2301
|
Rocha SF, Schiller M, Jing D, Li H, Butz S, Vestweber D, Biljes D, Drexler HC, Nieminen-Kelhä M, Vajkoczy P, Adams S, Benedito R, Adams RH. Esm1 Modulates Endothelial Tip Cell Behavior and Vascular Permeability by Enhancing VEGF Bioavailability. Circ Res 2014; 115:581-90. [DOI: 10.1161/circresaha.115.304718] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Susana F. Rocha
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Maria Schiller
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Ding Jing
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Hang Li
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Stefan Butz
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Dietmar Vestweber
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Daniel Biljes
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Hannes C.A. Drexler
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Melina Nieminen-Kelhä
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Peter Vajkoczy
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Susanne Adams
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Rui Benedito
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| | - Ralf H. Adams
- From the Max Planck Institute for Molecular Biomedicine, Münster, Germany (S.F.R., M.S., D.J., H.L., S.B., D.V., D.B., H.C.A.D., S.A., R.B., R.H.A.); University of Münster, Münster, Germany (R.H.A.); Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (S.F.R., R.B.); and Neurochirurgische Klinik, Charite Universitätsmedizin, Berlin, Germany (M.N.-K., P.V.)
| |
Collapse
|
2302
|
Pickup MW, Hover LD, Polikowsky ER, Chytil A, Gorska AE, Novitskiy SV, Moses HL, Owens P. BMPR2 loss in fibroblasts promotes mammary carcinoma metastasis via increased inflammation. Mol Oncol 2014; 9:179-91. [PMID: 25205038 DOI: 10.1016/j.molonc.2014.08.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 08/15/2014] [Indexed: 01/22/2023] Open
Abstract
Bone Morphogenetic Protein (BMP) receptors mediate a diverse range of signals to regulate both development and disease. BMP activity has been linked to both tumor promoting and suppressive functions in both tumor cells and their surrounding microenvironment. We sought to investigate the requirement for BMPR2 in stromal fibroblasts during mammary tumor formation and metastasis. We utilized FSP1 (Fibroblast Specific Protein-1) promoter driven Cre to genetically delete BMPR2 in mice expressing the MMTV.PyVmT mammary carcinoma oncogene. We found that abrogation of stromal BMPR2 expression via FSP1 driven Cre resulted in increased tumor metastasis. Additionally, similar to epithelial BMPR2 abrogation, stromal loss of BMPR2 results in increased inflammatory cell infiltration. We proceeded to isolate and establish fibroblast cell lines without BMPR2 and found a cell autonomous increase in inflammatory cytokine secretion. Fibroblasts were co-implanted with syngeneic tumor cells and resulted in accelerated tumor growth and increased metastasis when fibroblasts lacked BMPR2. We observed that the loss of BMPR2 results in increased chemokine expression, which facilitates inflammation by a sustained increase in myeloid cells. The chemokines increased in BMPR2 deleted cells correlated with poor outcome in human breast cancer patients. We conclude that BMPR2 has tumor suppressive functions in the stroma by regulating inflammation.
Collapse
Affiliation(s)
| | - Laura D Hover
- Department of Pathology, Microbiology and Immunology, USA
| | | | | | | | | | | | | |
Collapse
|
2303
|
A gene regulatory network controls the binary fate decision of rod and bipolar cells in the vertebrate retina. Dev Cell 2014; 30:513-27. [PMID: 25155555 PMCID: PMC4304698 DOI: 10.1016/j.devcel.2014.07.018] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/16/2014] [Accepted: 07/21/2014] [Indexed: 12/12/2022]
Abstract
Gene regulatory networks (GRNs) regulate critical events during development. In complex tissues, such as the mammalian central nervous system (CNS), networks likely provide the complex regulatory interactions needed to direct the specification of the many CNS cell types. Here, we dissect a GRN that regulates a binary fate decision between two siblings in the murine retina, the rod photoreceptor and bipolar interneuron. The GRN centers on Blimp1, one of the transcription factors (TFs) that regulates the rod versus bipolar cell fate decision. We identified a cis-regulatory module (CRM), B108, that mimics Blimp1 expression. Deletion of genomic B108 by CRISPR/Cas9 in vivo using electroporation abolished the function of Blimp1. Otx2 and RORβ were found to regulate Blimp1 expression via B108, and Blimp1 and Otx2 were shown to form a negative feedback loop that regulates the level of Otx2, which regulates the production of the correct ratio of rods and bipolar cells.
Collapse
|
2304
|
Carney CM, Muszynski JL, Strotman LN, Lewis SR, O'Connell RL, Beebe DJ, Theberge AB, Jorgensen JS. Cellular microenvironment dictates androgen production by murine fetal Leydig cells in primary culture. Biol Reprod 2014; 91:85. [PMID: 25143354 DOI: 10.1095/biolreprod.114.118570] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Despite the fact that fetal Leydig cells are recognized as the primary source of androgens in male embryos, the mechanisms by which steroidogenesis occurs within the developing testis remain unclear. A genetic approach was used to visualize and isolate fetal Leydig cells from remaining cells within developing mouse testes. Cyp11a1-Cre mice were bred to mT/mG dual reporter mice to target membrane-tagged enhanced green fluorescent protein (GFP) within steroidogenic cells, whereas other cells expressed membrane-tagged tandem-dimer tomato red. Fetal Leydig cell identity was validated using double-labeled immunohistochemistry against GFP and the steroidogenic enzyme 3beta-HSD, and cells were successfully isolated as indicated by qPCR results from sorted cell populations. Because fetal Leydig cells must collaborate with neighboring cells to synthesize testosterone, we hypothesized that the fetal Leydig cell microenvironment defined their capacity for androgen production. Microfluidic culture devices were used to measure androstenedione and testosterone production of fetal Leydig cells that were cultured in cell-cell contact within a mixed population, were isolated but remained in medium contact via compartmentalized co-culture with other testicular cells, or were isolated and cultured alone. Results showed that fetal Leydig cells maintained their identity and steroidogenic activity for 3-5 days in primary culture. Microenvironment dictated proficiency of testosterone production. As expected, fetal Leydig cells produced androstenedione but not testosterone when cultured in isolation. More testosterone accumulated in medium from mixed cultures than from compartmentalized co-cultures initially; however, co-cultures maintained testosterone synthesis for a longer time. These data suggest that a combination of cell-cell contact and soluble factors constitute the ideal microenvironment for fetal Leydig cell activity in primary culture.
Collapse
Affiliation(s)
- Colleen M Carney
- Department of Comparative Bioscience, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jessica L Muszynski
- Department of Comparative Bioscience, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Lindsay N Strotman
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Samantha R Lewis
- Department of Comparative Bioscience, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Rachel L O'Connell
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ashleigh B Theberge
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin Department of Urology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Joan S Jorgensen
- Department of Comparative Bioscience, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin Molecular and Environmental Toxicology Center, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
2305
|
Endothelial cell FGF signaling is required for injury response but not for vascular homeostasis. Proc Natl Acad Sci U S A 2014; 111:13379-84. [PMID: 25139991 DOI: 10.1073/pnas.1324235111] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Endothelial cells (ECs) express fibroblast growth factor receptors (FGFRs) and are exquisitely sensitive to FGF signals. However, whether the EC or another vascular cell type requires FGF signaling during development, homeostasis, and response to injury is not known. Here, we show that Flk1-Cre or Tie2-Cre mediated deletion of FGFR1 and FGFR2 (Fgfr1/2(Flk1-Cre) or Fgfr1/2(Tie2-Cre) mice), which results in deletion in endothelial and hematopoietic cells, is compatible with normal embryonic development. As adults, Fgfr1/2(Flk1-Cre) mice maintain normal blood pressure and vascular reactivity and integrity under homeostatic conditions. However, neovascularization after skin or eye injury was significantly impaired in both Fgfr1/2(Flk1-Cre) and Fgfr1/2(Tie2-Cre) mice, independent of either hematopoietic cell loss of FGFR1/2 or vascular endothelial growth factor receptor 2 (Vegfr2) haploinsufficiency. Also, impaired neovascularization was associated with delayed cutaneous wound healing. These findings reveal a key requirement for cell-autonomous EC FGFR signaling in injury-induced angiogenesis, but not for vascular homeostasis, identifying the EC FGFR signaling pathway as a target for diseases associated with aberrant vascular proliferation, such as age-related macular degeneration, and for modulating wound healing without the potential toxicity associated with direct manipulation of systemic FGF or VEGF activity.
Collapse
|
2306
|
Lgr5-positive cells are cancer stem cells in skin squamous cell carcinoma. Tumour Biol 2014; 35:11605-12. [DOI: 10.1007/s13277-014-2488-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Accepted: 08/08/2014] [Indexed: 10/24/2022] Open
|
2307
|
Bell EL, Nagamori I, Williams EO, Del Rosario AM, Bryson BD, Watson N, White FM, Sassone-Corsi P, Guarente L. SirT1 is required in the male germ cell for differentiation and fecundity in mice. Development 2014; 141:3495-504. [PMID: 25142464 DOI: 10.1242/dev.110627] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Sirtuins are NAD(+)-dependent deacylases that regulate numerous biological processes in response to the environment. SirT1 is the mammalian ortholog of yeast Sir2, and is involved in many metabolic pathways in somatic tissues. Whole body deletion of SirT1 alters reproductive function in oocytes and the testes, in part caused by defects in central neuro-endocrine control. To study the function of SirT1 specifically in the male germ line, we deleted this sirtuin in male germ cells and found that mutant mice had smaller testes, a delay in differentiation of pre-meiotic germ cells, decreased spermatozoa number, an increased proportion of abnormal spermatozoa and reduced fertility. At the molecular level, mutants do not have the characteristic increase in acetylation of histone H4 at residues K5, K8 and K12 during spermiogenesis and demonstrate corresponding defects in the histone to protamine transition. Our findings thus reveal a germ cell-autonomous role of SirT1 in spermatogenesis.
Collapse
Affiliation(s)
- Eric L Bell
- Massachusetts Institute of Technology, Department of Biology, Glenn Laboratory for the Science of Aging, Cambridge, MA 02139, USA
| | - Ippei Nagamori
- Center for Epigenetics and Metabolism, Department of Biological Chemistry, University of California, Irvine, CA 92697, USA Osaka University, Graduate School of Medicine, Osaka 565-0871, Japan
| | - Eric O Williams
- Massachusetts Institute of Technology, Department of Biology, Glenn Laboratory for the Science of Aging, Cambridge, MA 02139, USA
| | - Amanda M Del Rosario
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Bryan D Bryson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA Massachusettes Institute of Technology, Department of Biological Engineering, Cambridge, MA 02139, USA
| | - Nicki Watson
- W. M. Keck Microscopy Facility Whitehead Institute, Cambridge, MA 02139, USA
| | - Forest M White
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA Massachusettes Institute of Technology, Department of Biological Engineering, Cambridge, MA 02139, USA
| | - Paolo Sassone-Corsi
- Center for Epigenetics and Metabolism, Department of Biological Chemistry, University of California, Irvine, CA 92697, USA
| | - Leonard Guarente
- Massachusetts Institute of Technology, Department of Biology, Glenn Laboratory for the Science of Aging, Cambridge, MA 02139, USA Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
2308
|
Tong PL, Roediger B, Kolesnikoff N, Biro M, Tay SS, Jain R, Shaw LE, Grimbaldeston MA, Weninger W. The skin immune atlas: three-dimensional analysis of cutaneous leukocyte subsets by multiphoton microscopy. J Invest Dermatol 2014; 135:84-93. [PMID: 25007044 PMCID: PMC4268113 DOI: 10.1038/jid.2014.289] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 04/10/2014] [Accepted: 04/29/2014] [Indexed: 01/12/2023]
Abstract
Site-specific differences in skin response to pathogens and in the course of cutaneous inflammatory diseases are well appreciated. The composition and localization of cutaneous leukocytes has been studied extensively using histology and flow cytometry. However, the precise three-dimensional (3D) distribution of distinct immune cell subsets within skin at different body sites requires visualization of intact living skin. We used intravital multiphoton microscopy in transgenic reporter mice in combination with quantitative flow cytometry to generate a 3D immune cell atlas of mouse skin. The 3D location of innate and adaptive immune cells and site-specific differences in the densities of macrophages, T cells and mast cells at four defined sites (ear, back, footpad, tail) is presented. The combinatorial approach further demonstrates an as yet unreported age-dependent expansion of dermal gamma-delta T cells. Localization of dermal immune cells relative to anatomical structures was also determined. While dendritic cells were dispersed homogeneously within the dermis, mast cells preferentially localized to the perivascular space. Finally, we show the functional relevance of site-specific mast cell disparities using the passive cutaneous anaphylaxis model. These approaches are applicable to assessing immune cell variations and potential functional consequences in the setting of infection as well as the pathogenesis of inflammatory skin conditions.
