51
|
Understanding paraxial mesoderm development and sclerotome specification for skeletal repair. Exp Mol Med 2020; 52:1166-1177. [PMID: 32788657 PMCID: PMC8080658 DOI: 10.1038/s12276-020-0482-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/26/2022] Open
Abstract
Pluripotent stem cells (PSCs) are attractive regenerative therapy tools for skeletal tissues. However, a deep understanding of skeletal development is required in order to model this development with PSCs, and for the application of PSCs in clinical settings. Skeletal tissues originate from three types of cell populations: the paraxial mesoderm, lateral plate mesoderm, and neural crest. The paraxial mesoderm gives rise to the sclerotome mainly through somitogenesis. In this process, key developmental processes, including initiation of the segmentation clock, formation of the determination front, and the mesenchymal–epithelial transition, are sequentially coordinated. The sclerotome further forms vertebral columns and contributes to various other tissues, such as tendons, vessels (including the dorsal aorta), and even meninges. To understand the molecular mechanisms underlying these developmental processes, extensive studies have been conducted. These studies have demonstrated that a gradient of activities involving multiple signaling pathways specify the embryonic axis and induce cell-type-specific master transcription factors in a spatiotemporal manner. Moreover, applying the knowledge of mesoderm development, researchers have attempted to recapitulate the in vivo development processes in in vitro settings, using mouse and human PSCs. In this review, we summarize the state-of-the-art understanding of mesoderm development and in vitro modeling of mesoderm development using PSCs. We also discuss future perspectives on the use of PSCs to generate skeletal tissues for basic research and clinical applications. A deeper understanding of skeletal tissue development and improvements in tissue engineering will help pluripotent stem cell (PSC) therapies to reach their full potential for skeletal repair. The paraxial mesoderm, an embryonic germ layer, is crucial to the formation of healthy axial skeleton. Shoichiro Tani at the University of Tokyo, Japan, and co-workers reviewed current understanding of paraxial mesoderm development and studies involving in vitro PSC skeletal modeling. The formation of the paraxial mesoderm and associated connective tissues comprises multiple stages, and studies in vertebrate embryos have uncovered critical signaling pathways and cellular components important to PSC modeling. Although many individual cellular components can now be modeled, it remains challenging to recreate three-dimensional skeletal tissues. Such an achievement would facilitate a functioning model of bone metabolism, the next step in achieving skeletal regeneration.
Collapse
|
52
|
Norden PR, Sabine A, Wang Y, Demir CS, Liu T, Petrova TV, Kume T. Shear stimulation of FOXC1 and FOXC2 differentially regulates cytoskeletal activity during lymphatic valve maturation. eLife 2020; 9:53814. [PMID: 32510325 PMCID: PMC7302880 DOI: 10.7554/elife.53814] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 06/06/2020] [Indexed: 12/14/2022] Open
Abstract
Mutations in the transcription factor FOXC2 are predominately associated with lymphedema. Herein, we demonstrate a key role for related factor FOXC1, in addition to FOXC2, in regulating cytoskeletal activity in lymphatic valves. FOXC1 is induced by laminar, but not oscillatory, shear and inducible, endothelial-specific deletion impaired postnatal lymphatic valve maturation in mice. However, deletion of Foxc2 induced valve degeneration, which is exacerbated in Foxc1; Foxc2 mutants. FOXC1 knockdown (KD) in human lymphatic endothelial cells increased focal adhesions and actin stress fibers whereas FOXC2-KD increased focal adherens and disrupted cell junctions, mediated by increased ROCK activation. ROCK inhibition rescued cytoskeletal or junctional integrity changes induced by inactivation of FOXC1 and FOXC2 invitro and vivo respectively, but only ameliorated valve degeneration in Foxc2 mutants. These results identify both FOXC1 and FOXC2 as mediators of mechanotransduction in the postnatal lymphatic vasculature and posit cytoskeletal signaling as a therapeutic target in lymphatic pathologies.
Collapse
Affiliation(s)
- Pieter R Norden
- Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Amélie Sabine
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Epalinges, Switzerland
| | - Ying Wang
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, United States
| | - Cansaran Saygili Demir
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Epalinges, Switzerland
| | - Ting Liu
- Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, United States
| | - Tatiana V Petrova
- Department of Oncology, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Epalinges, Switzerland
| | - Tsutomu Kume
- Feinberg Cardiovascular and Renal Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, United States
| |
Collapse
|
53
|
Sanchez J, Miyake R, Cheng A, Liu T, Iseki S, Kume T. Conditional inactivation of Foxc1 and Foxc2 in neural crest cells leads to cardiac abnormalities. Genesis 2020; 58:e23364. [PMID: 32259372 DOI: 10.1002/dvg.23364] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 02/06/2023]
Abstract
Cardiac neural crest cells (cNCCs) are required for normal heart development. cNCCs are a multipotent and migratory cell lineage that differentiates into multiple cell types. cNCCs migrate into the developing heart to contribute to the septation of the cardiac outflow tract (OFT). Foxc1 and Foxc2 are closely related members of the FOX (Forkhead box) transcription factor family and are expressed in cNCC during heart development. However, the precise role of Foxc1 and Foxc2 in cNCCs has yet to be fully described. We found that compound NCC-specific Foxc1;Foxc2 mutant embryos exhibited persistent truncus arteriosus (PTA), ventricular septal defects (VSDs), and thinning of the ventricular myocardium. Loss of Foxc1/c2 expression in cNCCs resulted in abnormal patterns of cNCC migration into the OFT without the formation of the aorticopulmonary septum. Further, loss of Foxc1 expression in cNCCs resulted in normal OFT development but abnormal ventricular septal formation. In contrast, loss of Foxc2 expression in NCCs led to no obvious cardiac abnormalities. Together, we provide evidence that Foxc1 and Foxc2 in cNCCs are cooperatively required for proper cNCC migration, the formation of the OFT septation, and the development of the ventricles. Our data also suggests that Foxc1 expression may play a larger role in ventricular development compared to Foxc2.
Collapse
Affiliation(s)
- Joshua Sanchez
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Department of Medicine, Northwestern University, Chicago, Illinois
| | - Risa Miyake
- Section of Molecular Craniofacial Embryology, Tokyo Medical and Dental University Graduate School of Medical and Dental Sciences, Tokyo, Japan
| | - Andrew Cheng
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Department of Medicine, Northwestern University, Chicago, Illinois
| | - Ting Liu
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Department of Medicine, Northwestern University, Chicago, Illinois
| | - Sachiko Iseki
- Section of Molecular Craniofacial Embryology, Tokyo Medical and Dental University Graduate School of Medical and Dental Sciences, Tokyo, Japan
| | - Tsutomu Kume
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Department of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
54
|
Sissaoui S, Yu J, Yan A, Li R, Yukselen O, Kucukural A, Zhu LJ, Lawson ND. Genomic Characterization of Endothelial Enhancers Reveals a Multifunctional Role for NR2F2 in Regulation of Arteriovenous Gene Expression. Circ Res 2020; 126:875-888. [PMID: 32065070 PMCID: PMC7212523 DOI: 10.1161/circresaha.119.316075] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 02/14/2020] [Indexed: 02/07/2023]
Abstract
RATIONALE Significant progress has revealed transcriptional inputs that underlie regulation of artery and vein endothelial cell fates. However, little is known concerning genome-wide regulation of this process. Therefore, such studies are warranted to address this gap. OBJECTIVE To identify and characterize artery- and vein-specific endothelial enhancers in the human genome, thereby gaining insights into mechanisms by which blood vessel identity is regulated. METHODS AND RESULTS Using chromatin immunoprecipitation and deep sequencing for markers of active chromatin in human arterial and venous endothelial cells, we identified several thousand artery- and vein-specific regulatory elements. Computational analysis revealed that NR2F2 (nuclear receptor subfamily 2, group F, member 2) sites were overrepresented in vein-specific enhancers, suggesting a direct role in promoting vein identity. Subsequent integration of chromatin immunoprecipitation and deep sequencing data sets with RNA sequencing revealed that NR2F2 regulated 3 distinct aspects related to arteriovenous identity. First, consistent with previous genetic observations, NR2F2 directly activated enhancer elements flanking cell cycle genes to drive their expression. Second, NR2F2 was essential to directly activate vein-specific enhancers and their associated genes. Our genomic approach further revealed that NR2F2 acts with ERG (ETS-related gene) at many of these sites to drive vein-specific gene expression. Finally, NR2F2 directly repressed only a small number of artery enhancers in venous cells to prevent their activation, including a distal element upstream of the artery-specific transcription factor, HEY2 (hes related family bHLH transcription factor with YRPW motif 2). In arterial endothelial cells, this enhancer was normally bound by ERG, which was also required for arterial HEY2 expression. By contrast, in venous endothelial cells, NR2F2 was bound to this site, together with ERG, and prevented its activation. CONCLUSIONS By leveraging a genome-wide approach, we revealed mechanistic insights into how NR2F2 functions in multiple roles to maintain venous identity. Importantly, characterization of its role at a crucial artery enhancer upstream of HEY2 established a novel mechanism by which artery-specific expression can be achieved.
