteric length, branch number and ureteric volume, and increased average branch length compared with control cultures. A marked anterior-posterior asymmetry in both ureteric length, branch number and average branch length was observed in BMP4-treated metanephroi from both genotypes. A similar asymmetry was revealed in control metanephroi from both genotypes. This asymmetry is the result of reduced ureteric branching morphogenesis but not elongation in the posterior region of the kidney. These results suggest that despite reduced endogenous BMP4 mRNA levels, most BMP4(+/-) embryos can still facilitate normal ureteric branching morphogenesis during development. In addition, reduced endogenous levels of BMP4 do not alter the inhibitory effects of exogenous BMP4 on ureteric branching or amplification of normal renal asymmetry.
Collapse
Affiliation(s)
- Jason E Cain
- Department of Anatomy and Cell Biology, School of Biomedical Sciences, Monash University, Victoria, Australia
| | | |
Collapse
|
48
|
Michos O, Gonçalves A, Lopez-Rios J, Tiecke E, Naillat F, Beier K, Galli A, Vainio S, Zeller R. Reduction of BMP4 activity by gremlin 1 enables ureteric bud outgrowth and GDNF/WNT11 feedback signalling during kidney branching morphogenesis. Development 2007; 134:2397-405. [PMID: 17522159 DOI: 10.1242/dev.02861] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Antagonists act to restrict and negatively modulate the activity of secreted signals during progression of embryogenesis. In mouse embryos lacking the extra-cellular BMP antagonist gremlin 1 (Grem1), metanephric development is disrupted at the stage of initiating ureteric bud outgrowth. Treatment of mutant kidney rudiments in culture with recombinant gremlin 1 protein induces additional epithelial buds and restores outgrowth and branching. All epithelial buds express Wnt11, and Gdnf is significantly upregulated in the surrounding mesenchyme, indicating that epithelial-mesenchymal (e-m) feedback signalling is restored. In the wild type, Bmp4 is expressed by the mesenchyme enveloping the Wolffian duct and ureteric bud and Grem1 is upregulated in the mesenchyme around the nascent ureteric bud prior to initiation of its outgrowth. In agreement, BMP activity is reduced locally as revealed by lower levels of nuclear pSMAD protein in the mesenchyme. By contrast, in Grem1-deficient kidney rudiments, pSMAD proteins are detected in many cell nuclei in the metanephric mesenchyme, indicative of excessive BMP signal transduction. Indeed, genetic lowering of BMP4 levels in Grem1-deficient mouse embryos completely restores ureteric bud outgrowth and branching morphogenesis. The reduction of BMP4 levels in Grem1 mutant embryos enables normal progression of renal development and restores adult kidney morphology and functions. This study establishes that initiation of metanephric kidney development requires the reduction of BMP4 activity by the antagonist gremlin 1 in the mesenchyme, which in turn enables ureteric bud outgrowth and establishment of autoregulatory GDNF/WNT11 feedback signalling.
Collapse
Affiliation(s)
- Odyssé Michos
- Developmental Genetics, DKBW Centre for Biomedicine, University of Basel Medical Faculty, Mattenstrasse 28, CH-4058 Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Brenner-Anantharam A, Cebrian C, Guillaume R, Hurtado R, Sun TT, Herzlinger D. Tailbud-derived mesenchyme promotes urinary tract segmentation via BMP4 signaling. Development 2007; 134:1967-75. [PMID: 17442697 DOI: 10.1242/dev.004234] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Urinary tract morphogenesis requires the sub-division of the ureteric bud (UB) into the intra-renal collecting system and ureter, two tissues with unique structural and functional properties. In this report we investigate the cellular and molecular mechanisms that mediate their differentiation. Fate mapping experiments in the developing chick indicate that the UB is surrounded by two distinct mesenchymal populations: nephrogenic mesenchyme derived from the intermediate mesoderm and tailbud-derived mesoderm, which is selectively associated with the domain of the UB that differentiates into the ureter. Functional experiments utilizing murine metanephric kidney explants show that BMP4, a paracrine factor secreted by tailbud-derived mesenchyme, is required for ureter morphogenesis. Conversely, ectopic BMP4 signaling is sufficient to induce ureter morphogenesis in domains of the UB normally fated to differentiate into the intra-renal collecting system. Collectively, these results indicate that the border between the kidney and ureter forms where mesenchymal tissues originating in two different areas of the early embryo meet. These data raise the possibility that the susceptibility of this junction to congenital defects in humans, such as ureteral-pelvic obstructions, may be related to the complex morphogenetic movements that are required to integrate cells from these different lineages into a single functional structure.