Collapse
Affiliation(s)
- Philip L Tong
- Centenary Institute, Newtown, New South Wales, Australia; Discipline of Dermatology, The University of Sydney, Camperdown, New South Wales, Australia; Department of Dermatology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.
| | - Ben Roediger
- Centenary Institute, Newtown, New South Wales, Australia; Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Natasha Kolesnikoff
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia, Australia
| | - Maté Biro
- Centenary Institute, Newtown, New South Wales, Australia; Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Szun S Tay
- Centenary Institute, Newtown, New South Wales, Australia
| | - Rohit Jain
- Centenary Institute, Newtown, New South Wales, Australia; Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Lisa E Shaw
- Centenary Institute, Newtown, New South Wales, Australia
| | | | - Wolfgang Weninger
- Centenary Institute, Newtown, New South Wales, Australia; Discipline of Dermatology, The University of Sydney, Camperdown, New South Wales, Australia; Department of Dermatology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.
| |
Collapse
|
2309
|
Liang M, Liang A, Wang Y, Jiang J, Cheng J. Smooth muscle cells from the anastomosed artery are the major precursors for neointima formation in both artery and vein grafts. Basic Res Cardiol 2014; 109:431. [PMID: 25107324 DOI: 10.1007/s00395-014-0431-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 07/28/2014] [Accepted: 07/30/2014] [Indexed: 12/01/2022]
Abstract
Accumulation of smooth muscle cells (SMC) results in neointima formation in injured vessels. Two graft models consisting of vein and artery grafts were created by anastomosing common carotid arteries to donor vessels. To identify the origin of the neointima cells from anastomosed arteries, we use Wnt1-Cre/reporter mice to label and track SMCs in the common carotid artery. The contribution of SMCs in the neighboring arteries to neointima formation was studied. On evaluating the artery grafts after 1 month, >90 % of the labeled neointima cells were found to have originated from the anastomosing host arteries. Most of the neointima cells were also smooth muscle α-actin positive (SMA-α(+)) and expressed the smooth muscle myosin heavy chain (SMMHC), the SMC terminal differentiation marker. In vein grafts, about 60 % SMA-α-positive cells were from anastomosing arteries. Bone marrow cells did not contribute to neointima SMCs in vein grafts, but did co-stain with markers of inflammatory cells. Wnt1 expression was not detected in the neointima cells in the vein or artery grafts, or the injured femoral arteries. Neointima SMCs showed the synthetic phenotype and were positively labeled with BrdU in vitro and in vivo. Treatment with the IGF-1 receptor inhibitor suppressed SMC proliferation and neointima formation in vein grafts. Our results indicate that SMCs from the neighboring artery are predominantly present in the neointima formed in both vein and artery grafts and that Wnt1-Cre mice can be used to explore the role of SMCs originating from neighboring vessels in vascular remodeling.
Collapse
Affiliation(s)
- Ming Liang
- Department of Nephrology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | | | | | | | | |
Collapse
|
2310
|
Kim SC, Kellett T, Wang S, Nishi M, Nagre N, Zhou B, Flodby P, Shilo K, Ghadiali SN, Takeshima H, Hubmayr RD, Zhao X. TRIM72 is required for effective repair of alveolar epithelial cell wounding. Am J Physiol Lung Cell Mol Physiol 2014; 307:L449-59. [PMID: 25106429 DOI: 10.1152/ajplung.00172.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The molecular mechanisms for lung cell repair are largely unknown. Previous studies identified tripartite motif protein 72 (TRIM72) from striated muscle and linked its function to tissue repair. In this study, we characterized TRIM72 expression in lung tissues and investigated the role of TRIM72 in repair of alveolar epithelial cells. In vivo injury of lung cells was introduced by high tidal volume ventilation, and repair-defective cells were labeled with postinjury administration of propidium iodide. Primary alveolar epithelial cells were isolated and membrane wounding and repair were labeled separately. Our results show that absence of TRIM72 increases susceptibility to deformation-induced lung injury whereas TRIM72 overexpression is protective. In vitro cell wounding assay revealed that TRIM72 protects alveolar epithelial cells through promoting repair rather than increasing resistance to injury. The repair function of TRIM72 in lung cells is further linked to caveolin 1. These data suggest an essential role for TRIM72 in repair of alveolar epithelial cells under plasma membrane stress failure.
Collapse
Affiliation(s)
- Seong Chul Kim
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Thomas Kellett
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Shaohua Wang
- Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Miyuki Nishi
- Department of Biological Chemistry, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto, Japan
| | - Nagaraja Nagre
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Beiyun Zhou
- Will Rogers Institute Pulmonary Research Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of Southern California, Los Angeles, California
| | - Per Flodby
- Will Rogers Institute Pulmonary Research Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of Southern California, Los Angeles, California
| | - Konstantin Shilo
- Thoracic Pathology Division, Department of Pathology, The Ohio State University, Columbus, Ohio
| | - Samir N Ghadiali
- Biomedical Engineering Department, College of Engineering, The Ohio State University, Columbus, Ohio; and
| | - Hiroshi Takeshima
- Department of Biological Chemistry, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto, Japan
| | - Rolf D Hubmayr
- Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Xiaoli Zhao
- Division of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio; Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
| |
Collapse
|
2311
|
Choi YS, Zhang Y, Xu M, Yang Y, Ito M, Peng T, Cui Z, Nagy A, Hadjantonakis AK, Lang RA, Cotsarelis G, Andl T, Morrisey EE, Millar SE. Distinct functions for Wnt/β-catenin in hair follicle stem cell proliferation and survival and interfollicular epidermal homeostasis. Cell Stem Cell 2014; 13:720-33. [PMID: 24315444 DOI: 10.1016/j.stem.2013.10.003] [Citation(s) in RCA: 253] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Revised: 08/23/2013] [Accepted: 10/04/2013] [Indexed: 12/18/2022]
Abstract
Wnt/β-catenin signaling is a central regulator of adult stem cells. Variable sensitivity of Wnt reporter transgenes, β-catenin's dual roles in adhesion and signaling, and hair follicle degradation and inflammation resulting from broad deletion of epithelial β-catenin have precluded clear understanding of Wnt/β-catenin's functions in adult skin stem cells. By inducibly deleting β-catenin globally in skin epithelia, only in hair follicle stem cells, or only in interfollicular epidermis and comparing the phenotypes with those caused by ectopic expression of the Wnt/β-catenin inhibitor Dkk1, we show that this pathway is necessary for hair follicle stem cell proliferation. However, β-catenin is not required within hair follicle stem cells for their maintenance, and follicles resume proliferating after ectopic Dkk1 has been removed, indicating persistence of functional progenitors. We further unexpectedly discovered a broader role for Wnt/β-catenin signaling in contributing to progenitor cell proliferation in nonhairy epithelia and interfollicular epidermis under homeostatic, but not inflammatory, conditions.
Collapse
Affiliation(s)
- Yeon Sook Choi
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2312
|
Fausther M, Dranoff JA. Integrins, myofibroblasts, and organ fibrosis. Hepatology 2014; 60:756-8. [PMID: 24700390 PMCID: PMC4110176 DOI: 10.1002/hep.27155] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 03/27/2014] [Accepted: 03/30/2014] [Indexed: 12/14/2022]
Abstract
Myofibroblasts are the major source of extracellular matrix components that accumulate during tissue fibrosis, and hepatic stellate cells (HSCs) are believed to be the major source of myofibroblasts in the liver. To date, robust systems to genetically manipulate these cells have not been developed. We report that Cre under control of the promoter of Pdgfrb (Pdgfrb-Cre) inactivates loxP-flanked genes in mouse HSCs with high efficiency. We used this system to delete the gene encoding αV integrin subunit because various αV-containing integrins have been suggested as central mediators of fibrosis in multiple organs. Such depletion protected mice from carbon tetrachloride–induced hepatic fibrosis, whereas global loss of β3, β5 or β6 integrin or conditional loss of β8 integrins in HSCs did not. We also found that Pdgfrb-Cre effectively targeted myofibroblasts in multiple organs, and depletion of the αV integrin subunit using this system was protective in other models of organ fibrosis, including pulmonary and renal fibrosis. Pharmacological blockade of αV-containing integrins by a small molecule (CWHM 12) attenuated both liver and lung fibrosis, including in a therapeutic manner. These data identify a core pathway that regulates fibrosis and suggest that pharmacological targeting of all αV integrins may have clinical utility in the treatment of patients with a broad range of fibrotic diseases.
Collapse
|
2313
|
Dorfman MD, Garcia-Rudaz C, Alderman Z, Kerr B, Lomniczi A, Dissen GA, Castellano JM, Garcia-Galiano D, Gaytan F, Xu B, Tena-Sempere M, Ojeda SR. Loss of Ntrk2/Kiss1r signaling in oocytes causes premature ovarian failure. Endocrinology 2014; 155:3098-111. [PMID: 24877631 PMCID: PMC4097998 DOI: 10.1210/en.2014-1111] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Neurotrophins (NTs), once believed to be neural-specific trophic factors, are now known to also provide developmental cues to non-neural cells. In the ovary, NTs contribute to both the formation and development of follicles. Here we show that oocyte-specific deletion of the Ntrk2 gene that encodes the NTRK2 receptor (NTRK2) for neurotrophin-4/5 and brain-derived neurotrophic factor (BDNF) results in post-pubertal oocyte death, loss of follicular organization, and early adulthood infertility. Oocytes lacking NTRK2 do not respond to gonadotropins with activation of phosphatidylinositol 3-kinase (PI3K)-AKT-mediated signaling. Before puberty, oocytes only express a truncated NTRK2 form (NTRK2.T1), but at puberty full-length (NTRK2.FL) receptors are rapidly induced by the preovulatory gonadotropin surge. A cell line expressing both NTRK2.T1 and the kisspeptin receptor (KISS1R) responds to BDNF stimulation with activation of Ntrk2 expression only if kisspeptin is present. This suggests that BDNF and kisspeptin that are produced by granulosa cells (GCs) of periovulatory follicles act in concert to mediate the effect of gonadotropins on Ntrk2 expression in oocytes. In keeping with this finding, the oocytes of NTRK2-intact mice fail to respond to gonadotropins with increased Ntrk2 expression in the absence of KISS1R. Our results demonstrate that the preovulatory gonadotropin surge promotes oocyte survival at the onset of reproductive cyclicity by inducing oocyte expression of NTRK2.FL receptors that set in motion an AKT-mediated survival pathway. They also suggest that gonadotropins activate NTRK2.FL expression via a dual communication pathway involving BDNF and kisspeptin produced in GCs and their respective receptors NTRK2.T1 and KISS1R expressed in oocytes.
Collapse
Affiliation(s)
- Mauricio D Dorfman
- Division of Neuroscience (M.D.D., C.G.-R., Z.A., B.K., A.L., G.A.D., J.M.C., S.R.O.), Oregon National Primate Research Center/Oregon Health and Science University, Beaverton, Oregon 97006; Department of Cell Biology, Physiology and Immunology (D.G.-G., F.G., M.T.-S.), University of Córdoba; Centro de Investigacion Biomedica en Red (CIBER) Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III; and Instituto Maimónides de Investigaciones Biomédicas/Hospital Universitario Reina Sofia, Cordoba, Spain 14004; and Department of Pharmacology and Physiology (B.X.), Georgetown University Medical Center, Washington DC 20057
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2314
|
Gérard A, Patino-Lopez G, Beemiller P, Nambiar R, Ben-Aissa K, Liu Y, Totah FJ, Tyska MJ, Shaw S, Krummel MF. Detection of rare antigen-presenting cells through T cell-intrinsic meandering motility, mediated by Myo1g. Cell 2014; 158:492-505. [PMID: 25083865 PMCID: PMC4119593 DOI: 10.1016/j.cell.2014.05.044] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 04/15/2014] [Accepted: 05/20/2014] [Indexed: 10/25/2022]
Abstract
To mount an immune response, T lymphocytes must successfully search for foreign material bound to the surface of antigen-presenting cells. How T cells optimize their chances of encountering and responding to these antigens is unknown. T cell motility in tissues resembles a random or Levy walk and is regulated in part by external factors including chemokines and lymph-node topology, but motility parameters such as speed and propensity to turn may also be cell intrinsic. Here we found that the unconventional myosin 1g (Myo1g) motor generates membrane tension, enforces cell-intrinsic meandering search, and enhances T-DC interactions during lymph-node surveillance. Increased turning and meandering motility, as opposed to ballistic motility, is enhanced by Myo1g. Myo1g acts as a "turning motor" and generates a form of cellular "flânerie." Modeling and antigen challenges show that these intrinsically programmed elements of motility search are critical for the detection of rare cognate antigen-presenting cells.