Collapse
Affiliation(s)
- Samir Sissaoui
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605
| | - Jun Yu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605
| | - Aimin Yan
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605
| | - Rui Li
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605
| | - Onur Yukselen
- Department of Bioinformatics Core, University of Massachusetts Medical School, Worcester, MA, 01605
| | - Alper Kucukural
- Department of Bioinformatics Core, University of Massachusetts Medical School, Worcester, MA, 01605
- Department of Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605
- Department of Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605
- Department of Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, 01605
| | - Nathan D. Lawson
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605
| |
Collapse
|
55
|
Naito H, Iba T, Takakura N. Mechanisms of new blood-vessel formation and proliferative heterogeneity of endothelial cells. Int Immunol 2020; 32:295-305. [DOI: 10.1093/intimm/dxaa008] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/27/2020] [Indexed: 12/26/2022] Open
Abstract
Abstract
The vast blood-vessel network of the circulatory system is crucial for maintaining bodily homeostasis, delivering essential molecules and blood cells, and removing waste products. Blood-vessel dysfunction and dysregulation of new blood-vessel formation are related to the onset and progression of many diseases including cancer, ischemic disease, inflammation and immune disorders. Endothelial cells (ECs) are fundamental components of blood vessels and their proliferation is essential for new vessel formation, making them good therapeutic targets for regulating the latter. New blood-vessel formation occurs by vasculogenesis and angiogenesis during development. Induction of ECs termed tip, stalk and phalanx cells by interactions between vascular endothelial growth factor A (VEGF-A) and its receptors (VEGFR1–3) and between Notch and Delta-like Notch ligands (DLLs) is crucial for regulation of angiogenesis. Although the importance of angiogenesis is unequivocal in the adult, vasculogenesis effected by endothelial progenitor cells (EPCs) may also contribute to post-natal vessel formation. However, the definition of these cells is ambiguous and they include several distinct cell types under the simple classification of ‘EPC’. Furthermore, recent evidence indicates that ECs within the intima show clonal expansion in some situations and that they may harbor vascular-resident endothelial stem cells. In this article, we summarize recent knowledge on vascular development and new blood-vessel formation in the adult. We also introduce concepts of EC heterogeneity and EC clonal expansion, referring to our own recent findings.
Collapse
Affiliation(s)
- Hisamichi Naito
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Tomohiro Iba
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Nobuyuki Takakura
- Department of Signal Transduction, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Laboratory of Signal Transduction, World Premier Institute Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
56
|
Childhood glaucoma genes and phenotypes: Focus on FOXC1 mutations causing anterior segment dysgenesis and hearing loss. Exp Eye Res 2019; 190:107893. [PMID: 31836490 DOI: 10.1016/j.exer.2019.107893] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/16/2019] [Accepted: 12/04/2019] [Indexed: 12/27/2022]
Abstract
Childhood glaucoma is an important cause of blindness world-wide. Eleven genes are currently known to cause inherited forms of glaucoma with onset before age 20. While all the early-onset glaucoma genes cause severe disease, considerable phenotypic variability is observed among mutations carriers. In particular, FOXC1 genetic variants are associated with a broad range of phenotypes including multiple forms of glaucoma and also systemic abnormalities, especially hearing loss. FOXC1 is a member of the forkhead family of transcription factors and is involved in neural crest development necessary for formation of anterior eye structures and also pharyngeal arches that form the middle ear bones. In this study we review the clinical phenotypes reported for known FOXC1 mutations and show that mutations in patients with reported ocular anterior segment abnormalities and hearing loss primarily disrupt the critically important forkhead domain. These results suggest that optimal care for patients affected with anterior segment dysgenesis should include screening for FOXC1 mutations and also testing for hearing loss.
Collapse
|
57
|
Zhang SP, Yang RH, Shang J, Gao T, Wang R, Peng XD, Miao X, Pan L, Yuan WJ, Lin L, Hu QK. FOXC1 up-regulates the expression of toll-like receptors in myocardial ischaemia. J Cell Mol Med 2019; 23:7566-7580. [PMID: 31517441 PMCID: PMC6815849 DOI: 10.1111/jcmm.14626] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 05/20/2019] [Accepted: 08/06/2019] [Indexed: 01/01/2023] Open
Abstract
Myocardial ischaemia (MI) remains a major cause of death and disability worldwide. Accumulating evidence suggests a significant role for innate immunity, in which the family of toll‐like receptors (TLRs) acts as an essential player. We previously reported and reviewed the changes of Tlr expression in models of MI. However, the underlying mechanisms regulating Tlr expression in MI remain unclear. The present study first screened transcription factors (TFs) that potentially regulate Tlr gene transcription based on in silico analyses followed by experimental verification, using both in vivo and in vitro models. Forkhead box C1 (FOXC1) was identified as a putative TF, which was highly responsive to MI. Next, by focusing on two representative TLR subtypes, an intracellular subtype TLR3 and a cell‐surface subtype TLR4, the regulation of FOXC1 on Tlr expression was investigated. The overexpression or knockdown of FoxC1 was observed to up‐ or down‐regulate Tlr3/4 mRNA and protein levels, respectively. A dual‐luciferase assay showed that FOXC1 trans‐activated Tlr3/4 promoter, and a ChIP assay showed direct binding of FOXC1 to Tlr3/4 promoter. Last, a functional study of FOXC1 was performed, which revealed the pro‐inflammatory effects of FOXC1 and its destructive effects on infarct size and heart function in a mouse model of MI. The present study for the first time identified FOXC1 as a novel regulator of Tlr expression and described its function in MI.
Collapse
Affiliation(s)
- Shao-Ping Zhang
- Department of Physiology, Institute of Basic Medicine, Ningxia Medical University, Yinchuan, China.,Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ruo-Han Yang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China.,Department of Pharmacy, First People's Hospital, Guangyuan, China
| | - Jia Shang
- Department of Physiology, Institute of Basic Medicine, Ningxia Medical University, Yinchuan, China.,Department of Physiology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Ting Gao
- Department of Physiology, Institute of Basic Medicine, Ningxia Medical University, Yinchuan, China.,Department of Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Wang
- Department of Physiology, Institute of Basic Medicine, Ningxia Medical University, Yinchuan, China.,Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Xiao-Dong Peng
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Xiao Miao
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Pan
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wen-Jun Yuan
- Department of Physiology, Institute of Basic Medicine, Ningxia Medical University, Yinchuan, China.,Department of Physiology, Second Military Medical University, Shanghai, China
| | - Li Lin
- Department of Physiology, Institute of Basic Medicine, Ningxia Medical University, Yinchuan, China.,Department of Physiology, Second Military Medical University, Shanghai, China.,Key Laboratory of Arrhythmias of the Ministry of Education of China, Tongji University, Shanghai, China
| | - Qi-Kuan Hu
- Department of Physiology, Institute of Basic Medicine, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
58
|
Whitesell TR, Chrystal PW, Ryu JR, Munsie N, Grosse A, French CR, Workentine ML, Li R, Zhu LJ, Waskiewicz A, Lehmann OJ, Lawson ND, Childs SJ. foxc1 is required for embryonic head vascular smooth muscle differentiation in zebrafish. Dev Biol 2019; 453:34-47. [PMID: 31199900 DOI: 10.1016/j.ydbio.2019.06.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/29/2019] [Accepted: 06/09/2019] [Indexed: 11/15/2022]
Abstract
Vascular smooth muscle of the head derives from neural crest, but developmental mechanisms and early transcriptional drivers of the vSMC lineage are not well characterized. We find that in early development, the transcription factor foxc1b is expressed in mesenchymal cells that associate with the vascular endothelium. Using timelapse imaging, we observe that foxc1b expressing mesenchymal cells differentiate into acta2 expressing vascular mural cells. We show that in zebrafish, while foxc1b is co-expressed in acta2 positive smooth muscle cells that associate with large diameter vessels, it is not co-expressed in capillaries where pdgfrβ positive pericytes are located. In addition to being an early marker of the lineage, foxc1 is essential for vSMC differentiation; we find that foxc1 loss of function mutants have defective vSMC differentiation and that early genetic ablation of foxc1b or acta2 expressing populations blocks vSMC differentiation. Furthermore, foxc1 is expressed upstream of acta2 and is required for acta2 expression in vSMCs. Using RNA-Seq we determine an enriched intersectional gene expression profile using dual expression of foxc1b and acta2 to identify novel vSMC markers. Taken together, our data suggests that foxc1 is a marker of vSMCs and plays a critical functional role in promoting their differentiation.
Collapse
Affiliation(s)
- Thomas R Whitesell
- Alberta Children's Hospital Research Institute, University of Calgary, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N 4N1
| | - Paul W Chrystal
- Departments of Ophthalmology, and Medical Genetics, University of Alberta, Edmonton, Alberta, Canada; Department of Biological Sciences, CW405, Biological Sciences Bldg., 11455, Saskatchewan Dr., University of Alberta, Edmonton, AB, T6G 2E9, Canada; Women & Children's Health Research Institute, ECHA 4-081, 11405 87, Ave NW, University of Alberta, Edmonton, AB, T6G 1C9, Canada; Neurosciences and Mental Health Institute, 4-120 Katz Group Centre, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Jae-Ryeon Ryu
- Alberta Children's Hospital Research Institute, University of Calgary, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N 4N1
| | - Nicole Munsie
- Alberta Children's Hospital Research Institute, University of Calgary, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N 4N1
| | - Ann Grosse
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, USA, 01605
| | - Curtis R French
- Department of Biological Sciences, CW405, Biological Sciences Bldg., 11455, Saskatchewan Dr., University of Alberta, Edmonton, AB, T6G 2E9, Canada; Women & Children's Health Research Institute, ECHA 4-081, 11405 87, Ave NW, University of Alberta, Edmonton, AB, T6G 1C9, Canada; Neurosciences and Mental Health Institute, 4-120 Katz Group Centre, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Matthew L Workentine
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N 4N1
| | - Rui Li
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, USA, 01605
| | - Lihua Julie Zhu
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, USA, 01605; Program in Bioinformatics and Integrative Biology, Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA, 01605
| | - Andrew Waskiewicz
- Department of Biological Sciences, CW405, Biological Sciences Bldg., 11455, Saskatchewan Dr., University of Alberta, Edmonton, AB, T6G 2E9, Canada; Women & Children's Health Research Institute, ECHA 4-081, 11405 87, Ave NW, University of Alberta, Edmonton, AB, T6G 1C9, Canada; Neurosciences and Mental Health Institute, 4-120 Katz Group Centre, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Ordan J Lehmann
- Departments of Ophthalmology, and Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Nathan D Lawson
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, USA, 01605
| | - Sarah J Childs
- Alberta Children's Hospital Research Institute, University of Calgary, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N 4N1.