Collapse
Affiliation(s)
- Andrea Brenner-Anantharam
- Department of Physiology and Biophysics, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
50
|
Blauwkamp MN, Beyer LA, Kabara L, Takemura K, Buck T, King W, Dolan DF, Barald KF, Raphael Y, Koenig RJ. The role of bone morphogenetic protein 4 in inner ear development and function. Hear Res 2006; 225:71-9. [PMID: 17275231 PMCID: PMC1868473 DOI: 10.1016/j.heares.2006.12.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2006] [Revised: 12/06/2006] [Accepted: 12/07/2006] [Indexed: 11/26/2022]
Abstract
Bone Morphogenetic Protein 4 (BMP4) is a member of the TGF-beta superfamily and is known to be important for the normal development of many tissues and organs, including the inner ear. Bmp4 homozygous null mice die as embryos, but Bmp4 heterozygous null (Bmp4(+/-)) mice are viable and some adults exhibit a circling phenotype, suggestive of an inner ear defect. To understand the role of BMP4 in inner ear development and function, we have begun to study C57BL/6 Bmp4(+/-) mice. Quantitative testing of the vestibulo-collic reflex, which helps maintain head stability, demonstrated that Bmp4(+/-) mice that exhibit circling behavior have a poor response in the yaw axis, consistent with semicircular canal dysfunction. Although the hair cells of the ampullae were grossly normal, the stereocilia were greatly reduced in number. Auditory brainstem responses showed that Bmp4(+/-) mice have elevated hearing thresholds and immunohistochemical staining demonstrated decreased numbers of neuronal processes in the organ of Corti. Thus Bmp4(+/-) mice have structural and functional deficits in the inner ear.
Collapse
Affiliation(s)
- Marsha N. Blauwkamp
- Cellular and Molecular Biology Program 2966 Taubman Medical Library, University of Michigan, Ann Arbor MI 48109-0619, USA
| | - Lisa A. Beyer
- Kresge Hearing Research Institute, Department of Otolaryngology 1301 East Ann Street, Ann Arbor, MI 48109-0506, USA
| | - Lisa Kabara
- Kresge Hearing Research Institute, Department of Otolaryngology 1301 East Ann Street, Ann Arbor, MI 48109-0506, USA
| | - Keiji Takemura
- Kresge Hearing Research Institute, Department of Otolaryngology 1301 East Ann Street, Ann Arbor, MI 48109-0506, USA
| | - Timothy Buck
- Kresge Hearing Research Institute, Department of Otolaryngology 1301 East Ann Street, Ann Arbor, MI 48109-0506, USA
| | - W.M. King
- Kresge Hearing Research Institute, Department of Otolaryngology 1301 East Ann Street, Ann Arbor, MI 48109-0506, USA
| | - David F. Dolan
- Kresge Hearing Research Institute, Department of Otolaryngology 1301 East Ann Street, Ann Arbor, MI 48109-0506, USA
| | - Kate F. Barald
- Cellular and Molecular Biology Program 2966 Taubman Medical Library, University of Michigan, Ann Arbor MI 48109-0619, USA
- Departments of Cell and Developmental Biology, and Biomedical Engineering 3053 BSRB, 109 Zina Pitcher Place, University of Michigan, Ann Arbor, MI, 48109-2200, USA
| | - Yehoash Raphael
- Kresge Hearing Research Institute, Department of Otolaryngology 1301 East Ann Street, Ann Arbor, MI 48109-0506, USA
| | - Ronald J. Koenig
- Cellular and Molecular Biology Program 2966 Taubman Medical Library, University of Michigan, Ann Arbor MI 48109-0619, USA
- Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes 5560 MSRB-II, 1150 W. Medical Center Drive, University of Michigan, Ann Arbor MI, 48109-0678, USA
| |
Collapse
|