Collapse
Affiliation(s)
- Audrey Gérard
- Department of Pathology, University of California, San Francisco, 513 Parnassus Ave, HSW512, San Francisco, CA 94143-0511, USA
| | - Genaro Patino-Lopez
- Experimental Immunology Branch National Cancer Institute, Bethesda, MD 20892-1360, USA
| | - Peter Beemiller
- Department of Pathology, University of California, San Francisco, 513 Parnassus Ave, HSW512, San Francisco, CA 94143-0511, USA
| | - Rajalakshmi Nambiar
- Cell and Developmental Biology Department, Vanderbilt University School of Medicine, Nashville, TN 37205, USA
| | - Khadija Ben-Aissa
- Experimental Immunology Branch National Cancer Institute, Bethesda, MD 20892-1360, USA
| | - Yin Liu
- Experimental Immunology Branch National Cancer Institute, Bethesda, MD 20892-1360, USA
| | - Fadi J. Totah
- Department of Pathology, University of California, San Francisco, 513 Parnassus Ave, HSW512, San Francisco, CA 94143-0511, USA
| | - Matthew J. Tyska
- Cell and Developmental Biology Department, Vanderbilt University School of Medicine, Nashville, TN 37205, USA
| | - Stephen Shaw
- Experimental Immunology Branch National Cancer Institute, Bethesda, MD 20892-1360, USA
| | - Matthew F. Krummel
- Department of Pathology, University of California, San Francisco, 513 Parnassus Ave, HSW512, San Francisco, CA 94143-0511, USA
| |
Collapse
|
2315
|
Murphy MM, Keefe AC, Lawson JA, Flygare SD, Yandell M, Kardon G. Transiently active Wnt/β-catenin signaling is not required but must be silenced for stem cell function during muscle regeneration. Stem Cell Reports 2014; 3:475-88. [PMID: 25241745 PMCID: PMC4266007 DOI: 10.1016/j.stemcr.2014.06.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 06/27/2014] [Accepted: 06/30/2014] [Indexed: 12/22/2022] Open
Abstract
Adult muscle’s exceptional capacity for regeneration is mediated by muscle stem cells, termed satellite cells. As with many stem cells, Wnt/β-catenin signaling has been proposed to be critical in satellite cells during regeneration. Using new genetic reagents, we explicitly test in vivo whether Wnt/β-catenin signaling is necessary and sufficient within satellite cells and their derivatives for regeneration. We find that signaling is transiently active in transit-amplifying myoblasts, but is not required for regeneration or satellite cell self-renewal. Instead, downregulation of transiently activated β-catenin is important to limit the regenerative response, as continuous regeneration is deleterious. Wnt/β-catenin activation in adult satellite cells may simply be a vestige of their developmental lineage, in which β-catenin signaling is critical for fetal myogenesis. In the adult, surprisingly, we show that it is not activation but rather silencing of Wnt/β-catenin signaling that is important for muscle regeneration. Wnt/β-catenin signaling is transiently active in myoblasts during muscle regeneration β-catenin is not required in myogenic cells for muscle regeneration β-catenin signaling in myoblasts must be silenced to limit the regenerative response β-catenin requirement and sensitivity differs in fetal and adult muscle stem cells
Collapse
Affiliation(s)
- Malea M Murphy
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexandra C Keefe
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Jennifer A Lawson
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Steven D Flygare
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Mark Yandell
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Gabrielle Kardon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
2316
|
Piran R, Lee SH, Li CR, Charbono A, Bradley LM, Levine F. Pharmacological induction of pancreatic islet cell transdifferentiation: relevance to type I diabetes. Cell Death Dis 2014; 5:e1357. [PMID: 25077543 PMCID: PMC4123101 DOI: 10.1038/cddis.2014.311] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 05/19/2014] [Accepted: 06/16/2014] [Indexed: 12/22/2022]
Abstract
Type I diabetes (T1D) is an autoimmune disease in which an immune response to pancreatic β-cells results in their loss over time. Although the conventional view is that this loss is due to autoimmune destruction, we present evidence of an additional phenomenon in which autoimmunity promotes islet endocrine cell transdifferentiation. The end result is a large excess of δ-cells, resulting from α- to β- to δ-cell transdifferentiation. Intermediates in the process of transdifferentiation were present in murine and human T1D. Here, we report that the peptide caerulein was sufficient in the context of severe β-cell deficiency to induce efficient induction of α- to β- to δ-cell transdifferentiation in a manner very similar to what occurred in T1D. This was demonstrated by genetic lineage tracing and time course analysis. Islet transdifferentiation proceeded in an islet autonomous manner, indicating the existence of a sensing mechanism that controls the transdifferentiation process within each islet. The finding of evidence for islet cell transdifferentiation in rodent and human T1D and its induction by a single peptide in a model of T1D has important implications for the development of β-cell regeneration therapies for diabetes.
Collapse
Affiliation(s)
- R Piran
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - S-H Lee
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - C-R Li
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - A Charbono
- Animal Facility, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - L M Bradley
- Infectious and Inflammatory Disease Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - F Levine
- Sanford Children's Health Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
2317
|
Feil S, Fehrenbacher B, Lukowski R, Essmann F, Schulze-Osthoff K, Schaller M, Feil R. Transdifferentiation of vascular smooth muscle cells to macrophage-like cells during atherogenesis. Circ Res 2014; 115:662-7. [PMID: 25070003 DOI: 10.1161/circresaha.115.304634] [Citation(s) in RCA: 414] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Atherosclerosis is a widespread and devastating disease, but the origins of cells within atherosclerotic plaques are not well defined. OBJECTIVE To investigate the specific contribution of vascular smooth muscle cells (SMCs) to atherosclerotic plaque formation by genetic inducible fate mapping in mice. METHODS AND RESULTS Vascular SMCs were genetically pulse-labeled using the tamoxifen-dependent Cre recombinase, CreER(T2), expressed from the endogenous SM22α locus combined with Cre-activatable reporter genes that were integrated into the ROSA26 locus. Mature SMCs in the arterial media were labeled by tamoxifen treatment of young apolipoprotein E-deficient mice before the development of atherosclerosis and then their fate was monitored in older atherosclerotic animals. We found that medial SMCs can undergo clonal expansion and convert to macrophage-like cells that have lost classic SMC marker expression and make up a major component of advanced atherosclerotic lesions. CONCLUSIONS This study provides strong in vivo evidence for smooth muscle-to-macrophage transdifferentiation and supports an important role of SMC plasticity in atherogenesis. Targeting this type of SMC phenotypic conversion might be a novel strategy for the treatment of atherosclerosis, as well as other diseases with a smooth muscle component.
Collapse
Affiliation(s)
- Susanne Feil
- From the Interfakultäres Institut für Biochemie (S.F., F.E., K.S.-O., R.F.), Department of Dermatology (B.F., M.S.), and Pharmakologie, Toxikologie und Klinische Pharmazie (R.L.), University of Tübingen, Tübingen, Germany
| | - Birgit Fehrenbacher
- From the Interfakultäres Institut für Biochemie (S.F., F.E., K.S.-O., R.F.), Department of Dermatology (B.F., M.S.), and Pharmakologie, Toxikologie und Klinische Pharmazie (R.L.), University of Tübingen, Tübingen, Germany
| | - Robert Lukowski
- From the Interfakultäres Institut für Biochemie (S.F., F.E., K.S.-O., R.F.), Department of Dermatology (B.F., M.S.), and Pharmakologie, Toxikologie und Klinische Pharmazie (R.L.), University of Tübingen, Tübingen, Germany
| | - Frank Essmann
- From the Interfakultäres Institut für Biochemie (S.F., F.E., K.S.-O., R.F.), Department of Dermatology (B.F., M.S.), and Pharmakologie, Toxikologie und Klinische Pharmazie (R.L.), University of Tübingen, Tübingen, Germany
| | - Klaus Schulze-Osthoff
- From the Interfakultäres Institut für Biochemie (S.F., F.E., K.S.-O., R.F.), Department of Dermatology (B.F., M.S.), and Pharmakologie, Toxikologie und Klinische Pharmazie (R.L.), University of Tübingen, Tübingen, Germany
| | - Martin Schaller
- From the Interfakultäres Institut für Biochemie (S.F., F.E., K.S.-O., R.F.), Department of Dermatology (B.F., M.S.), and Pharmakologie, Toxikologie und Klinische Pharmazie (R.L.), University of Tübingen, Tübingen, Germany
| | - Robert Feil
- From the Interfakultäres Institut für Biochemie (S.F., F.E., K.S.-O., R.F.), Department of Dermatology (B.F., M.S.), and Pharmakologie, Toxikologie und Klinische Pharmazie (R.L.), University of Tübingen, Tübingen, Germany.
| |
Collapse
|
2318
|
Coulson-Thomas VJ, Gesteira TF, Esko J, Kao W. Heparan sulfate regulates hair follicle and sebaceous gland morphogenesis and homeostasis. J Biol Chem 2014; 289:25211-26. [PMID: 25053416 DOI: 10.1074/jbc.m114.572511] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Hair follicle (HF) morphogenesis and cycling are a result of intricate autonomous epithelial-mesenchymal interactions. Once the first HF cycle is complete it repeatedly undergoes cyclic transformations. Heparan sulfate (HS) proteoglycans are found on the cell surface and in the extracellular matrix where they influence a variety of biological processes by interacting with physiologically important proteins, such as growth factors. Inhibition of heparanase (an HS endoglycosidase) in in vitro cultured HFs has been shown to induce a catagen-like process. Therefore, this study aimed to elucidate the precise role of HS in HF morphogenesis and cycling. An inducible tetratransgenic mouse model was generated to excise exostosin glycosyltransferase 1 (Ext1) in keratin 14-positive cells from P21. Interestingly, EXT1(StEpiΔ/StEpiΔ) mice presented solely anagen HFs. Moreover, waxing the fur to synchronize the HFs revealed accelerated hair regrowth in the EXT1(StEpiΔ/StEpiΔ) mice and hindered cycling into catagen. The ablation of HS in the interfollicular epidermal cells of mature skin led to the spontaneous formation of new HFs and an increase in Sonic Hedgehog expression resembling wild-type mice at P0, thereby indicating that the HS/Sonic Hedgehog signaling pathway regulates HF formation during embryogenesis and prevents HF formation in mature skin. Finally, the knock-out of HS also led to the morphogenesis and hyperplasia of sebaceous glands and sweat glands in mature mice, leading to exacerbated sebum production and accumulation on the skin surface. Therefore, our findings clearly show that an intricate control of HS levels is required for HF, sebaceous gland, and sweat gland morphogenesis and HF cycling.
Collapse
Affiliation(s)
| | - Tarsis Ferreira Gesteira
- From the Department of Ophthalmology, University of Cincinnati, Cincinnati, Ohio 45267-0838, Division of Developmental Biology, Cincinnati Children's Hospital and Research, Cincinnati, Ohio 45229-3039, and
| | - Jeffrey Esko
- Department of Cellular and Molecular Medicine, Glycobiology Research and Training Center, University of California, San Diego, La Jolla, California 92093-0687
| | - Winston Kao
- From the Department of Ophthalmology, University of Cincinnati, Cincinnati, Ohio 45267-0838
| |
Collapse
|
2319
|
Kizhatil K, Ryan M, Marchant JK, Henrich S, John SWM. Schlemm's canal is a unique vessel with a combination of blood vascular and lymphatic phenotypes that forms by a novel developmental process. PLoS Biol 2014; 12:e1001912. [PMID: 25051267 PMCID: PMC4106723 DOI: 10.1371/journal.pbio.1001912] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/12/2014] [Indexed: 01/04/2023] Open
Abstract
A draining vessel in the eye arises via a novel hybrid process of vascular development and is important for understanding ocular fluid homeostasis and glaucoma. Schlemm's canal (SC) plays central roles in ocular physiology. These roles depend on the molecular phenotypes of SC endothelial cells (SECs). Both the specific phenotype of SECs and development of SC remain poorly defined. To allow a modern and extensive analysis of SC and its origins, we developed a new whole-mount procedure to visualize its development in the context of surrounding tissues. We then applied genetic lineage tracing, specific-fluorescent reporter genes, immunofluorescence, high-resolution confocal microscopy, and three-dimensional (3D) rendering to study SC. Using these techniques, we show that SECs have a unique phenotype that is a blend of both blood and lymphatic endothelial cell phenotypes. By analyzing whole mounts of postnatal mouse eyes progressively to adulthood, we show that SC develops from blood vessels through a newly discovered process that we name “canalogenesis.” Functional inhibition of KDR (VEGFR2), a critical receptor in initiating angiogenesis, shows that this receptor is required during canalogenesis. Unlike angiogenesis and similar to stages of vasculogenesis, during canalogenesis tip cells divide and form branched chains prior to vessel formation. Differing from both angiogenesis and vasculogenesis, during canalogenesis SECs express Prox1, a master regulator of lymphangiogenesis and lymphatic phenotypes. Thus, SC development resembles a blend of vascular developmental programs. These advances define SC as a unique vessel with a combination of blood vascular and lymphatic phenotypes. They are important for dissecting its functions that are essential for ocular health and normal vision. Schlemm's canal serves as a drainage tube for fluid from the anterior chamber of the eye and is directly relevant to glaucoma, a disease that causes vision loss in over 70 million people. Aqueous humor enters the canal and then drains into connected veins. Molecular understanding of the development of Schlemm's canal and its drainage functions has remained limited. We provide a detailed characterization of Schlemm's canal development, and in so doing discover a novel process of vascular development that we name “canalogenesis.” We show that although the process requires a functional KDR receptor, which is also critical in blood vessel development, the endothelial cells of Schlemm's canal have a unique hybrid molecular phenotype, expressing proteins that are characteristic of both blood and lymphatic vessels. Of note, the expression of Prox1, a master regulator of lymphatic fate, and other lymphatic proteins are largely restricted to specialized cells of the inner wall of Schlemm's canal through which the aqueous humor passes as it exits the eye. Thus, Prox1 and other lymphatic proteins may be critical for the functional specialization of these cells for aqueous humor drainage. Schlemm's canal is thus a unique vessel with a combination of blood vascular and lymphatic characteristics.