| |
Collapse
|
59
|
Della Gaspera B, Mateus A, Andéol Y, Weill L, Charbonnier F, Chanoine C. Lineage tracing of sclerotome cells in amphibian reveals that multipotent somitic cells originate from lateral somitic frontier. Dev Biol 2019; 453:11-18. [PMID: 31128088 DOI: 10.1016/j.ydbio.2019.05.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/18/2019] [Accepted: 05/21/2019] [Indexed: 11/29/2022]
Abstract
The two somite compartments, dorso-lateral dermomyotome and medio-ventral sclerotome are major vertebrate novelties, but little is known about their evolutionary origin. We determined that sclerotome cells in Xenopus come from lateral somitic frontier (LSF) by lineage tracing, ablation experiments and histological analysis. We identified Twist1 as marker of migrating sclerotome progenitors in two amphibians, Xenopus and axolotl. From these results, three conclusions can be drawn. First, LSF is made up of multipotent somitic cells (MSCs) since LSF gives rise to sclerotome but also to dermomytome as already shown in Xenopus. Second, the basic scheme of somite compartmentalization is conserved from cephalochordates to anamniotes since in both cases, lateral cells envelop dorsally and ventrally the ancestral myotome, suggesting that lateral MSCs should already exist in cephalochordates. Third, the transition from anamniote to amniote vertebrates is characterized by extension of the MSCs domain to the entire somite at the expense of ancestral myotome since amniote somite is a naive tissue that subdivides into sclerotome and dermomyotome. Like neural crest pluripotent cells, MSCs are at the origin of major vertebrate novelties, namely hypaxial region of the somite, dermomyotome and sclerotome compartments. Hence, change in MSCs properties and location is involved in somite evolution.
Collapse
Affiliation(s)
- Bruno Della Gaspera
- UMR INSERM 1124, Université de Paris, Faculté des sciences biomédicales et fondamentales, 45 rue des Saints-Pères, F-75270, Paris Cedex 06, France.
| | - Alice Mateus
- UMR INSERM 1124, Université de Paris, Faculté des sciences biomédicales et fondamentales, 45 rue des Saints-Pères, F-75270, Paris Cedex 06, France
| | - Yannick Andéol
- Equipe UR6, Enzymologie de l'ARN, Sorbonne Université, Faculté des Sciences et Technologies, 9 quai St Bernard, 75251, Paris Cedex 05, France
| | - Laure Weill
- UMR INSERM 1124, Université de Paris, Faculté des sciences biomédicales et fondamentales, 45 rue des Saints-Pères, F-75270, Paris Cedex 06, France
| | - Frédéric Charbonnier
- UMR INSERM 1124, Université de Paris, Faculté des sciences biomédicales et fondamentales, 45 rue des Saints-Pères, F-75270, Paris Cedex 06, France
| | - Christophe Chanoine
- UMR INSERM 1124, Université de Paris, Faculté des sciences biomédicales et fondamentales, 45 rue des Saints-Pères, F-75270, Paris Cedex 06, France.
| |
Collapse
|
60
|
Chen X, Wei H, Li J, Liang X, Dai S, Jiang L, Guo M, Qu L, Chen Z, Chen L, Chen Y. Structural basis for DNA recognition by FOXC2. Nucleic Acids Res 2019; 47:3752-3764. [PMID: 30722065 PMCID: PMC6468292 DOI: 10.1093/nar/gkz077] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 01/26/2019] [Accepted: 01/30/2019] [Indexed: 12/15/2022] Open
Abstract
The FOXC family of transcription factors (FOXC1 and FOXC2) plays essential roles in the regulation of embryonic, ocular, and cardiac development. Mutations and abnormal expression of FOXC proteins are implicated in genetic diseases as well as cancer. In this study, we determined two crystal structures of the DNA-binding domain (DBD) of human FOXC2 protein, in complex with different DNA sites. The FOXC2-DBD adopts the winged-helix fold with helix H3 contributing to all the base specific contacts, while the N-terminus, wing 1, and the C-terminus of FOXC2-DBD all make additional contacts with the phosphate groups of DNA. Our structural, biochemical, and bioinformatics analyses allow us to revise the previously proposed DNA recognition mechanism and provide a model of DNA binding for the FOXC proteins. In addition, our structural analysis and accompanying biochemical assays provide a molecular basis for understanding disease-causing mutations in FOXC1 and FOXC2.
Collapse
Affiliation(s)
- Xiaojuan Chen
- NHC Key Laboratory of Cancer Proteomics and Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Key Laboratory of Medical Genetics and College of Life Science, Central South University, Changsha, Hunan 410008, China
| | - Hudie Wei
- NHC Key Laboratory of Cancer Proteomics and Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jun Li
- NHC Key Laboratory of Cancer Proteomics and Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xujun Liang
- NHC Key Laboratory of Cancer Proteomics and Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shuyan Dai
- NHC Key Laboratory of Cancer Proteomics and Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Longying Jiang
- NHC Key Laboratory of Cancer Proteomics and Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ming Guo
- NHC Key Laboratory of Cancer Proteomics and Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Lingzhi Qu
- NHC Key Laboratory of Cancer Proteomics and Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhuchu Chen
- NHC Key Laboratory of Cancer Proteomics and Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Lin Chen
- Molecular and Computational Biology Program, Department of Biological Sciences and Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA
| | - Yongheng Chen
- NHC Key Laboratory of Cancer Proteomics and Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Key Laboratory of Medical Genetics and College of Life Science, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
61
|
Shen H, Jin M, Gu S, Wu Y, Yang M, Hua X. CD97 Is Decreased in Preeclamptic Placentas and Promotes Human Trophoblast Invasion Through PI3K/Akt/mTOR Signaling Pathway. Reprod Sci 2019:1933719119828067. [PMID: 30791863 DOI: 10.1177/1933719119828067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Preeclampsia (PE) is a pregnancy disorder leading to the morbidity and mortality. Despite the development of the understanding of etiology, the only effective treatment of PE is the delivery of the placenta. An improved mastery on the regulation of trophoblast invasion could be meaningful to alleviate the disease burden of PE. Relative expression of CD97 in PE and normal placental tissues was evaluated by quantitative real-time polymerase chain reaction, immunohistology, and Western blot. The CD97 siRNA and expression vector was transfected to cultured human trophoblast HTR-8/SVneo, and the cell invasion as well as the protein expression in PI3K/Akt/mTOR signaling pathway were evaluated. Expression of CD97 is significantly downregulated in PE placental tissues compared to normal controls. The Si-CD97 inhibits HTR-8/SVneo trophoblast cells invasion, as well as the activation of PI3K/Akt/mTOR signaling pathway. In accordance, overexpression of CD97 promotes trophoblast cell invasion. In addition, CD97 regulates FOXC2 expression and showed similar effects on PI3K/Akt/mTOR signaling pathway as specific FOXC2 inhibitor. In short, this study demonstrated the downregulation of CD97 expression in preeclamptic placentas. Further mechanism investigation revealed that CD97 promoted trophoblast invasion by targeting FOXC2 via PI3K/Akt/mTOR signaling pathway, laying the foundation for the development of PE intervention strategy by targeting CD97 in placentation and pathogenesis of PE.
Collapse
Affiliation(s)
- Huaxiang Shen
- 1 Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
- 2 Department of Obstetrics, Jiaxing Maternity and Child Health Hospital, Jiaxing, Zhejiang, People's Republic of China
| | - Minfei Jin
- 1 Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Shengyi Gu
- 1 Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Yuelin Wu
- 1 Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Mengnan Yang
- 1 Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Xiaolin Hua
- 1 Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
62
|
Gilding LN, Somervaille TCP. The Diverse Consequences of FOXC1 Deregulation in Cancer. Cancers (Basel) 2019; 11:E184. [PMID: 30764547 PMCID: PMC6406774 DOI: 10.3390/cancers11020184] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 02/06/2023] Open
Abstract
Forkhead box C1 (FOXC1) is a transcription factor with essential roles in mesenchymal lineage specification and organ development during normal embryogenesis. In keeping with these developmental properties, mutations that impair the activity of FOXC1 result in the heritable Axenfeld-Rieger Syndrome and other congenital disorders. Crucially, gain of FOXC1 function is emerging as a recurrent feature of malignancy; FOXC1 overexpression is now documented in more than 16 cancer types, often in association with an unfavorable prognosis. This review explores current evidence for FOXC1 deregulation in cancer and the putative mechanisms by which FOXC1 confers its oncogenic effects.