Collapse
Affiliation(s)
- Krishnakumar Kizhatil
- The Howard Hughes Medical Institute, and The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Margaret Ryan
- The Howard Hughes Medical Institute, and The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Jeffrey K. Marchant
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Stephen Henrich
- The Howard Hughes Medical Institute, and The Jackson Laboratory, Bar Harbor, Maine, United States of America
| | - Simon W. M. John
- The Howard Hughes Medical Institute, and The Jackson Laboratory, Bar Harbor, Maine, United States of America
- Department of Ophthalmology and Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
2320
|
Greggio C, De Franceschi F, Figueiredo-Larsen M, Grapin-Botton A. In vitro pancreas organogenesis from dispersed mouse embryonic progenitors. J Vis Exp 2014. [PMID: 25079453 DOI: 10.3791/51725] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The pancreas is an essential organ that regulates glucose homeostasis and secretes digestive enzymes. Research on pancreas embryogenesis has led to the development of protocols to produce pancreatic cells from stem cells (1). The whole embryonic organ can be cultured at multiple stages of development (2-4). These culture methods have been useful to test drugs and to image developmental processes. However the expansion of the organ is very limited and morphogenesis is not faithfully recapitulated since the organ flattens. We propose three-dimensional (3D) culture conditions that enable the efficient expansion of dissociated mouse embryonic pancreatic progenitors. By manipulating the composition of the culture medium it is possible to generate either hollow spheres, mainly composed of pancreatic progenitors expanding in their initial state, or, complex organoids which progress to more mature expanding progenitors and differentiate into endocrine, acinar and ductal cells and which spontaneously self-organize to resemble the embryonic pancreas. We show here that the in vitro process recapitulates many aspects of natural pancreas development. This culture system is suitable to investigate how cells cooperate to form an organ by reducing its initial complexity to few progenitors. It is a model that reproduces the 3D architecture of the pancreas and that is therefore useful to study morphogenesis, including polarization of epithelial structures and branching. It is also appropriate to assess the response to mechanical cues of the niche such as stiffness and the effects on cell´s tensegrity.
Collapse
Affiliation(s)
- Chiara Greggio
- Ecole Polytechnique Fédérale de Lausanne, School of Life Sciences, Swiss Institute for Experimental Cancer Research
| | - Filippo De Franceschi
- Ecole Polytechnique Fédérale de Lausanne, School of Life Sciences, Swiss Institute for Experimental Cancer Research
| | | | - Anne Grapin-Botton
- Ecole Polytechnique Fédérale de Lausanne, School of Life Sciences, Swiss Institute for Experimental Cancer Research; DanStem, University of Copenhagen;
| |
Collapse
|
2321
|
Moding EJ, Lee CL, Castle KD, Oh P, Mao L, Zha S, Min HD, Ma Y, Das S, Kirsch DG. Atm deletion with dual recombinase technology preferentially radiosensitizes tumor endothelium. J Clin Invest 2014; 124:3325-38. [PMID: 25036710 DOI: 10.1172/jci73932] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 05/15/2014] [Indexed: 02/06/2023] Open
Abstract
Cells isolated from patients with ataxia telangiectasia are exquisitely sensitive to ionizing radiation. Kinase inhibitors of ATM, the gene mutated in ataxia telangiectasia, can sensitize tumor cells to radiation therapy, but concern that inhibiting ATM in normal tissues will also increase normal tissue toxicity from radiation has limited their clinical application. Endothelial cell damage can contribute to the development of long-term side effects after radiation therapy, but the role of endothelial cell death in tumor response to radiation therapy remains controversial. Here, we developed dual recombinase technology using both FlpO and Cre recombinases to generate primary sarcomas in mice with endothelial cell-specific deletion of Atm to determine whether loss of Atm in endothelial cells sensitizes tumors and normal tissues to radiation. Although deletion of Atm in proliferating tumor endothelial cells enhanced the response of sarcomas to radiation, Atm deletion in quiescent endothelial cells of the heart did not sensitize mice to radiation-induced myocardial necrosis. Blocking cell cycle progression reversed the effect of Atm loss on tumor endothelial cell radiosensitivity. These results indicate that endothelial cells must progress through the cell cycle in order to be radiosensitized by Atm deletion.
Collapse
|
2322
|
Howard JM, Nuguid JM, Ngole D, Nguyen H. Tcf3 expression marks both stem and progenitor cells in multiple epithelia. Development 2014; 141:3143-52. [PMID: 25038042 DOI: 10.1242/dev.106989] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The Lef/Tcf-family transcription factor Tcf3 has important roles in development, stem cell function and malignancy. Previous gain- and loss-of-function studies have suggested that Tcf3 is a mediator of self-renewal and an undifferentiated state in stem and progenitor cells in skin, but little is known of its role in other postnatal tissues. Here, we explore the distribution and behavior of Tcf3-expressing cells in several adult tissues using a novel Tcf3-CreER knock-in mouse model. By lineage tracing in dorsal skin, we verify that Tcf3-expressing cells in the hair follicle bulge are self-renewing stem cells with multilineage potential. We then demonstrate, for the first time, the presence of Tcf3-expressing cells in the basal layer of several other stratified epithelia, including the paw skin, tongue and esophagus. By lineage tracing, we demonstrate that the Tcf3-expressing population in these tissues includes persistent stem cells, transient progenitors and cells undergoing active differentiation. Our observations here suggest that the role of Tcf3 in cell-fate decision is more complex than previously appreciated and is highly dependent on cellular context.
Collapse
Affiliation(s)
- Jeffrey M Howard
- Stem Cells and Regenerative Medicine (STaR) Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA Medical Scientist Training Program, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Justine M Nuguid
- Stem Cells and Regenerative Medicine (STaR) Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA School of Medicine, Southern Illinois University, PO Box 19620, Springfield, IL 62794, USA
| | - Diana Ngole
- Stem Cells and Regenerative Medicine (STaR) Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hoang Nguyen
- Stem Cells and Regenerative Medicine (STaR) Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA Department of Dermatology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
2323
|
Tian X, Hu T, Zhang H, He L, Huang X, Liu Q, Yu W, He L, Yang Z, Yan Y, Yang X, Zhong TP, Pu WT, Zhou B. Vessel formation. De novo formation of a distinct coronary vascular population in neonatal heart. Science 2014; 345:90-4. [PMID: 24994653 DOI: 10.1126/science.1251487] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The postnatal coronary vessels have been viewed as developing through expansion of vessels formed during the fetal period. Using genetic lineage tracing, we found that a substantial portion of postnatal coronary vessels arise de novo in the neonatal mouse heart, rather than expanding from preexisting embryonic vasculature. Our data show that lineage conversion of neonatal endocardial cells during trabecular compaction generates a distinct compartment of the coronary circulation located within the inner half of the ventricular wall. This lineage conversion occurs within a brief period after birth and provides an efficient means of rapidly augmenting the coronary vasculature. This mechanism of postnatal coronary vascular growth provides avenues for understanding and stimulating cardiovascular regeneration following injury and disease.
Collapse
Affiliation(s)
- Xueying Tian
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tianyuan Hu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hui Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Lingjuan He
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiuzhen Huang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Qiaozhen Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wei Yu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Liang He
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhen Yang
- Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan Yan
- Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiao Yang
- State Key Laboratory of Proteomics, Institute of Biotechnology, Beijing 100071, China
| | - Tao P Zhong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - William T Pu
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA. Department of Cardiology, Children's Hospital Boston, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Bin Zhou
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
2324
|
Dual fluorescent reporter pig for Cre recombination: transgene placement at the ROSA26 locus. PLoS One 2014; 9:e102455. [PMID: 25025770 PMCID: PMC4099177 DOI: 10.1371/journal.pone.0102455] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 06/18/2014] [Indexed: 01/27/2023] Open
Abstract
We are extending the Cre/loxP site-specific recombination system to pigs, focussing on conditional and tissue-specific expression of oncogenic mutations to model human cancers. Identifying the location, pattern and extent of Cre recombination in vivo is an important aspect of this technology. Here we report pigs with a dual fluorochrome cassette under the control of the strong CAG promoter that switches expression after Cre-recombination, from membrane-targeted tandem dimer Tomato to membrane-targeted green fluorescent protein. The reporter cassette was placed at the porcine ROSA26 locus by conventional gene targeting using primary mesenchymal stem cells, and animals generated by nuclear transfer. Gene targeting efficiency was high, and analysis of foetal organs and primary cells indicated that the reporter is highly expressed and functional. Cre reporter pigs will provide a multipurpose indicator of Cre recombinase activity, an important new tool for the rapidly expanding field of porcine genetic modification.
Collapse
|
2325
|
Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology. Nat Commun 2014; 4:2823. [PMID: 24264436 PMCID: PMC4059406 DOI: 10.1038/ncomms3823] [Citation(s) in RCA: 1040] [Impact Index Per Article: 94.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 10/25/2013] [Indexed: 02/07/2023] Open
Abstract
Although organ fibrosis causes significant morbidity and mortality in chronic diseases, the lack of detailed knowledge about specific cellular contributors mediating fibrogenesis hampers the design of effective antifibrotic therapies. Different cellular sources, including tissue-resident and bone marrow-derived fibroblasts, pericytes and epithelial cells, have been suggested to give rise to myofibroblasts, but their relative contributions remain controversial, with profound differences between organs and different diseases. Here we employ a novel Cre-transgenic mouse that marks 99% of hepatic stellate cells (HSCs), a liver-specific pericyte population, to demonstrate that HSCs give rise to 82-96% of myofibroblasts in models of toxic, cholestatic and fatty liver disease. Moreover, we exclude that HSCs function as facultative epithelial progenitor cells in the injured liver. On the basis these findings, HSCs should be considered the primary cellular target for antifibrotic therapies across all types of liver disease.
Collapse
|
2326
|
Hung CM, Calejman CM, Sanchez-Gurmaches J, Li H, Clish CB, Hettmer S, Wagers AJ, Guertin DA. Rictor/mTORC2 loss in the Myf5 lineage reprograms brown fat metabolism and protects mice against obesity and metabolic disease. Cell Rep 2014; 8:256-71. [PMID: 25001283 DOI: 10.1016/j.celrep.2014.06.007] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 04/30/2014] [Accepted: 06/04/2014] [Indexed: 12/24/2022] Open
Abstract
The in vivo functions of mechanistic target of rapamycin complex 2 (mTORC2) and the signaling mechanisms that control brown adipose tissue (BAT) fuel utilization and activity are not well understood. Here, by conditionally deleting Rictor in the Myf5 lineage, we provide in vivo evidence that mTORC2 is dispensable for skeletal muscle development and regeneration but essential for BAT growth. Furthermore, deleting Rictor in Myf5 precursors shifts BAT metabolism to a more oxidative and less lipogenic state and protects mice from obesity and metabolic disease at thermoneutrality. We additionally find that Rictor is required for brown adipocyte differentiation in vitro and that the mechanism specifically requires AKT1 hydrophobic motif phosphorylation but is independent of pan-AKT signaling and is rescued with BMP7. Our findings provide insights into the signaling circuitry that regulates brown adipocytes and could have important implications for developing therapies aimed at increasing energy expenditure as a means to combat human obesity.
Collapse
Affiliation(s)
- Chien-Min Hung
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Camila Martinez Calejman
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Joan Sanchez-Gurmaches
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Huawei Li
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | - Simone Hettmer
- Howard Hughes Medical Institute, USA; Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 01238, USA; Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA; Department of Pediatric Oncology, Dana Farber Cancer Institute, Boston, MA 02155, USA; Division of Pediatric Hematology/Oncology, Children's Hospital, Boston, MA 02155, USA
| | - Amy J Wagers
- Howard Hughes Medical Institute, USA; Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 01238, USA; Joslin Diabetes Center, One Joslin Place, Boston, MA 02215, USA; Department of Pediatric Oncology, Dana Farber Cancer Institute, Boston, MA 02155, USA
| | - David A Guertin
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
2327
|
Jeffery E, Berry R, Church CD, Yu S, Shook BA, Horsley V, Rosen ED, Rodeheffer MS. Characterization of Cre recombinase models for the study of adipose tissue. Adipocyte 2014; 3:206-11. [PMID: 25068087 PMCID: PMC4110097 DOI: 10.4161/adip.29674] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 06/20/2014] [Accepted: 06/20/2014] [Indexed: 02/06/2023] Open
Abstract
The study of adipose tissue in vivo has been significantly advanced through the use of genetic mouse models. While the aP2-Cre(BI) and aP2-Cre(Salk) lines have been widely used to target adipose tissue, the specificity of these lines for adipocytes has recently been questioned. Here we characterize Cre recombinase activity in multiple cell populations of the major adipose tissue depots of these and other Cre lines using the membrane-Tomato/membrane-GFP (mT/mG) dual fluorescent reporter. We find that the aP2-Cre(BI) and aP2-Cre(Salk) lines lack specificity for adipocytes within adipose tissue, and that the aP2-Cre(BI) line does not efficiently target adipocytes in white adipose depots. Alternatively, the Adiponectin-CreERT line shows high efficiency and specificity for adipocytes, while the PdgfRα-CreERUCL and PdgfRα-CreERJHU lines do not efficiently target adipocyte precursor cells in the major adipose depots. Instead, we show that the PdgfRα-Cre line is preferable for studies targeting adipocyte precursor cells in vivo.
Collapse
Affiliation(s)
- Elise Jeffery
- Department of Cell Biology; Yale University; New Haven, CT USA
| | - Ryan Berry
- Department of Molecular, Cell and Developmental Biology; Yale University; New Haven, CT USA
| | | | - Songtao Yu
- Department of Pediatrics; Children’s Memorial Research Center; Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Brett A Shook
- Department of Molecular, Cell and Developmental Biology; Yale University; New Haven, CT USA
| | - Valerie Horsley
- Department of Molecular, Cell and Developmental Biology; Yale University; New Haven, CT USA
- Yale Stem Cell Center; Yale University; New Haven, CT USA
| | - Evan D Rosen
- Division of Endocrinology; Beth Israel Deaconess Medical Center; Boston, MA USA
- Harvard Medical School; Boston, MA USA
| | - Matthew S Rodeheffer
- Department of Molecular, Cell and Developmental Biology; Yale University; New Haven, CT USA
- Section of Comparative Medicine; Yale University; New Haven, CT USA
- Yale Stem Cell Center; Yale University; New Haven, CT USA
| |
Collapse
|
2328
|
Tevosian SG. Transgenic mouse models in the study of reproduction: insights into GATA protein function. Reproduction 2014; 148:R1-R14. [DOI: 10.1530/rep-14-0086] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
For the past 2 decades, transgenic technology in mice has allowed for an unprecedented insight into the transcriptional control of reproductive development and function. The key factor among the mouse genetic tools that made this rapid advance possible is a conditional transgenic approach, a particularly versatile method of creating gene deletions and substitutions in the mouse genome. A centerpiece of this strategy is an enzyme, Cre recombinase, which is expressed from defined DNA regulatory elements that are active in the tissue of choice. The regulatory DNA element (either genetically engineered or natural) assures Cre expression only in predetermined cell types, leading to the guided deletion of genetically modified (flanked by loxP or ‘floxed’ byloxP) gene loci. This review summarizes and compares the studies in which genes encoding GATA family transcription factors were targeted either globally or by Cre recombinases active in the somatic cells of ovaries and testes. The conditional gene loss experiments require detailed knowledge of the spatial and temporal expression of Cre activity, and the challenges in interpreting the outcomes are highlighted. These studies also expose the complexity of GATA-dependent regulation of gonadal gene expression and suggest that gene function is highly context dependent.
Collapse
|
2329
|
DeGraff DJ, Grabowska MM, Case T, Yu X, Herrick MK, Hayward W, Strand DW, Cates JM, Hayward SW, Gao N, Walter MA, Buttyan R, Yi Y, Kaestner KH, Matusik RJ. FOXA1 deletion in luminal epithelium causes prostatic hyperplasia and alteration of differentiated phenotype. J Transl Med 2014; 94:726-39. [PMID: 24840332 PMCID: PMC4451837 DOI: 10.1038/labinvest.2014.64] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 02/20/2014] [Accepted: 03/26/2014] [Indexed: 01/25/2023] Open
Abstract
The forkhead box (Fox) superfamily of transcription factors has essential roles in organogenesis and tissue differentiation. Foxa1 and Foxa2 are expressed during prostate budding and ductal morphogenesis, whereas Foxa1 expression is retained in adult prostate epithelium. Previous characterization of prostatic tissue rescued from embryonic Foxa1 knockout mice revealed Foxa1 to be essential for ductal morphogenesis and epithelial maturation. However, it is unknown whether Foxa1 is required to maintain the differentiated status in adult prostate epithelium. Here, we employed the PBCre4 transgenic system and determined the impact of prostate-specific Foxa1 deletion in adult murine epithelium. PBCre4/Foxa1(loxp/loxp) mouse prostates showed progressive florid hyperplasia with extensive cribriform patterning, with the anterior prostate being most affected. Immunohistochemistry studies show mosaic Foxa1 KO consistent with PBCre4 activity, with Foxa1 KO epithelial cells specifically exhibiting altered cell morphology, increased proliferation, and elevated expression of basal cell markers. Castration studies showed that, while PBCre4/Foxa1(loxp/loxp) prostates did not exhibit altered sensitivity in response to hormone ablation compared with control prostates, the number of Foxa1-positive cells in mosaic Foxa1 KO prostates was significantly reduced compared with Foxa1-negative cells following castration. Unexpectedly, gene expression profile analyses revealed that Foxa1 deletion caused abnormal expression of seminal vesicle-associated genes in KO prostates. In summary, these results indicate Foxa1 expression is required for the maintenance of prostatic cellular differentiation.
Collapse
Affiliation(s)
- David J. DeGraff
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | | | - Tom Case
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - Xiuping Yu
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - Mary K. Herrick
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - William Hayward
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - Douglas W. Strand
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - Justin M. Cates
- Department of Pathology, Vanderbilt University Medical Center, Nashville TN
| | - Simon W. Hayward
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark NJ
| | | | | | - Yajun Yi
- Institute for Integrative Genomics and Department of Medicine, Vanderbilt University, Nashville TN
| | | | - Robert J. Matusik
- Department of Urologic Surgery, Vanderbilt University Medical Center, Nashville TN,Department of Cell and Developmental Biology, Vanderbilt University, Nashville TN,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville TN,Correspondence and reprint requests should be made to: Robert J. Matusik, Ph.D., William L. Bray Chair of Urologic Surgery, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232,
| |
Collapse
|
2330
|
Lua I, James D, Wang J, Wang KS, Asahina K. Mesodermal mesenchymal cells give rise to myofibroblasts, but not epithelial cells, in mouse liver injury. Hepatology 2014; 60:311-22. [PMID: 24488807 PMCID: PMC4077971 DOI: 10.1002/hep.27035] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 01/26/2014] [Indexed: 12/18/2022]
Abstract
UNLABELLED Hepatic stellate cells (HSCs) and portal fibroblasts (PFs) are believed to be the major source of myofibroblasts that participate in fibrogenesis by way of synthesis of proinflammatory cytokines and extracellular matrices. Previous lineage tracing studies using MesP1(Cre) and Rosa26lacZ(flox) mice demonstrated that MesP1+ mesoderm gives rise to mesothelial cells (MCs), which differentiate into HSCs and PFs during liver development. In contrast, several in vivo and in vitro studies reported that HSCs can differentiate into other cell types, including hepatocytes, cholangiocytes, and progenitor cell types known as oval cells, thereby acting as stem cells in the liver. To test whether HSCs give rise to epithelial cells in adult liver, we determined the hepatic lineages of HSCs and PFs using MesP1(Cre) and Rosa26mTmG(flox) mice. Genetic cell lineage tracing revealed that the MesP1+ mesoderm gives rise to MCs, HSCs, and PFs, but not to hepatocytes or cholangiocytes, in the adult liver. Upon carbon tetrachloride injection or bile duct ligation surgery-mediated liver injury, mesodermal mesenchymal cells, including HSCs and PFs, differentiate into myofibroblasts but not into hepatocytes or cholangiocytes. Furthermore, differentiation of the mesodermal mesenchymal cells into oval cells was not observed. These results indicate that HSCs are not sufficiently multipotent to produce hepatocytes, cholangiocytes, or oval cells by way of mesenchymal-epithelial transition in vivo. CONCLUSION Cell lineage tracing demonstrated that mesodermal mesenchymal cells including HSCs are the major source of myofibroblasts but do not differentiate into epithelial cell types such as hepatocytes, cholangiocytes, and oval cells.
Collapse
Affiliation(s)
- Ingrid Lua
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, Keck School of Medicine, University of Southern California
| | - David James
- Saban Research Institute, Children’s Hospital Los Angeles
| | - Jiaohong Wang
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, Keck School of Medicine, University of Southern California
| | - Kasper S. Wang
- Saban Research Institute, Children’s Hospital Los Angeles
| | - Kinji Asahina
- Southern California Research Center for ALPD and Cirrhosis, Department of Pathology, Keck School of Medicine, University of Southern California
| |
Collapse
|
2331
|
He Y, Zhang H, Yung A, Villeda SA, Jaeger PA, Olayiwola O, Fainberg N, Wyss-Coray T. ALK5-dependent TGF-β signaling is a major determinant of late-stage adult neurogenesis. Nat Neurosci 2014; 17:943-52. [PMID: 24859199 PMCID: PMC4096284 DOI: 10.1038/nn.3732] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 04/30/2014] [Indexed: 01/19/2023]
Abstract
The transforming growth factor-β (TGF-β) signaling pathway serves critical functions in CNS development, but, apart from its proposed neuroprotective actions, its physiological role in the adult brain is unclear. We observed a prominent activation of TGF-β signaling in the adult dentate gyrus and expression of downstream Smad proteins in this neurogenic zone. Consistent with a function of TGF-β signaling in adult neurogenesis, genetic deletion of the TGF-β receptor ALK5 reduced the number, migration and dendritic arborization of newborn neurons. Conversely, constitutive activation of neuronal ALK5 in forebrain caused a marked increase in these aspects of neurogenesis and was associated with higher expression of c-Fos in newborn neurons and with stronger memory function. Our findings describe an unexpected role for ALK5-dependent TGF-β signaling as a regulator of the late stages of adult hippocampal neurogenesis, which may have implications for changes in neurogenesis during aging and disease.
Collapse
Affiliation(s)
- Yingbo He
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Hui Zhang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Andrea Yung
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Saul A Villeda
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Philipp A Jaeger
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Oluwatobi Olayiwola
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Nina Fainberg
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305, USA
- Center for Tissue Regeneration, Repair and Rehabilitation, VA Palo Alto Health Care System, Palo Alto, California 94304, USA
| |
Collapse
|
2332
|
Mangiavini L, Merceron C, Araldi E, Khatri R, Gerard-O'Riley R, Wilson TL, Rankin EB, Giaccia AJ, Schipani E. Loss of VHL in mesenchymal progenitors of the limb bud alters multiple steps of endochondral bone development. Dev Biol 2014; 393:124-36. [PMID: 24972088 DOI: 10.1016/j.ydbio.2014.06.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 05/24/2014] [Accepted: 06/17/2014] [Indexed: 01/21/2023]
Abstract
Adaptation to low oxygen tension (hypoxia) is a critical event during development. The transcription factors Hypoxia Inducible Factor-1α (HIF-1α) and HIF-2α are essential mediators of the homeostatic responses that allow hypoxic cells to survive and differentiate. Von Hippel-Lindau protein (VHL) is the E3 ubiquitin ligase that targets HIFs to the proteasome for degradation in normoxia. We have previously demonstrated that the transcription factor HIF-1α is essential for survival and differentiation of growth plate chondrocytes, whereas HIF-2α is not necessary for fetal growth plate development. We have also shown that VHL is important for endochondral bone development, since loss of VHL in chondrocytes causes severe dwarfism. In this study, in order to expand our understanding of the role of VHL in chondrogenesis, we conditionally deleted VHL in mesenchymal progenitors of the limb bud, i.e. in cells not yet committed to the chondrocyte lineage. Deficiency of VHL in limb bud mesenchyme does not alter the timely differentiation of mesenchymal cells into chondrocytes. However, it causes structural collapse of the cartilaginous growth plate as a result of impaired proliferation, delayed terminal differentiation, and ectopic death of chondrocytes. This phenotype is associated to delayed replacement of cartilage by bone. Notably, loss of HIF-2α fully rescues the late formation of the bone marrow cavity in VHL mutant mice, though it does not affect any other detectable abnormality of the VHL mutant growth plates. Our findings demonstrate that VHL regulates bone morphogenesis as its loss considerably alters size, shape and overall development of the skeletal elements.