Collapse
Affiliation(s)
- L Niall Gilding
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4JG, UK.
| | - Tim C P Somervaille
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4JG, UK.
| |
Collapse
|
63
|
Whitsett JA, Kalin TV, Xu Y, Kalinichenko VV. Building and Regenerating the Lung Cell by Cell. Physiol Rev 2019; 99:513-554. [PMID: 30427276 DOI: 10.1152/physrev.00001.2018] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The unique architecture of the mammalian lung is required for adaptation to air breathing at birth and thereafter. Understanding the cellular and molecular mechanisms controlling its morphogenesis provides the framework for understanding the pathogenesis of acute and chronic lung diseases. Recent single-cell RNA sequencing data and high-resolution imaging identify the remarkable heterogeneity of pulmonary cell types and provides cell selective gene expression underlying lung development. We will address fundamental issues related to the diversity of pulmonary cells, to the formation and function of the mammalian lung, and will review recent advances regarding the cellular and molecular pathways involved in lung organogenesis. What cells form the lung in the early embryo? How are cell proliferation, migration, and differentiation regulated during lung morphogenesis? How do cells interact during lung formation and repair? How do signaling and transcriptional programs determine cell-cell interactions necessary for lung morphogenesis and function?
Collapse
Affiliation(s)
- Jeffrey A Whitsett
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Tanya V Kalin
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Yan Xu
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| | - Vladimir V Kalinichenko
- Perinatal Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Division of Neonatology, Perinatal and Pulmonary Biology, Cincinnati, Ohio
| |
Collapse
|
64
|
Magli A, Baik J, Mills LJ, Kwak IY, Dillon BS, Mondragon Gonzalez R, Stafford DA, Swanson SA, Stewart R, Thomson JA, Garry DJ, Dynlacht BD, Perlingeiro RCR. Time-dependent Pax3-mediated chromatin remodeling and cooperation with Six4 and Tead2 specify the skeletal myogenic lineage in developing mesoderm. PLoS Biol 2019; 17:e3000153. [PMID: 30807574 PMCID: PMC6390996 DOI: 10.1371/journal.pbio.3000153] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 02/01/2019] [Indexed: 12/26/2022] Open
Abstract
The transcriptional mechanisms driving lineage specification during development are still largely unknown, as the interplay of multiple transcription factors makes it difficult to dissect these molecular events. Using a cell-based differentiation platform to probe transcription function, we investigated the role of the key paraxial mesoderm and skeletal myogenic commitment factors-mesogenin 1 (Msgn1), T-box 6 (Tbx6), forkhead box C1 (Foxc1), paired box 3 (Pax3), Paraxis, mesenchyme homeobox 1 (Meox1), sine oculis-related homeobox 1 (Six1), and myogenic factor 5 (Myf5)-in paraxial mesoderm and skeletal myogenesis. From this study, we define a genetic hierarchy, with Pax3 emerging as the gatekeeper between the presomitic mesoderm and the myogenic lineage. By assaying chromatin accessibility, genomic binding and transcription profiling in mesodermal cells from mouse and human Pax3-induced embryonic stem cells and Pax3-null embryonic day (E)9.5 mouse embryos, we identified conserved Pax3 functions in the activation of the skeletal myogenic lineage through modulation of Hedgehog, Notch, and bone morphogenetic protein (BMP) signaling pathways. In addition, we demonstrate that Pax3 molecular function involves chromatin remodeling of its bound elements through an increase in chromatin accessibility and cooperation with sine oculis-related homeobox 4 (Six4) and TEA domain family member 2 (Tead2) factors. To our knowledge, these data provide the first integrated analysis of Pax3 function, demonstrating its ability to remodel chromatin in mesodermal cells from developing embryos and proving a mechanistic footing for the transcriptional hierarchy driving myogenesis.
Collapse
Affiliation(s)
- Alessandro Magli
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - June Baik
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Lauren J. Mills
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Il-Youp Kwak
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Bridget S. Dillon
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ricardo Mondragon Gonzalez
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - David A. Stafford
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Scott A. Swanson
- Morgridge Institute for Research, Madison, Wisconsin, United States of America
| | - Ron Stewart
- Morgridge Institute for Research, Madison, Wisconsin, United States of America
| | - James A. Thomson
- Morgridge Institute for Research, Madison, Wisconsin, United States of America
| | - Daniel J. Garry
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Brian D. Dynlacht
- Department of Pathology, New York University Cancer Institute, New York University School of Medicine, New York, New York, United States of America
| | - Rita C. R. Perlingeiro
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
65
|
Medina-Trillo C, Aroca-Aguilar JD, Ferre-Fernández JJ, Alexandre-Moreno S, Morales L, Méndez-Hernández CD, García-Feijoo J, Escribano J. Role of FOXC2 and PITX2 rare variants associated with mild functional alterations as modifier factors in congenital glaucoma. PLoS One 2019; 14:e0211029. [PMID: 30657791 PMCID: PMC6338360 DOI: 10.1371/journal.pone.0211029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Accepted: 01/07/2019] [Indexed: 11/19/2022] Open
Abstract
Congenital glaucoma (CG) is a severe and inherited childhood optical neuropathy that leads to irreversible visual loss and blindness in children. CG pathogenesis remains largely unexplained in most patients. Herein we have extended our previous studies to evaluate the role of FOXC2 and PITX2 variants in CG. Variants of the proximal promoter and transcribed sequence of these two genes were analyzed by Sanger sequencing in a cohort of 133 CG families. To investigate possible oligogenic inheritance involving FOXC2 or PITX2 and CYP1B1, we also analyzed FOXC2 and PITX2 variants in a group of 25 CG cases who were known to carry CYP1B1 glaucoma-associated genotypes. The functional effect of three identified variants was assessed by transactivation luciferase reporter assays, protein stability and subcellular localization analyses. We found eight probands (6.0%) who carried four rare FOXC2 variants in the heterozygous state. In addition, we found an elevated frequency (8%) of heterozygous and rare PITX2 variants in the group of CG cases who were known to carry CYP1B1 glaucoma-associated genotypes, and one of these PITX2 variants arose de novo. To the best of our knowledge, two of the identified variants (FOXC2: c.1183C>A, p.(H395N); and PITX2: c.535C>A, p.(P179T)) have not been previously identified. Examination of the genotype-phenotype correlation in this group suggests that the presence of the infrequent PITX2 variants increase the severity of the phenotype. Transactivation reporter analyses showed partial functional alteration of three identified amino acid substitutions (FOXC2: p.(C498R) and p.(H395N); PITX2: p.(P179T)). In summary, the increased frequency in PCG patients of rare FOXC2 and PITX2 variants with mild functional alterations, suggests they play a role as putative modifier factors in this disease further supporting that CG is not a simple monogenic disease and provides novel insights into the complex pathological mechanisms that underlie CG.
Collapse
Affiliation(s)
- Cristina Medina-Trillo
- Área de Genética, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, SPAIN
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Universidad de Castilla-La Mancha, Albacete, SPAIN
- Cooperative Research Network on Prevention, Early Detection and Treatment of Prevalent Degenerative and Chronic Ocular Pathology (OftaRed), Instituto de Salud Carlos III, Madrid, SPAIN
| | - José-Daniel Aroca-Aguilar
- Área de Genética, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, SPAIN
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Universidad de Castilla-La Mancha, Albacete, SPAIN
- Cooperative Research Network on Prevention, Early Detection and Treatment of Prevalent Degenerative and Chronic Ocular Pathology (OftaRed), Instituto de Salud Carlos III, Madrid, SPAIN
| | - Jesús-José Ferre-Fernández
- Área de Genética, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, SPAIN
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Universidad de Castilla-La Mancha, Albacete, SPAIN
- Cooperative Research Network on Prevention, Early Detection and Treatment of Prevalent Degenerative and Chronic Ocular Pathology (OftaRed), Instituto de Salud Carlos III, Madrid, SPAIN
| | - Susana Alexandre-Moreno
- Área de Genética, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, SPAIN
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Universidad de Castilla-La Mancha, Albacete, SPAIN
- Cooperative Research Network on Prevention, Early Detection and Treatment of Prevalent Degenerative and Chronic Ocular Pathology (OftaRed), Instituto de Salud Carlos III, Madrid, SPAIN
| | - Laura Morales
- Cooperative Research Network on Prevention, Early Detection and Treatment of Prevalent Degenerative and Chronic Ocular Pathology (OftaRed), Instituto de Salud Carlos III, Madrid, SPAIN
- Servicio de Oftalmología, Hospital San Carlos, Madrid, SPAIN
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, SPAIN
| | - Carmen-Dora Méndez-Hernández
- Cooperative Research Network on Prevention, Early Detection and Treatment of Prevalent Degenerative and Chronic Ocular Pathology (OftaRed), Instituto de Salud Carlos III, Madrid, SPAIN
- Servicio de Oftalmología, Hospital San Carlos, Madrid, SPAIN
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, SPAIN
| | - Julián García-Feijoo
- Cooperative Research Network on Prevention, Early Detection and Treatment of Prevalent Degenerative and Chronic Ocular Pathology (OftaRed), Instituto de Salud Carlos III, Madrid, SPAIN
- Servicio de Oftalmología, Hospital San Carlos, Madrid, SPAIN
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos, Madrid, SPAIN
| | - Julio Escribano
- Área de Genética, Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, SPAIN
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Universidad de Castilla-La Mancha, Albacete, SPAIN
- Cooperative Research Network on Prevention, Early Detection and Treatment of Prevalent Degenerative and Chronic Ocular Pathology (OftaRed), Instituto de Salud Carlos III, Madrid, SPAIN
| |
Collapse
|
66
|
Hariri H, Kurban M, Al-Haddad C, Fahed AC, Poladian S, Khalil A, Abbas O, Arabi M, Bitar F, Nemer G. Degenerated hair follicle cells and partial loss of sebaceous and eccrine glands in a familial case of axenfeld-rieger syndrome: An emerging role for the FOXC1/NFATC1 genetic axis. J Dermatol Sci 2018; 92:237-244. [PMID: 30514661 DOI: 10.1016/j.jdermsci.2018.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 11/05/2018] [Accepted: 11/05/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND Cutaneous malformations are at times associated with some forms of congenital heart defects. Many a times subtle cutaneous phenotypes maybe overlooked as their significance on the lives of individuals is minimal. Lebanon represents an area of high consanguinity, where the rates can go beyond 70% in some districts. For the past 6 years, we have been studying several genodermatoses in Lebanon including those with cardiac malformations. OBJECTIVES The main aim of this study is to document the genetic basis of a familial case of Axenfeld-Rieger Syndrome (ARS) with a mild cutaneous phenotype represented histologically with degeneration/ absence of hair follicles and incomplete formation of sebaceous and eccrine glands, in addition to the cardiac and ocular phenotypes. METHODS Whole exome sequencing was performed on two identical-twins with ARS along with their affected father and non-affected mother. Sanger sequencing was used to confirm the mutation, and the effects of the mutations on protein function was assessed in vitro using transient transfections. RESULTS A novel mutation inFOXC1 designated p.L240Rfs*75 was found in both twins and their father. The affected individuals share also a rare documented variant in NFATC1 designated p.V197 M. Both were absent from 200 Lebanese exomes. Our in vitro results suggested a gain of function activity of the FOXC1/NFATC1 complex, confirming its documented role in controlling murine hair follicle stem cells quiescence and regeneration. CONCLUSION This is the first documented human case with a mutation inFOXC1 regulating multi-organ developmental pathways that reflect a conserved mechanism in cell differentiation and proliferation.
Collapse
Affiliation(s)
- Hadla Hariri
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Mazen Kurban
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon; Department of Dermatology, American University of Beirut, Beirut, Lebanon; Department of Dermatology, Columbia University, New York, NY, United States
| | | | - Akl C Fahed
- Department of Genetics, Harvard Medical School, Boston, MA, United States; Department of Medicine, Massachusetts General Hospital, Boston, MA, United States; Howard Hughes Medical Institute and Division of Cardiology, Brigham and Women's Hospital, Boston, MA, United States
| | - Sarin Poladian
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Athar Khalil
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Oussama Abbas
- Department of Dermatology, American University of Beirut, Beirut, Lebanon
| | - Mariam Arabi
- Department of Pediatrics and Adolescent Medicine, American University of Beirut, Beirut, Lebanon
| | - Fadi Bitar
- Department of Pediatrics and Adolescent Medicine, American University of Beirut, Beirut, Lebanon
| | - Georges Nemer
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
67
|
Pan H, Peng Z, Lin J, Ren X, Zhang G, Cui Y. Forkhead box C1 boosts triple-negative breast cancer metastasis through activating the transcription of chemokine receptor-4. Cancer Sci 2018; 109:3794-3804. [PMID: 30290049 PMCID: PMC6272100 DOI: 10.1111/cas.13823] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 09/07/2018] [Accepted: 09/16/2018] [Indexed: 02/05/2023] Open
Abstract
The transcription factor forkhead box C1 (FOXC1) has recently been proposed as a crucial regulator of triple-negative breast cancer (TNBC) and associated with TNBC metastasis. However, the mechanism of FOXC1 in TNBC development and metastasis is elusive. In this study, overexpression of FOXC1 in MDA-MB-231 cells significantly enhanced, whereas knockdown of FOXC1 in BT549 cells significantly reduced, the capabilities of TNBC cell invasion and motility in vitro and metastasis to the lung in vivo, when compared to their respective control cells. Mechanistic studies revealed that FOXC1 increased the expression of CXC chemokine receptor-4 (CXCR4), probably through transcriptional activation. AMD3100, an inhibitor of CXCR4, could block cell migration. In a zebrafish tumor model, AMD3100 could suppress cell invasion and metastasis. In addition, overexpressing CXCR4 in FOXC1-knockdown BT549 cells increased the capabilities of TNBC cell invasion and motility. In contrast, inhibition of CXCR4 with either AMD3100 or siRNA in MDA-MB-231 cells overexpressing FOXC1 reduced the capabilities of invasion and motility. Taken together, our results reveal a potential mechanism for FOXC1-induced TNBC metastasis.
Collapse
Affiliation(s)
- Hongchao Pan
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and TreatmentCancer Hospital of Shantou University Medical CollegeShantouChina
| | - Zhilan Peng
- College of Food Science and TechnologyGuangdong Ocean UniversityZhanjiangChina
| | - Jiediao Lin
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and TreatmentCancer Hospital of Shantou University Medical CollegeShantouChina
| | - Xiaosha Ren
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and TreatmentCancer Hospital of Shantou University Medical CollegeShantouChina
| | - Guojun Zhang
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and TreatmentCancer Hospital of Shantou University Medical CollegeShantouChina
| | - Yukun Cui
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and TreatmentCancer Hospital of Shantou University Medical CollegeShantouChina
| |
Collapse
|
68
|
Tsuji-Tamura K, Ogawa M. Morphology regulation in vascular endothelial cells. Inflamm Regen 2018; 38:25. [PMID: 30214642 PMCID: PMC6130072 DOI: 10.1186/s41232-018-0083-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/07/2018] [Indexed: 12/22/2022] Open
Abstract
Morphological change in endothelial cells is an initial and crucial step in the process of establishing a functional vascular network. Following or associated with differentiation and proliferation, endothelial cells elongate and assemble into linear cord-like vessels, subsequently forming a perfusable vascular tube. In vivo and in vitro studies have begun to outline the underlying genetic and signaling mechanisms behind endothelial cell morphology regulation. This review focuses on the transcription factors and signaling pathways regulating endothelial cell behavior, involved in morphology, during vascular development.
Collapse
Affiliation(s)
- Kiyomi Tsuji-Tamura
- 1Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 860-0811 Japan.,2Present Address: Oral Biochemistry and Molecular Biology, Department of Oral Health Science, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan
| | - Minetaro Ogawa
- 1Department of Cell Differentiation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 860-0811 Japan
| |
Collapse
|
69
|
Mya N, Furutera T, Okuhara S, Kume T, Takechi M, Iseki S. Transcription factor Foxc1 is involved in anterior part of cranial base formation. Congenit Anom (Kyoto) 2018; 58:158-166. [PMID: 29322554 DOI: 10.1111/cga.12268] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 12/31/2017] [Accepted: 01/06/2018] [Indexed: 01/12/2023]
Abstract
The cranial base is a structure mainly formed through endochondral ossification and integrated into the craniofacial complex, which acts as an underlying platform for the developing brain. Foxc1 is an indispensable regulator during intramembranous and endochondral ossification. In this study, we found that the spontaneous loss of Foxc1 function in a mouse (congenital hydrocephalous), Foxc1ch/ch , demonstrated the anterior cranial base defects, including unossified presphenoid and lack of middle part of the basisphenoid bone. Hypoplastic presphenoid primordial cartilage (basal portion of the trabecular cartilage [bTB]) and a lack of the middle part of basisphenoid primordial cartilage (the hypophyseal cartilage) were consistently observed at earlier developmental stage. Foxc1 was expressed robustly and ubiquitously in undifferentiated mesenchyme of the cranial base-forming area in E11.0 wild-type fetuses. Once chondrogenesis commenced, the expression was downregulated and later limited to the perichondrium. Detection of transcripts of Collagen type2 A1 (Col2a1) revealed that both bTB and the anterior part of the hypophyseal cartilage developing anterior to the persistent epithelial stalk of the anterior lobe of the pituitary gland were suppressed in the Foxc1ch/ch . Proliferation activity of chondrocyte precursor cells was higher in the Foxc1ch/ch . Loss of Foxc1 function only in the neural crest cell lineage (Wnt1-cre;Foxc1ch/flox ) showed ossification of the posterior part of the hypophyseal cartilage derived from the mesoderm. These findings suggest that Foxc1 is an important regulator to further chondrogenesis and initiate the ossification of the presphenoid and basisphenoid bones.
Collapse
Affiliation(s)
- Nandar Mya
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshiko Furutera
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shigeru Okuhara
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tsutomu Kume
- Feinberg Cardiovascular Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Masaki Takechi
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sachiko Iseki
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
70
|
Fan TP, Ting HC, Yu JK, Su YH. Reiterative use of FGF signaling in mesoderm development during embryogenesis and metamorphosis in the hemichordate Ptychodera flava. BMC Evol Biol 2018; 18:120. [PMID: 30075704 PMCID: PMC6091094 DOI: 10.1186/s12862-018-1235-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 07/26/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Mesoderm is generally considered to be a germ layer that is unique to Bilateria, and it develops into diverse tissues, including muscle, and in the case of vertebrates, the skeleton and notochord. Studies on various deuterostome animals have demonstrated that fibroblast growth factor (FGF) signaling is required for the formation of many mesodermal structures, such as vertebrate somites, from which muscles are differentiated, and muscles in sea urchin embryos, suggesting an ancient role of FGF signaling in muscle development. However, the formation of trunk muscles in invertebrate chordates is FGF-independent, leading to ambiguity about this ancient role in deuterostomes. To further understand the role of FGF signaling during deuterostome evolution, we investigated the development of mesodermal structures during embryogenesis and metamorphosis in Ptychodera flava, an indirect-developing hemichordate that has larval morphology similar to echinoderms and adult body features that are similar to chordates. RESULTS Here we show that genes encoding FGF ligands, FGF receptors and transcription factors that are known to be involved in mesoderm formation and myogenesis are expressed dynamically during embryogenesis and metamorphosis. FGF signaling at the early gastrula stage is required for the specification of the mesodermal cell fate in P. flava. The mesoderm cells are then differentiated stepwise into the hydroporic canal, the pharyngeal muscle and the muscle string; formation of the last two muscular structures are controlled by FGF signaling. Moreover, augmentation of FGF signaling during metamorphosis accelerated the process, facilitating the transformation from cilia-driven swimming larvae into muscle-driven worm-like juveniles. CONCLUSIONS Our data show that FGF signaling is required for mesoderm induction and myogenesis in the P. flava embryo, and it is reiteratively used for the morphological transition during metamorphosis. The dependence of muscle development on FGF signaling in both planktonic larvae and sand-burrowing worms supports its ancestral role in deuterostomes.