Collapse
Affiliation(s)
- Laura Mangiavini
- Department of Orthopaedic Surgery, Medical School, University of Michigan, Ann Arbor, MI 48109, USA; Division of Endocrinology, Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Orthopaedic and Traumatology, Milano-Bicocca University, 20900 Monza (MB), Italy
| | - Christophe Merceron
- Department of Orthopaedic Surgery, Medical School, University of Michigan, Ann Arbor, MI 48109, USA; Division of Endocrinology, Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Inserm, UMRS 791-LIOAD, Centre for Osteoarticular and Dental Tissue Engineering, Group STEP 'Skeletal Tissue Engineering and Physiopathology', 44042 Nantes, France; LUNAM, Nantes University, Faculty of Dental Surgery, Nantes, France
| | - Elisa Araldi
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Richa Khatri
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Rita Gerard-O'Riley
- Division of Endocrinology, Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Tremika LeShan Wilson
- Department of Orthopaedic Surgery, Medical School, University of Michigan, Ann Arbor, MI 48109, USA; Division of Endocrinology, Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Erinn B Rankin
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, Stanford, CA 94303-5152, USA
| | - Amato J Giaccia
- Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University, Stanford, CA 94303-5152, USA
| | - Ernestina Schipani
- Department of Orthopaedic Surgery, Medical School, University of Michigan, Ann Arbor, MI 48109, USA; Division of Endocrinology, Department of Medicine, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Division of Endocrinology, Department of Medicine, Medical School, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
2333
|
Pohlkamp T, Steller L, May P, Günther T, Schüle R, Frotscher M, Herz J, Bock HH. Generation and characterization of an Nse-CreERT2 transgenic line suitable for inducible gene manipulation in cerebellar granule cells. PLoS One 2014; 9:e100384. [PMID: 24950299 PMCID: PMC4065071 DOI: 10.1371/journal.pone.0100384] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 05/27/2014] [Indexed: 12/15/2022] Open
Abstract
We created an Nse-CreERT2 mouse line expressing the tamoxifen-inducible CreERT2 recombinase under the control of the neuron-specific enolase (Nse) promoter. By using Cre reporter lines we could show that this Nse-CreERT2 line has recombination activity in the granule cells of all cerebellar lobules as well as in postmitotic granule cell precursors in the external granular layer of the developing cerebellum. A few hippocampal dentate gyrus granule cells showed Cre-mediated recombination as well. Cre activity could be induced in both the developing and adult mouse brain. The established mouse line constitutes a valuable tool to study the function of genes expressed by cerebellar granule cells in the developing and adult brain. In combination with reporter lines it is a useful model to analyze the development and maintenance of the cerebellar architecture including granule cell distribution, migration, and the extension of granule cell fibers in vivo.
Collapse
Affiliation(s)
- Theresa Pohlkamp
- Center for Neuroscience, Department of Neuroanatomy, Albert-Ludwigs-University, Freiburg, Germany
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail: (TP); (JH); (HHB)
| | - Laura Steller
- Center for Neuroscience, Department of Neuroanatomy, Albert-Ludwigs-University, Freiburg, Germany
| | - Petra May
- Center for Neuroscience, Department of Neuroanatomy, Albert-Ludwigs-University, Freiburg, Germany
- Clinic for Gastroenterology, Hepatology and Infectiology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Thomas Günther
- Department of Urology, University Hospital Freiburg, Freiburg, Germany
| | - Roland Schüle
- Department of Urology, University Hospital Freiburg, Freiburg, Germany
| | - Michael Frotscher
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, Hamburg, Germany
| | - Joachim Herz
- Center for Neuroscience, Department of Neuroanatomy, Albert-Ludwigs-University, Freiburg, Germany
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail: (TP); (JH); (HHB)
| | - Hans H. Bock
- Center for Neuroscience, Department of Neuroanatomy, Albert-Ludwigs-University, Freiburg, Germany
- Clinic for Gastroenterology, Hepatology and Infectiology, Heinrich-Heine-University, Düsseldorf, Germany
- * E-mail: (TP); (JH); (HHB)
| |
Collapse
|
2334
|
Nadeau V, Charron J. Essential role of the ERK/MAPK pathway in blood-placental barrier formation. Development 2014; 141:2825-37. [PMID: 24948605 DOI: 10.1242/dev.107409] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The mammalian genome contains two ERK/MAP kinase kinase genes, Map2k1 and Map2k2, which encode dual-specificity kinases responsible for ERK activation. Loss of Map2k1 function in mouse causes embryonic lethality due to placental defects, whereas Map2k2 mutants have a normal lifespan. The majority of Map2k1(+/-) Map2k2(+/-) embryos die during gestation from the underdevelopment of the placenta labyrinth, demonstrating that both kinases are involved in placenta formation. Map2k1(+/-) Map2k2(+/-) mutants show reduced vascularization of the labyrinth and defective formation of syncytiotrophoblast layer II (SynT-II) leading to the accumulation of multinucleated trophoblast giant cells (MTGs). To define the cell type-specific contribution of the ERK/MAPK pathway to placenta development, we performed deletions of Map2k1 function in different Map2k1 Map2k2 allelic backgrounds. Loss of MAP kinase kinase activity in pericytes or in allantois-derived tissues worsens the MTG phenotype. These results define the contribution of the ERK/MAPK pathway in specific embryonic and extraembryonic cell populations for normal placentation. Our data also indicate that MTGs could result from the aberrant fusion of SynT-I and -II. Using mouse genetics, we demonstrate that the normal development of SynT-I into a thin layer of multinucleated cells depends on the presence of SynT-II. Lastly, the combined mutations of Map2k1 and Map2k2 alter the expression of several genes involved in cell fate specification, cell fusion and cell polarity. Thus, appropriate ERK/MAPK signaling in defined cell types is required for the proper growth, differentiation and morphogenesis of the placenta.
Collapse
Affiliation(s)
- Valérie Nadeau
- Centre de recherche sur le cancer de l'Université Laval, Centre Hospitalier Universitaire de Québec, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, Canada G1R 2J6
| | - Jean Charron
- Centre de recherche sur le cancer de l'Université Laval, Centre Hospitalier Universitaire de Québec, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, Canada G1R 2J6
| |
Collapse
|
2335
|
Adipocytes arise from multiple lineages that are heterogeneously and dynamically distributed. Nat Commun 2014; 5:4099. [PMID: 24942009 PMCID: PMC4066194 DOI: 10.1038/ncomms5099] [Citation(s) in RCA: 275] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 05/12/2014] [Indexed: 12/15/2022] Open
Abstract
Adipose tissue development is poorly understood. Here we use a lineage-tracing strategy optimal for adipocytes to provide evidence that Myf5 precursors are not the exclusive source of brown adipocytes and contribute more to the mature white and brite adipocyte populations than previously thought. Moreover, Myf5 lineage distribution in adipose tissue changes in response to modifiable and non-modifiable factors. We also find that the Pax3 lineage largely overlaps with the Myf5 lineage in brown fat and subcutaneous white fat, but exhibits gender-linked divergence in visceral white fat while the MyoD1 lineage does not give rise to any adipocytes. Finally, by deleting insulin receptor beta in the Myf5 lineage, we provide in vivo evidence that the insulin receptor is essential for adipogenesis and that adipocyte lineages have plasticity. These data establish a conceptual framework for adipose tissue development and could explain body fat patterning variations in healthy and lipodystrophic or obese humans.
Collapse
|
2336
|
Brunet I, Gordon E, Han J, Cristofaro B, Broqueres-You D, Liu C, Bouvrée K, Zhang J, del Toro R, Mathivet T, Larrivée B, Jagu J, Pibouin-Fragner L, Pardanaud L, Machado MJC, Kennedy TE, Zhuang Z, Simons M, Levy BI, Tessier-Lavigne M, Grenz A, Eltzschig H, Eichmann A. Netrin-1 controls sympathetic arterial innervation. J Clin Invest 2014; 124:3230-40. [PMID: 24937433 DOI: 10.1172/jci75181] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 04/03/2014] [Indexed: 12/21/2022] Open
Abstract
Autonomic sympathetic nerves innervate peripheral resistance arteries, thereby regulating vascular tone and controlling blood supply to organs. Despite the fundamental importance of blood flow control, how sympathetic arterial innervation develops remains largely unknown. Here, we identified the axon guidance cue netrin-1 as an essential factor required for development of arterial innervation in mice. Netrin-1 was produced by arterial smooth muscle cells (SMCs) at the onset of innervation, and arterial innervation required the interaction of netrin-1 with its receptor, deleted in colorectal cancer (DCC), on sympathetic growth cones. Function-blocking approaches, including cell type-specific deletion of the genes encoding Ntn1 in SMCs and Dcc in sympathetic neurons, led to severe and selective reduction of sympathetic innervation and to defective vasoconstriction in resistance arteries. These findings indicate that netrin-1 and DCC are critical for the control of arterial innervation and blood flow regulation in peripheral organs.
Collapse
|
2337
|
Lund DK, Mouly V, Cornelison DDW. MMP-14 is necessary but not sufficient for invasion of three-dimensional collagen by human muscle satellite cells. Am J Physiol Cell Physiol 2014; 307:C140-9. [PMID: 24898588 DOI: 10.1152/ajpcell.00032.2014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The twenty-five known matrix metalloproteases (MMPs) and their endogenous inhibitors, tissue inhibitors of metalloproteases (TIMPs), mediate cell invasion through the extracellular matrix (ECM). In a comparative three-dimensional assay, we analyzed human and mouse satellite cells' competence to invade an artificial ECM (collagen I). We identified a single MMP that 1) is expressed by human muscle satellite cells; 2) is induced at the mRNA/protein level by adhesion to collagen I; and 3) is necessary for invasion into a collagen I matrix. Interestingly, murine satellite cells neither express this MMP, nor invade the collagen matrix. However, exogenous human MMP-14 is not sufficient to induce invasion of a collagen matrix by murine cells, emphasizing species differences.
Collapse
Affiliation(s)
- Dane K Lund
- Division of Biology and Bond Life Sciences Center, University of Missouri, Columbia, Missouri; and
| | - Vincent Mouly
- Institut de Myologie, Université Pierre et Marie Curie, Paris, France
| | - D D W Cornelison
- Division of Biology and Bond Life Sciences Center, University of Missouri, Columbia, Missouri; and
| |
Collapse
|
2338
|
Yu YR, You LR, Yan YT, Chen CM. Role of OVCA1/DPH1 in craniofacial abnormalities of Miller–Dieker syndrome. Hum Mol Genet 2014; 23:5579-96. [DOI: 10.1093/hmg/ddu273] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
2339
|
Notch regulation of myogenic versus endothelial fates of cells that migrate from the somite to the limb. Proc Natl Acad Sci U S A 2014; 111:8844-9. [PMID: 24927569 DOI: 10.1073/pnas.1407606111] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Multipotent Pax3-positive (Pax3(+)) cells in the somites give rise to skeletal muscle and to cells of the vasculature. We had previously proposed that this cell-fate choice depends on the equilibrium between Pax3 and Foxc2 expression. In this study, we report that the Notch pathway promotes vascular versus skeletal muscle cell fates. Overactivating the Notch pathway specifically in Pax3(+) progenitors, via a conditional Pax3(NICD) allele, results in an increase of the number of smooth muscle and endothelial cells contributing to the aorta. At limb level, Pax3(+) cells in the somite give rise to skeletal muscles and to a subpopulation of endothelial cells in blood vessels of the limb. We now demonstrate that in addition to the inhibitory role of Notch signaling on skeletal muscle cell differentiation, the Notch pathway affects the Pax3:Foxc2 balance and promotes the endothelial versus myogenic cell fate, before migration to the limb, in multipotent Pax3(+) cells in the somite of the mouse embryo.