Collapse
Affiliation(s)
- Tzu-Pei Fan
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, National Chung Hsing University and Academia Sinica, Taipei, 11529, Taiwan.,Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Rd., Sec. 2, Nankang, Taipei, 11529, Taiwan.,Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Hsiu-Chi Ting
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Rd., Sec. 2, Nankang, Taipei, 11529, Taiwan
| | - Jr-Kai Yu
- Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Rd., Sec. 2, Nankang, Taipei, 11529, Taiwan
| | - Yi-Hsien Su
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, National Chung Hsing University and Academia Sinica, Taipei, 11529, Taiwan. .,Institute of Cellular and Organismic Biology, Academia Sinica, 128 Academia Rd., Sec. 2, Nankang, Taipei, 11529, Taiwan. .,Biotechnology Center, National Chung Hsing University, Taichung, 40227, Taiwan.
| |
Collapse
|
71
|
Crawford JM, Lal BK, Durán WN, Pappas PJ. Pathophysiology of venous ulceration. J Vasc Surg Venous Lymphat Disord 2018. [PMID: 28624002 DOI: 10.1016/j.jvsv.2017.03.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Our understanding of the pathophysiologic process of venous ulceration has dramatically increased during the past two decades because of dedicated, venous-specific basic science research. Currently, the mechanisms regulating venous ulceration are a combination of macroscopic and microscopic pathologic processes. Macroscopic alterations refer to pathologic processes related to varicose vein formation, vein wall architecture, and cellular abnormalities that impair venous function. These processes are primarily caused by genetic factors that lead to the destruction of normal vein wall architecture and venous hypertension. Venous hypertension causes a chronic inflammatory response that over time can cause venous ulceration. The inciting inflammatory injury is chronic extravasation of macromolecules and red blood cell degradation products and iron overload. Chronic inflammation causes white blood cell extravasation into the dermis with secretion of numerous proinflammatory cytokines. These cytokines transform the phenotype of fibroblasts to a contractile phenotype that increases tension in the dermis. In addition, iron overload keeps macrophages in an M1 phenotype, which leads to tissue destruction instead of dermal repair. Current surgical and medical therapies are primarily directed at eliminating venous hypertension and promoting venous ulcer wound healing. Despite advances in our understanding of venous ulcer formation and healing, ulcers still take an average of 6 months to heal, and ulcer recurrence rates at 5 years are >58%. To improve the care of patients with venous ulcers, we need to further our understanding of the underlying pathologic events that lead to ulcer formation, prevent healing, and decrease ulcer-free recurrence intervals.
Collapse
Affiliation(s)
| | - Brajesh K Lal
- University of Maryland School of Medicine and the Baltimore Veterans Affairs Hospital, Baltimore, Md
| | | | | |
Collapse
|
72
|
Xu P, Balczerski B, Ciozda A, Louie K, Oralova V, Huysseune A, Crump JG. Fox proteins are modular competency factors for facial cartilage and tooth specification. Development 2018; 145:dev.165498. [PMID: 29777011 DOI: 10.1242/dev.165498] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/11/2018] [Indexed: 12/30/2022]
Abstract
Facial form depends on the precise positioning of cartilage, bone, and tooth fields in the embryonic pharyngeal arches. How complex signaling information is integrated to specify these cell types remains a mystery. We find that modular expression of Forkhead domain transcription factors (Fox proteins) in the zebrafish face arises through integration of Hh, Fgf, Bmp, Edn1 and Jagged-Notch pathways. Whereas loss of C-class Fox proteins results in reduced upper facial cartilages, loss of F-class Fox proteins results in distal jaw truncations and absent midline cartilages and teeth. We show that Fox proteins are required for Sox9a to promote chondrogenic gene expression. Fox proteins are sufficient in neural crest-derived cells for cartilage development, and neural crest-specific misexpression of Fox proteins expands the cartilage domain but inhibits bone. These results support a modular role for Fox proteins in establishing the competency of progenitors to form cartilage and teeth in the face.
Collapse
Affiliation(s)
- Pengfei Xu
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Bartosz Balczerski
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Amanda Ciozda
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Kristin Louie
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Veronika Oralova
- Evolutionary Developmental Biology, Ghent University, B-9000 Ghent, Belgium
| | - Ann Huysseune
- Evolutionary Developmental Biology, Ghent University, B-9000 Ghent, Belgium
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| |
Collapse
|
73
|
Wei C, Lin H, Cui S. The Forkhead Transcription Factor FOXC2 Is Required for Maintaining Murine Spermatogonial Stem Cells. Stem Cells Dev 2018; 27:624-636. [DOI: 10.1089/scd.2017.0233] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Chao Wei
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Hao Lin
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Sheng Cui
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| |
Collapse
|
74
|
Lescroart F, Wang X, Lin X, Swedlund B, Gargouri S, Sànchez-Dànes A, Moignard V, Dubois C, Paulissen C, Kinston S, Göttgens B, Blanpain C. Defining the earliest step of cardiovascular lineage segregation by single-cell RNA-seq. Science 2018; 359:1177-1181. [PMID: 29371425 PMCID: PMC6556615 DOI: 10.1126/science.aao4174] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 01/11/2018] [Indexed: 02/02/2023]
Abstract
Mouse heart development arises from Mesp1-expressing cardiovascular progenitors (CPs) that are specified during gastrulation. The molecular processes that control early regional and lineage segregation of CPs have been unclear. We performed single-cell RNA sequencing of wild-type and Mesp1-null CPs in mice. We showed that populations of Mesp1 CPs are molecularly distinct and span the continuum between epiblast and later mesodermal cells, including hematopoietic progenitors. Single-cell transcriptome analysis of Mesp1-deficient CPs showed that Mesp1 is required for the exit from the pluripotent state and the induction of the cardiovascular gene expression program. We identified distinct populations of Mesp1 CPs that correspond to progenitors committed to different cell lineages and regions of the heart, identifying the molecular features associated with early lineage restriction and regional segregation of the heart at the early stage of mouse gastrulation.
Collapse
Affiliation(s)
- Fabienne Lescroart
- Université Libre de Bruxelles, Laboratory of Stem Cells and Cancer, Brussels B-1070, Belgium
| | - Xiaonan Wang
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
- Wellcome and Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Xionghui Lin
- Université Libre de Bruxelles, Laboratory of Stem Cells and Cancer, Brussels B-1070, Belgium
| | - Benjamin Swedlund
- Université Libre de Bruxelles, Laboratory of Stem Cells and Cancer, Brussels B-1070, Belgium
| | - Souhir Gargouri
- Université Libre de Bruxelles, Laboratory of Stem Cells and Cancer, Brussels B-1070, Belgium
| | - Adriana Sànchez-Dànes
- Université Libre de Bruxelles, Laboratory of Stem Cells and Cancer, Brussels B-1070, Belgium
| | - Victoria Moignard
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
- Wellcome and Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Christine Dubois
- Université Libre de Bruxelles, Laboratory of Stem Cells and Cancer, Brussels B-1070, Belgium
| | - Catherine Paulissen
- Université Libre de Bruxelles, Laboratory of Stem Cells and Cancer, Brussels B-1070, Belgium
| | - Sarah Kinston
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK
- Wellcome and Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Berthold Göttgens
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK.
- Wellcome and Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Cédric Blanpain
- Université Libre de Bruxelles, Laboratory of Stem Cells and Cancer, Brussels B-1070, Belgium.
- WELBIO, Université Libre de Bruxelles, Brussels B-1070, Belgium
| |
Collapse
|
75
|
Ahn J, Han KS, Heo JH, Bang D, Kang YH, Jin HA, Hong SJ, Lee JH, Ham WS. FOXC2 and CLIP4 : a potential biomarker for synchronous metastasis of ≤7-cm clear cell renal cell carcinomas. Oncotarget 2018; 7:51423-51434. [PMID: 27283491 PMCID: PMC5239485 DOI: 10.18632/oncotarget.9842] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/20/2016] [Indexed: 12/15/2022] Open
Abstract
Renal cell carcinomas (RCC) smaller than 7-cm are heterogeneous and exhibit metastatic potential in approximately 15% of cases. Although large-scale characterization of mutations in clear cell RCC (ccRCC), the most common RCC subtype, has been established, the genetic alterations related to ≤7-cm ccRCCs undergoing synchronous metastasis are poorly understood. To discover biomarkers that can be used to estimate the risk of synchronous metastasis in these ccRCC patients, we performed whole exome sequencing on the formalin-fixed paraffin-embedded (FFPE) samples of 10 ccRCC patients with ≤7-cm tumors and synchronous metastasis and expanded our study using The Cancer Genome Atlas (TCGA) ccRCC dataset (n = 201). Recurrent mutations were selected according to functional prediction and statistical significance. Mutations in three candidate genes, RELN (1 out of 10), FOXC2 (1 out of 10), and CLIP4 (2 out of 10) were found in expanded analysis using a TCGA cohort. Furthermore, siRNA-mediated target gene knockdown (FOXC2 and CLIP4) and overexpression (RELN) assays showed that FOXC2 and CLIP4 significantly increased cell migration and viability in ccRCCs. Our study demonstrated that FOXC2 and CLIP4 activity correlates to the presence of ≤7-cm ccRCCs with synchronous metastasis and may be potential molecular predictors of synchronous metastasis of ≤7-cm ccRCCs.
Collapse
Affiliation(s)
- Jinwoo Ahn
- Department of Chemistry, Yonsei University, Seoul, Korea
| | - Kyung Seok Han
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Jun Hyeok Heo
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Duhee Bang
- Department of Chemistry, Yonsei University, Seoul, Korea
| | - You Hyun Kang
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Hyun A Jin
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Joon Hong
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hyun Lee
- Department of Clinical Pharmacology and Therapeutics, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Won Sik Ham
- Department of Urology and Urological Science Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
76
|
Bolte C, Whitsett JA, Kalin TV, Kalinichenko VV. Transcription Factors Regulating Embryonic Development of Pulmonary Vasculature. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2018; 228:1-20. [PMID: 29288383 DOI: 10.1007/978-3-319-68483-3_1] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Lung morphogenesis is a highly orchestrated process beginning with the appearance of lung buds on approximately embryonic day 9.5 in the mouse. Endodermally derived epithelial cells of the primitive lung buds undergo branching morphogenesis to generate the tree-like network of epithelial-lined tubules. The pulmonary vasculature develops in close proximity to epithelial progenitor cells in a process that is regulated by interactions between the developing epithelium and underlying mesenchyme. Studies in transgenic and knockout mouse models demonstrate that normal lung morphogenesis requires coordinated interactions between cells lining the tubules, which end in peripheral saccules, juxtaposed to an extensive network of capillaries. Multiple growth factors, microRNAs, transcription factors, and their associated signaling cascades regulate cellular proliferation, migration, survival, and differentiation during formation of the peripheral lung. Dysregulation of signaling events caused by gene mutations, teratogens, or premature birth causes severe congenital and acquired lung diseases in which normal alveolar architecture and the pulmonary capillary network are disrupted. Herein, we review scientific progress regarding signaling and transcriptional mechanisms regulating the development of pulmonary vasculature during lung morphogenesis.
Collapse
Affiliation(s)
- Craig Bolte
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, OH, USA.,Division of Pulmonary Biology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Jeffrey A Whitsett
- Division of Pulmonary Biology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA.,Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Tanya V Kalin
- Division of Pulmonary Biology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Vladimir V Kalinichenko
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, OH, USA. .,Division of Pulmonary Biology, Cincinnati Children's Research Foundation, Cincinnati, OH, USA. .,Division of Developmental Biology, Perinatal Institute, Cincinnati Children's Research Foundation, Cincinnati, OH, USA.
| |
Collapse
|
77
|
Yue Y, Jiang M, He L, Zhang Z, Zhang Q, Gu C, Liu M, Li N, Zhao Q. The transcription factor Foxc1a in zebrafish directly regulates expression of nkx2.5, encoding a transcriptional regulator of cardiac progenitor cells. J Biol Chem 2017; 293:638-650. [PMID: 29162723 DOI: 10.1074/jbc.ra117.000414] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/17/2017] [Indexed: 01/19/2023] Open
Abstract
Cardiogenesis is a tightly controlled biological process required for formation of a functional heart. The transcription factor Foxc1 not only plays a crucial role in outflow tract development in mice, but is also involved in cardiac structure formation and normal function in humans. However, the molecular mechanisms by which Foxc1 controls cardiac development remain poorly understood. Previously, we reported that zebrafish embryos deficient in foxc1a, an ortholog of mammalian Foxc1, display pericardial edemas and die 9-10 days postfertilization. To further investigate Foxc1a's role in zebrafish cardiogenesis and identify its downstream target genes during early heart development, we comprehensively analyzed the cardiovascular phenotype of foxc1a-null zebrafish embryos. Our results confirmed that foxc1a-null mutants exhibit disrupted cardiac morphology, structure, and function. Performing transcriptome analysis on the foxc1a mutants, we found that the expression of the cardiac progenitor marker gene nkx2.5 was significantly decreased, but the expression of germ layer-patterning genes was unaffected. Dual-fluorescence in situ hybridization assays revealed that foxc1a and nkx2.5 are co-expressed in the anterior lateral plate mesoderm at the somite stage. Chromatin immunoprecipitation and promoter truncation assays disclosed that Foxc1a regulates nkx2.5 expression via direct binding to two noncanonical binding sites in the proximal nkx2.5 promoter. Moreover, functional rescue experiments revealed that developmental stage-specific nkx2.5 overexpression partially rescues the cardiac defects of the foxc1a-null embryos. Taken together, our results indicate that during zebrafish cardiogenesis, Foxc1a is active directly upstream of nkx2.5.
Collapse
Affiliation(s)
- Yunyun Yue
- From the Model Animal Research Center, Ministry of Education Key Laboratory of Model Animal for Disease Study, Nanjing University, 12 Xuefu Road, Pukou High-tech Development Zone, Nanjing 210061, China
| | - Mingyang Jiang
- From the Model Animal Research Center, Ministry of Education Key Laboratory of Model Animal for Disease Study, Nanjing University, 12 Xuefu Road, Pukou High-tech Development Zone, Nanjing 210061, China
| | - Luqingqing He
- From the Model Animal Research Center, Ministry of Education Key Laboratory of Model Animal for Disease Study, Nanjing University, 12 Xuefu Road, Pukou High-tech Development Zone, Nanjing 210061, China
| | - Zhaojunjie Zhang
- From the Model Animal Research Center, Ministry of Education Key Laboratory of Model Animal for Disease Study, Nanjing University, 12 Xuefu Road, Pukou High-tech Development Zone, Nanjing 210061, China
| | - Qinxin Zhang
- From the Model Animal Research Center, Ministry of Education Key Laboratory of Model Animal for Disease Study, Nanjing University, 12 Xuefu Road, Pukou High-tech Development Zone, Nanjing 210061, China
| | - Chun Gu
- From the Model Animal Research Center, Ministry of Education Key Laboratory of Model Animal for Disease Study, Nanjing University, 12 Xuefu Road, Pukou High-tech Development Zone, Nanjing 210061, China
| | - Meijing Liu
- From the Model Animal Research Center, Ministry of Education Key Laboratory of Model Animal for Disease Study, Nanjing University, 12 Xuefu Road, Pukou High-tech Development Zone, Nanjing 210061, China
| | - Nan Li
- From the Model Animal Research Center, Ministry of Education Key Laboratory of Model Animal for Disease Study, Nanjing University, 12 Xuefu Road, Pukou High-tech Development Zone, Nanjing 210061, China
| | - Qingshun Zhao
- From the Model Animal Research Center, Ministry of Education Key Laboratory of Model Animal for Disease Study, Nanjing University, 12 Xuefu Road, Pukou High-tech Development Zone, Nanjing 210061, China
| |
Collapse
|
78
|
Wang J, Li W, Zheng X, Pang X, Du G. Research progress on the forkhead box C1. Oncotarget 2017; 9:12471-12478. [PMID: 29552326 PMCID: PMC5844762 DOI: 10.18632/oncotarget.22527] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 11/01/2017] [Indexed: 01/19/2023] Open
Abstract
FOXC1 is a vital member of FOX families which play important roles in biological processes including proliferation, differentiation, apoptosis, migration, invasion, metabolism, and longevity. Here we are focusing on roles of FOXC1 and their mechanisms in cancers. FOXC1 promoted progress of many cancers, such as breast cancer (especially basal-like breast cancer), hepatocellular carcinoma, gastric cancer and so on. FOXC1 was also found to be associated with drug resistance of cancers. FOXC1 promoted metastasis of cancers by increasing expression of MMP7, NEDD9 and Snail. Proliferation and invasion of cancers were increased by FOXC1 by mediating NF-κB, MST1R and KLF4 expression. FOXC1 was associated with development by regulating expression of FGF19 and MSX1. Recently, FOXC1 was found to be required for niche of stem cells or development of stem cells by mediating expression of Gli2, CXCL12, SCF, NFATC1, BMP and Myh7. Overall, FOXC1 exerts its functions by many mechanisms and may be used as a potential biomarker for diseases.