Collapse
|
2340
|
Direct transcriptional repression of Zfp423 by Zfp521 mediates a bone morphogenic protein-dependent osteoblast versus adipocyte lineage commitment switch. Mol Cell Biol 2014; 34:3076-85. [PMID: 24891617 DOI: 10.1128/mcb.00185-14] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Osteoblasts and adipocytes arise from a common mesenchymal precursor cell. The cell fate decision of a mesenchymal precursor cell is under the influence of molecular cues and signaling pathways that lead to the activation or repression of lineage-specific transcription factors. The molecular mechanisms determining osteoblast versus adipocyte lineage specificity in response to bone morphogenic protein (BMP) remain unclear. In this study, we describe the mechanism through which Zfp521 (ZNF521), a regulator of lineage progression in multiple immature cell populations, regulates lineage specification of mesenchymal progenitor cells during BMP-induced differentiation events. In vivo deletion or in vitro knockdown of Zfp521 in mesenchymal precursors resulted in increased expression of the adipocyte determinant factor Zfp423 (ZNF423). This was concurrent with the loss of histone H3K9 methylation and an increase in histone H3K9 acetylation at the Zfp423 promoter, which together are indicative of decreased gene repression. Indeed, we found that Zfp521 occupies and represses the promoter and intronic enhancer regions of Zfp423. Accordingly, conditional deletion of Zfp521 inhibited heterotopic bone formation in response to local injection of BMP2. In contrast, marrow adiposity within BMP2-induced bone was markedly enhanced in Zfp521-deficient mice, suggesting that precursor cells lacking Zfp521 differentiate preferentially into adipocytes instead of osteoblasts in response to BMP2. Consistent with a cell-autonomous role of Zfp521 in mesenchymal precursors, knockdown of Zfp521 in stromal cells prevented BMP2-induced osteoblast marker expression and simultaneously enhanced lipid accumulation and expression of adipocyte-related genes. Taken together, the data suggest that Zfp521 is a cell fate switch critical for BMP-induced osteoblast commitment and identify Zfp521 as the intrinsic repressor of Zfp423 and hence of adipocyte commitment during BMP-induced mesenchymal precursor differentiation.
Collapse
|
2341
|
Seidelmann SB, Lighthouse JK, Greif DM. Development and pathologies of the arterial wall. Cell Mol Life Sci 2014; 71:1977-99. [PMID: 24071897 PMCID: PMC11113178 DOI: 10.1007/s00018-013-1478-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 09/11/2013] [Accepted: 09/12/2013] [Indexed: 01/13/2023]
Abstract
Arteries consist of an inner single layer of endothelial cells surrounded by layers of smooth muscle and an outer adventitia. The majority of vascular developmental studies focus on the construction of endothelial networks through the process of angiogenesis. Although many devastating vascular diseases involve abnormalities in components of the smooth muscle and adventitia (i.e., the vascular wall), the morphogenesis of these layers has received relatively less attention. Here, we briefly review key elements underlying endothelial layer formation and then focus on vascular wall development, specifically on smooth muscle cell origins and differentiation, patterning of the vascular wall, and the role of extracellular matrix and adventitial progenitor cells. Finally, we discuss select human diseases characterized by marked vascular wall abnormalities. We propose that continuing to apply approaches from developmental biology to the study of vascular disease will stimulate important advancements in elucidating disease mechanism and devising novel therapeutic strategies.
Collapse
MESH Headings
- Angiogenic Proteins/genetics
- Angiogenic Proteins/metabolism
- Animals
- Arteries/growth & development
- Arteries/metabolism
- Arteries/pathology
- Cardiovascular Diseases/genetics
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/pathology
- Cell Differentiation
- Cell Lineage/genetics
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Endothelium, Vascular/growth & development
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Gene Expression Regulation, Developmental
- Humans
- Morphogenesis/genetics
- Muscle, Smooth, Vascular/growth & development
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neovascularization, Pathologic
- Neovascularization, Physiologic
Collapse
Affiliation(s)
- Sara B. Seidelmann
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, 300 George St., Rm 773J, New Haven, CT 06511 USA
| | - Janet K. Lighthouse
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, 300 George St., Rm 773J, New Haven, CT 06511 USA
| | - Daniel M. Greif
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, 300 George St., Rm 773J, New Haven, CT 06511 USA
| |
Collapse
|
2342
|
Duval N, Daubas P, Bourcier de Carbon C, St Cloment C, Tinevez JY, Lopes M, Ribes V, Robert B. Msx1 and Msx2 act as essential activators of Atoh1 expression in the murine spinal cord. Development 2014; 141:1726-36. [PMID: 24715462 DOI: 10.1242/dev.099002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Dorsal spinal neurogenesis is orchestrated by the combined action of signals secreted from the roof plate organizer and a downstream transcriptional cascade. Within this cascade, Msx1 and Msx2, two homeodomain transcription factors (TFs), are induced earlier than bHLH neuralizing TFs. Whereas bHLH TFs have been shown to specify neuronal cell fate, the function of Msx genes remains poorly defined. We describe dramatic alterations of neuronal patterning in Msx1/Msx2 double-mutant mouse embryos. The most dorsal spinal progenitor pool fails to express the bHLH neuralizing TF Atoh1, which results in a lack of Lhx2-positive and Barhl2-positive dI1 interneurons. Neurog1 and Ascl1 expression territories are dorsalized, leading to ectopic dorsal differentiation of dI2 and dI3 interneurons. In proportion, the amount of Neurog1-expressing progenitors appears unaffected, whereas the number of Ascl1-positive cells is increased. These defects occur while BMP signaling is still active in the Msx1/Msx2 mutant embryos. Cell lineage analysis and co-immunolabeling demonstrate that Atoh1-positive cells derive from progenitors expressing both Msx1 and Msx2. In vitro, Msx1 and Msx2 proteins activate Atoh1 transcription by specifically interacting with several homeodomain binding sites in the Atoh1 3' enhancer. In vivo, Msx1 and Msx2 are required for Atoh1 3' enhancer activity and ChIP experiments confirm Msx1 binding to this regulatory sequence. These data support a novel function of Msx1 and Msx2 as transcriptional activators. Our study provides new insights into the transcriptional control of spinal cord patterning by BMP signaling, with Msx1 and Msx2 acting upstream of Atoh1.
Collapse
Affiliation(s)
- Nathalie Duval
- Institut Pasteur, Morphogenesis Molecular Genetics, CNRS URA 2578, 75015 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
2343
|
Bowen ME, Ayturk UM, Kurek KC, Yang W, Warman ML. SHP2 regulates chondrocyte terminal differentiation, growth plate architecture and skeletal cell fates. PLoS Genet 2014; 10:e1004364. [PMID: 24875294 PMCID: PMC4038465 DOI: 10.1371/journal.pgen.1004364] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 03/24/2014] [Indexed: 12/04/2022] Open
Abstract
Loss of PTPN11/SHP2 in mice or in human metachondromatosis (MC) patients causes benign cartilage tumors on the bone surface (exostoses) and within bones (enchondromas). To elucidate the mechanisms underlying cartilage tumor formation, we investigated the role of SHP2 in the specification, maturation and organization of chondrocytes. Firstly, we studied chondrocyte maturation by performing RNA-seq on primary chondrocyte pellet cultures. We found that SHP2 depletion, or inhibition of the ERK1/2 pathway, delays the terminal differentiation of chondrocytes from the early-hypertrophic to the late-hypertrophic stage. Secondly, we studied chondrocyte maturation and organization in mice with a mosaic postnatal inactivation of Ptpn11 in chondrocytes. We found that the vertebral growth plates of these mice have expanded domains of early-hypertrophic chondrocytes that have not yet terminally differentiated, and their enchondroma-like lesions arise from chondrocytes displaced from the growth plate due to a disruption in the organization of maturation and ossification zones. Furthermore, we observed that lesions from human MC patients also display disorganized chondrocyte maturation zones. Next, we found that inactivation of Ptpn11 in Fsp1-Cre-expressing fibroblasts induces exostosis-like outgrowths, suggesting that loss of SHP2 in cells on the bone surface and at bone-ligament attachment sites induces ectopic chondrogenesis. Finally, we performed lineage tracing to show that exostoses and enchondromas in mice likely contain mixtures of wild-type and SHP2-deficient chondrocytes. Together, these data indicate that in patients with MC, who are heterozygous for inherited PTPN11 loss-of-function mutations, second-hit mutations in PTPN11 can induce enchondromas by disrupting the organization and delaying the terminal differentiation of growth plate chondrocytes, and can induce exostoses by causing ectopic chondrogenesis of cells on the bone surface. Furthermore, the data are consistent with paracrine signaling from SHP2-deficient cells causing SHP2-sufficient cells to be incorporated into the lesions. Patients with the inherited disorder, metachondromatosis (MC), develop multiple benign cartilage tumors during childhood. MC patients carry heterozygous loss-of-function mutations in the PTPN11 gene, and their cartilage tumors likely arise when the second PTPN11 allele is lost due to a somatic mutation. PTPN11 encodes a phosphatase called SHP2 that is involved in a variety of signaling pathways. Here, we use mouse models and cell culture assays to investigate the mechanisms by which loss of SHP2 promotes cartilage tumor formation. We show that cartilage tumors that form inside bones (enchondromas) likely arise due to disorganized growth and delayed terminal differentiation of growth plate chondrocytes, while cartilage tumors that form on the bone surface (exostoses) can arise due to ectopic chondrogenesis of fibroblast-like cells that surround bones. We also suggest that paracrine signals from SHP2-deficient cells cause neighboring SHP2-sufficient cells to contribute to exostoses and enchondromas. Finally, we provide in vitro data that the ERK1/2 pathway is regulated by SHP2 and promotes chondrocyte terminal differentiation. Together, our data provide insight into the mechanisms underlying cartilage tumor formation and implicate SHP2 as a key regulator of chondrocyte specification, organization and maturation.
Collapse
Affiliation(s)
- Margot E. Bowen
- Orthopaedic Research Laboratories, Boston Children's Hospital, Boston, Massachusetts, United States of America
| | - Ugur M. Ayturk
- Orthopaedic Research Laboratories, Boston Children's Hospital, Boston, Massachusetts, United States of America
| | - Kyle C. Kurek
- Orthopaedic Research Laboratories, Boston Children's Hospital, Boston, Massachusetts, United States of America
| | - Wentian Yang
- Department of Orthopaedics, Brown University, Providence, Rhode Island, United States of America
| | - Matthew L. Warman
- Orthopaedic Research Laboratories, Boston Children's Hospital, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
2344
|
Two nested developmental waves demarcate a compartment boundary in the mouse lung. Nat Commun 2014; 5:3923. [PMID: 24879355 PMCID: PMC4115076 DOI: 10.1038/ncomms4923] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 04/19/2014] [Indexed: 01/05/2023] Open
Abstract
The lung is a branched tubular network with two distinct compartments — the proximal conducting airways and the peripheral gas exchange region — separated by a discrete boundary termed the bronchoalveolar duct junction (BADJ). Here we image the developing mouse lung in three dimensions and show that two nested developmental waves demarcate the BADJ under the control of a global hormonal signal. A first wave of branching morphogenesis progresses throughout embryonic development, generating branches for both compartments. A second wave of conducting airway differentiation follows the first wave but terminates earlier, specifying the proximal compartment and setting the BADJ. The second wave is terminated by a glucocorticoid signaling: premature activation or loss of glucocorticoid signaling causes a proximal or distal shift, respectively, in BADJ location. The results demonstrate a novel mechanism of boundary formation in complex, three-dimensional organs and provide new insights into glucocorticoid therapies for lung defects in premature birth.
Collapse
|
2345
|
Chivukula RR, Shi G, Acharya A, Mills EW, Zeitels LR, Anandam JL, Abdelnaby AA, Balch GC, Mansour JC, Yopp AC, Maitra A, Mendell JT. An essential mesenchymal function for miR-143/145 in intestinal epithelial regeneration. Cell 2014; 157:1104-16. [PMID: 24855947 PMCID: PMC4175516 DOI: 10.1016/j.cell.2014.03.055] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 01/24/2014] [Accepted: 03/10/2014] [Indexed: 12/15/2022]
Abstract
Downregulation of the miR-143/145 microRNA (miRNA) cluster has been repeatedly reported in colon cancer and other epithelial tumors. In addition, overexpression of these miRNAs inhibits tumorigenesis, leading to broad consensus that they function as cell-autonomous epithelial tumor suppressors. We generated mice with deletion of miR-143/145 to investigate the functions of these miRNAs in intestinal physiology and disease in vivo. Although intestinal development proceeded normally in the absence of these miRNAs, epithelial regeneration after injury was dramatically impaired. Surprisingly, we found that miR-143/145 are expressed and function exclusively within the mesenchymal compartment of intestine. Defective epithelial regeneration in miR-143/145-deficient mice resulted from the dysfunction of smooth muscle and myofibroblasts and was associated with derepression of the miR-143 target Igfbp5, which impaired IGF signaling after epithelial injury. These results provide important insights into the regulation of epithelial wound healing and argue against a cell-autonomous tumor suppressor role for miR-143/145 in colon cancer.
Collapse
Affiliation(s)
- Raghu R Chivukula
- Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Guanglu Shi
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Asha Acharya
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eric W Mills
- Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lauren R Zeitels
- Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Joselin L Anandam
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Abier A Abdelnaby
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Glen C Balch
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - John C Mansour
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Adam C Yopp
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anirban Maitra
- Sheikh Ahmed Bin Zayed Al Nahyan Center for Pancreatic Cancer Research, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joshua T Mendell
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA; Harold C. Simmons Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
2346
|
Turner CJ, Badu-Nkansah K, Crowley D, van der Flier A, Hynes RO. Integrin-α5β1 is not required for mural cell functions during development of blood vessels but is required for lymphatic-blood vessel separation and lymphovenous valve formation. Dev Biol 2014; 392:381-92. [PMID: 24858485 DOI: 10.1016/j.ydbio.2014.05.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 04/22/2014] [Accepted: 05/08/2014] [Indexed: 11/25/2022]
Abstract
Integrin α5β1 is essential for vascular development but it remains unclear precisely where and how it functions. Here, we report that deletion of the gene encoding the integrin-α5 subunit (Itga5) using the Pdgfrb-Cre transgenic mouse line, leads to oedema, haemorrhage and increased levels of embryonic lethality. Unexpectedly, these defects were not caused by loss of α5 from Pdgfrb-Cre expressing mural cells (pericytes and vascular smooth muscle cells), which wrap around the endothelium and stabilise blood vessels, nor by defects in the heart or great vessels, but were due to abnormal development of the lymphatic vasculature. Reminiscent of the pathologies seen in the human lymphatic malformation, fetal cystic hygroma, α5 mutants display defects both in the separation of their blood and lymphatic vasculature and in the formation of the lymphovenous valves. As a consequence, α5-deficient mice develop dilated, blood-filled lymphatic vessels and lymphatic capillaries that are ectopically covered with smooth muscle cells. Analysis of the expression of Pdgfrb during lymphatic development suggests that these defects probably arise from loss of α5β1 integrin in subsets of specialised Prox1(+)Pdgfrb(+) venous endothelial cells that are essential for the separation of the jugular lymph sac from the cardinal vein and formation of the lymphovenous valve leaflets.
Collapse
Affiliation(s)
- Christopher J Turner
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kwabena Badu-Nkansah
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Denise Crowley
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arjan van der Flier
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Richard O Hynes
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
2347
|
McBride SH, McKenzie JA, Bedrick BS, Kuhlmann P, Pasteris JD, Rosen V, Silva MJ. Long bone structure and strength depend on BMP2 from osteoblasts and osteocytes, but not vascular endothelial cells. PLoS One 2014; 9:e96862. [PMID: 24837969 PMCID: PMC4024030 DOI: 10.1371/journal.pone.0096862] [Citation(s) in RCA: 25] [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: 12/06/2013] [Accepted: 04/13/2014] [Indexed: 11/19/2022] Open
Abstract
The importance of bone morphogenetic protein 2 (BMP2) in the skeleton is well known. BMP2 is expressed in a variety of tissues during development, growth and healing. In this study we sought to better identify the role of tissue-specific BMP2 during post-natal growth and to determine if BMP2 knockout affects the ability of terminally differentiated cells to create high quality bone material. We targeted BMP2 knockout to two differentiated cell types known to express BMP2 during growth and healing, early-stage osteoblasts and their progeny (osterix promoted Cre) and vascular endothelial cells (vascular-endothelial-cadherin promoted Cre). Our objectives were to assess post-natal bone growth, structure and strength. We hypothesized that removal of BMP2 from osteogenic and vascular cells (separately) would result in smaller skeletons with inferior bone material properties. At 12 and 24 weeks of age the osteoblast knockout of BMP2 reduced body weight by 20%, but the vascular knockout had no effect. Analysis of bone in the tibia revealed reductions in cortical and cancellous bone size and volume in the osteoblast knockout, but not in the vascular endothelial knockout. Furthermore, forelimb strength testing revealed a 30% reduction in ultimate force at both 12 and 24 weeks in the osteoblast knockout of BMP2, but no change in the vascular endothelial knockout. Moreover, mechanical strength testing of femurs from osteoblast knockout mice demonstrated an increased Young's modulus (greater than 35%) but decreased post-yield displacement (greater than 50%) at both 12 and 24 weeks of age. In summary, the osteoblast knockout of BMP2 reduced bone size and altered mechanical properties at the whole-bone and material levels. Osteoblast-derived BMP2 has an important role in post-natal skeletal growth, structure and strength, while vascular endothelial-derived BMP2 does not.
Collapse
Affiliation(s)
- Sarah H. McBride
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University in St. Louis, St. Louis, Missouri, United States of America
- Department of Orthopaedic Surgery, Saint Louis University, St. Louis, Missouri, United States of America
| | - Jennifer A. McKenzie
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Bronwyn S. Bedrick
- Department of Earth and Planetary Sciences, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Paige Kuhlmann
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Jill D. Pasteris
- Department of Earth and Planetary Sciences, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts, United States of America
| | - Matthew J. Silva
- Department of Orthopaedic Surgery, Musculoskeletal Research Center, Washington University in St. Louis, St. Louis, Missouri, United States of America
| |
Collapse
|
2348
|
Zhang H, von Gise A, Liu Q, Hu T, Tian X, He L, Pu W, Huang X, He L, Cai CL, Camargo FD, Pu WT, Zhou B. Yap1 is required for endothelial to mesenchymal transition of the atrioventricular cushion. J Biol Chem 2014; 289:18681-92. [PMID: 24831012 DOI: 10.1074/jbc.m114.554584] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cardiac malformations due to aberrant development of the atrioventricular (AV) valves are among the most common forms of congenital heart diseases. Normally, heart valve mesenchyme is formed from an endothelial to mesenchymal transition (EMT) of endothelial cells of the endocardial cushions. Yes-associated protein 1 (YAP1) has been reported to regulate EMT in vitro, in addition to its known role as a major regulator of organ size and cell proliferation in vertebrates, leading us to hypothesize that YAP1 is required for heart valve development. We tested this hypothesis by conditional inactivation of YAP1 in endothelial cells and their derivatives. This resulted in markedly hypocellular endocardial cushions due to impaired formation of heart valve mesenchyme by EMT and to reduced endocardial cell proliferation. In endothelial cells, TGFβ induces nuclear localization of Smad2/3/4 complex, which activates expression of Snail, Twist1, and Slug, key transcription factors required for EMT. YAP1 interacts with this complex, and loss of YAP1 disrupts TGFβ-induced up-regulation of Snail, Twist1, and Slug. Together, our results identify a role of YAP1 in regulating EMT through modulation of TGFβ-Smad signaling and through proliferative activity during cardiac cushion development.
Collapse
Affiliation(s)
- Hui Zhang
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Alexander von Gise
- the Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts 02115, the Department of Pediatric Cardiology and Intensive Care, MHH-Hannover Medical School, 30669 Hannover, Germany
| | - Qiaozhen Liu
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tianyuan Hu
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xueying Tian
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lingjuan He
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenjuan Pu
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiuzhen Huang
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liang He
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chen-Leng Cai
- the Department of Developmental and Regenerative Biology, Center for Molecular Cardiology of the Child Health and Development Institute, the Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, New York 10029
| | - Fernando D Camargo
- the Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, and
| | - William T Pu
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bin Zhou
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China,
| |
Collapse
|
2349
|
Rinkevich Y, Montoro DT, Contreras-Trujillo H, Harari-Steinberg O, Newman AM, Tsai JM, Lim X, Van-Amerongen R, Bowman A, Januszyk M, Pleniceanu O, Nusse R, Longaker MT, Weissman IL, Dekel B. In vivo clonal analysis reveals lineage-restricted progenitor characteristics in mammalian kidney development, maintenance, and regeneration. Cell Rep 2014; 7:1270-83. [PMID: 24835991 DOI: 10.1016/j.celrep.2014.04.018] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Revised: 03/02/2014] [Accepted: 04/09/2014] [Indexed: 12/18/2022] Open
Abstract
The mechanism and magnitude by which the mammalian kidney generates and maintains its proximal tubules, distal tubules, and collecting ducts remain controversial. Here, we use long-term in vivo genetic lineage tracing and clonal analysis of individual cells from kidneys undergoing development, maintenance, and regeneration. We show that the adult mammalian kidney undergoes continuous tubulogenesis via expansions of fate-restricted clones. Kidneys recovering from damage undergo tubulogenesis through expansions of clones with segment-specific borders, and renal spheres developing in vitro from individual cells maintain distinct, segment-specific fates. Analysis of mice derived by transfer of color-marked embryonic stem cells (ESCs) into uncolored blastocysts demonstrates that nephrons are polyclonal, developing from expansions of singly fated clones. Finally, we show that adult renal clones are derived from Wnt-responsive precursors, and their tracing in vivo generates tubules that are segment specific. Collectively, these analyses demonstrate that fate-restricted precursors functioning as unipotent progenitors continuously maintain and self-preserve the mouse kidney throughout life.
Collapse
Affiliation(s)
- Yuval Rinkevich
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Daniel T Montoro
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Humberto Contreras-Trujillo
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Orit Harari-Steinberg
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv 52621, Israel
| | - Aaron M Newman
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jonathan M Tsai
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xinhong Lim
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Renee Van-Amerongen
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Angela Bowman
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael Januszyk
- Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Oren Pleniceanu
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv 52621, Israel
| | - Roel Nusse
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Developmental Biology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael T Longaker
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Hagey Laboratory for Pediatric Regenerative Medicine, Department of Surgery, Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Irving L Weissman
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Ludwig Center for Cancer Stem Cell Research, Stanford University, Stanford, CA 94305, USA
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv 52621, Israel.
| |
Collapse
|
2350
|
McAllister F, Bailey JM, Alsina J, Nirschl CJ, Sharma R, Fan H, Rattigan Y, Roeser JC, Lankapalli RH, Zhang H, Jaffee EM, Drake CG, Housseau F, Maitra A, Kolls JK, Sears CL, Pardoll DM, Leach SD. Oncogenic Kras activates a hematopoietic-to-epithelial IL-17 signaling axis in preinvasive pancreatic neoplasia. Cancer Cell 2014; 25:621-37. [PMID: 24823639 PMCID: PMC4072043 DOI: 10.1016/j.ccr.2014.03.014] [Citation(s) in RCA: 334] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 01/12/2014] [Accepted: 03/12/2014] [Indexed: 02/06/2023]
Abstract
Many human cancers are dramatically accelerated by chronic inflammation. However, the specific cellular and molecular elements mediating this effect remain largely unknown. Using a murine model of pancreatic intraepithelial neoplasia (PanIN), we found that Kras(G12D) induces expression of functional IL-17 receptors on PanIN epithelial cells and also stimulates infiltration of the pancreatic stroma by IL-17-producing immune cells. Both effects are augmented by associated chronic pancreatitis, resulting in functional in vivo changes in PanIN epithelial gene expression. Forced IL-17 overexpression dramatically accelerates PanIN initiation and progression, while inhibition of IL-17 signaling using genetic or pharmacologic techniques effectively prevents PanIN formation. Together, these studies suggest that a hematopoietic-to-epithelial IL-17 signaling axis is a potent and requisite driver of PanIN formation.
Collapse
MESH Headings
- Animals
- Carcinoma in Situ/genetics
- Carcinoma in Situ/metabolism
- Carcinoma in Situ/prevention & control
- Cell Transformation, Neoplastic
- Chemoprevention
- Epithelial Cells/metabolism
- Hematopoietic System/cytology
- Hematopoietic System/metabolism
- Humans
- Inflammation
- Interleukin-17/antagonists & inhibitors
- Interleukin-17/genetics
- Interleukin-17/metabolism
- Mice
- Mice, Transgenic
- Pancreas/pathology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/prevention & control
- Proto-Oncogene Proteins p21(ras)/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/biosynthesis
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Interleukin-17/biosynthesis
- Receptors, Interleukin-17/metabolism
- Signal Transduction/genetics
- Th17 Cells/immunology
Collapse
Affiliation(s)
- Florencia McAllister
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA; Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jennifer M Bailey
- Department of Surgery and McKusick Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Janivette Alsina
- Department of Surgery and McKusick Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Rajni Sharma
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Hongni Fan
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yanique Rattigan
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jeffrey C Roeser
- Department of Surgery and McKusick Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Hao Zhang
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Elizabeth M Jaffee
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Charles G Drake
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Franck Housseau
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Anirban Maitra
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jay K Kolls
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Cynthia L Sears
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Steven D Leach
- Department of Surgery and McKusick Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|