Collapse
Affiliation(s)
- Jinhua Wang
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China.,Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Wan Li
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Xiangjin Zheng
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Xiaocong Pang
- Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Guanhua Du
- The State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Beijing 100050, China.,Key Laboratory of Drug Target Research and Drug Screen, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
79
|
Gao B, Qu Y, Han B, Nagaoka Y, Katsumata M, Deng N, Bose S, Jin L, Giuliano AE, Cui X. Inhibition of lobuloalveolar development by FOXC1 overexpression in the mouse mammary gland. Sci Rep 2017; 7:14017. [PMID: 29070831 PMCID: PMC5656618 DOI: 10.1038/s41598-017-14342-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 10/10/2017] [Indexed: 11/09/2022] Open
Abstract
The forkhead box transcription factor FOXC1 plays a critical role in embryogenesis and the development of many organs. Its mutations and high expression are associated with many human diseases including breast cancer. Although FOXC1 knockout mouse studies showed that it is not required for mammary gland development during puberty, it is not clear whether its overexpression alters normal mammary development in vivo. To address this question, we generated transgenic mice with mammary-specific FOXC1 overexpression. We report that transgenic FOXC1 overexpression suppresses lobuloalveologenesis and lactation in mice. This phenotype is associated with higher percentages of estrogen receptor-, progesterone receptor-, or ki67-positive mammary epithelial cells in the transgenic mice at the lactation stage. We also show that expression of the Elf5 transcription factor, a master regulator of mammary alveologenesis and luminal cell differentiation, is markedly reduced in mammary epithelial cells of transgenic mice. Likewise, levels of activated Stat5, another inducer of alveolar expansion and a known mediator of the Elf5 effect, are also lowered in those cells. In contrast, the cytokeratin 8-positive mammary cell population with progenitor properties is elevated in the transgenic mice at the lactation stage, suggesting inhibition of mammary cell differentiation. These results may implicate FOXC1 as a new important regulator of mammary gland development.
Collapse
Affiliation(s)
- Bowen Gao
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048,, USA
| | - Ying Qu
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048,, USA
| | - Bingchen Han
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048,, USA
| | - Yoshiko Nagaoka
- Department of Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048,, USA
| | - Makoto Katsumata
- Department of Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048,, USA
| | - Nan Deng
- Biostatistics and Bioinformatics Research Center, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048,, USA
| | - Shikha Bose
- Department of Pathology, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048,, USA
| | - Liting Jin
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048,, USA.,Department of Breast Surgery, Hubei Cancer Hospital, Wuhan, Hubei, 430079,, China
| | - Armando E Giuliano
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048,, USA
| | - Xiaojiang Cui
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048,, USA.
| |
Collapse
|
80
|
Fogel JL, Lakeland DL, Mah IK, Mariani FV. A minimally sufficient model for rib proximal-distal patterning based on genetic analysis and agent-based simulations. eLife 2017; 6:e29144. [PMID: 29068314 PMCID: PMC5693115 DOI: 10.7554/elife.29144] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/24/2017] [Indexed: 12/19/2022] Open
Abstract
For decades, the mechanism of skeletal patterning along a proximal-distal axis has been an area of intense inquiry. Here, we examine the development of the ribs, simple structures that in most terrestrial vertebrates consist of two skeletal elements-a proximal bone and a distal cartilage portion. While the ribs have been shown to arise from the somites, little is known about how the two segments are specified. During our examination of genetically modified mice, we discovered a series of progressively worsening phenotypes that could not be easily explained. Here, we combine genetic analysis of rib development with agent-based simulations to conclude that proximal-distal patterning and outgrowth could occur based on simple rules. In our model, specification occurs during somite stages due to varying Hedgehog protein levels, while later expansion refines the pattern. This framework is broadly applicable for understanding the mechanisms of skeletal patterning along a proximal-distal axis.
Collapse
Affiliation(s)
- Jennifer L Fogel
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell ResearchUniversity of Southern CaliforniaLos AngelesUnited States
| | | | - In Kyoung Mah
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell ResearchUniversity of Southern CaliforniaLos AngelesUnited States
| | - Francesca V Mariani
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell ResearchUniversity of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
81
|
Abstract
Skeletal muscle is the largest tissue in the body and loss of its function or its regenerative properties results in debilitating musculoskeletal disorders. Understanding the mechanisms that drive skeletal muscle formation will not only help to unravel the molecular basis of skeletal muscle diseases, but also provide a roadmap for recapitulating skeletal myogenesis in vitro from pluripotent stem cells (PSCs). PSCs have become an important tool for probing developmental questions, while differentiated cell types allow the development of novel therapeutic strategies. In this Review, we provide a comprehensive overview of skeletal myogenesis from the earliest premyogenic progenitor stage to terminally differentiated myofibers, and discuss how this knowledge has been applied to differentiate PSCs into muscle fibers and their progenitors in vitro.
Collapse
Affiliation(s)
- Jérome Chal
- Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women's Hospital, 77 Avenue Louis Pasteur, Boston, MA 02115, USA .,Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS (UMR 7104), Inserm U964, Université de Strasbourg, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
82
|
Khalil A, Al-Haddad C, Hariri H, Shibbani K, Bitar F, Kurban M, Nemer G, Arabi M. A Novel Mutation in FOXC1 in a Lebanese Family with Congenital Heart Disease and Anterior Segment Dysgenesis: Potential Roles for NFATC1 and DPT in the Phenotypic Variations. Front Cardiovasc Med 2017; 4:58. [PMID: 28979898 PMCID: PMC5611365 DOI: 10.3389/fcvm.2017.00058] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/31/2017] [Indexed: 01/06/2023] Open
Abstract
Congenital heart diseases (CHDs) are still the leading cause of death in neonates. Anterior segment dysgenesis is a broad clinical phenotype that affects the normal development of the eye, leading in most of the cases to glaucoma which is still a major cause of blindness for children and adolescents. Despite tremendous insights gained from genetic studies, a clear genotype–phenotype correlation is still difficult to draw. In Lebanon, a small country with still a high rate of consanguineous marriages, there are little data on the epidemiology of glaucoma amongst children with or without CHD. We carried out whole exome sequencing (WES) on a family with anterior segment dysgenesis, and CHD composed of three affected children with glaucoma, two of them with structural cardiac defects and three healthy siblings. The results unravel a novel mutation in FOXC1 (p. R127H) segregating with the phenotype and inherited from the mother, who did not develop glaucoma. We propose a digenic model for glaucoma in this family by combining the FOXC1 variant with a missense variant inherited from the father in the dermatopontin (DPT) gene. We also unravel a novel NFATC1 missense mutation predicted to be deleterious and present only in the patient with a severe ocular and cardiac phenotype. This is the first report on FOXC1 using WES to genetically characterize a family with both ocular and cardiac malformations. Our results support the usage of such technology to have a better genotype–phenotype picture for Mendelian-inherited diseases for which expressivity and penetrance are still not answered.
Collapse
Affiliation(s)
- Athar Khalil
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | | | - Hadla Hariri
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Kamel Shibbani
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Fadi Bitar
- Department of Pediatrics and Adolescent Medicine, American University of Beirut, Beirut, Lebanon
| | - Mazen Kurban
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon.,Department of Dermatology, American University of Beirut, Beirut, Lebanon.,Department of Dermatology, Columbia University, New York, NY, United States
| | - Georges Nemer
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Mariam Arabi
- Department of Pediatrics and Adolescent Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
83
|
Hill JT, Demarest B, Gorsi B, Smith M, Yost HJ. Heart morphogenesis gene regulatory networks revealed by temporal expression analysis. Development 2017; 144:3487-3498. [PMID: 28807900 DOI: 10.1242/dev.154146] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/07/2017] [Indexed: 12/18/2022]
Abstract
During embryogenesis the heart forms as a linear tube that then undergoes multiple simultaneous morphogenetic events to obtain its mature shape. To understand the gene regulatory networks (GRNs) driving this phase of heart development, during which many congenital heart disease malformations likely arise, we conducted an RNA-seq timecourse in zebrafish from 30 hpf to 72 hpf and identified 5861 genes with altered expression. We clustered the genes by temporal expression pattern, identified transcription factor binding motifs enriched in each cluster, and generated a model GRN for the major gene batteries in heart morphogenesis. This approach predicted hundreds of regulatory interactions and found batteries enriched in specific cell and tissue types, indicating that the approach can be used to narrow the search for novel genetic markers and regulatory interactions. Subsequent analyses confirmed the GRN using two mutants, Tbx5 and nkx2-5, and identified sets of duplicated zebrafish genes that do not show temporal subfunctionalization. This dataset provides an essential resource for future studies on the genetic/epigenetic pathways implicated in congenital heart defects and the mechanisms of cardiac transcriptional regulation.
Collapse
Affiliation(s)
- Jonathon T Hill
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA .,Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Bradley Demarest
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Bushra Gorsi
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Megan Smith
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - H Joseph Yost
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
84
|
McQueen C, Pownall ME. An analysis of MyoD-dependent transcription using CRISPR/Cas9 gene targeting in Xenopus tropicalis embryos. Mech Dev 2017; 146:1-9. [DOI: 10.1016/j.mod.2017.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/18/2017] [Accepted: 05/19/2017] [Indexed: 12/11/2022]
|
85
|
Ovaert C, Busa T, Faure E, Missirian C, Philip N, Paoli F, Milh M, Macé L, Zaffran S. FOXC1 haploinsufficiency due to 6p25 deletion in a patient with rapidly progressing aortic valve disease. Am J Med Genet A 2017; 173:2489-2493. [PMID: 28657660 DOI: 10.1002/ajmg.a.38331] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 05/15/2017] [Accepted: 05/24/2017] [Indexed: 01/12/2023]
Abstract
6p25 deletion is a rare but well-known entity. The main clinical features include an abnormal facial appearance, developmental delay, and ocular anomalies. Cardiac anomalies are frequently seen but remain poorly delineated. We describe a 4-year-old girl with 6p25.3 deletion, which includes the FOXC1 gene, typical dysmorphic features associated with developmental delay and oculo-motor anomalies. Aortic valve dysplasia was diagnosed early in life. The cardiac lesion progressed very rapidly between the age of 3 and 4 years requiring aortic valve replacement. Genomic analysis of blood and excised valve tissue showed down-regulation of FOXC1 but also FOXC2 expression in the diseased aortic valve. This allows us to speculate on the potential role of FOXC1 in aortic valve anomalies.
Collapse
Affiliation(s)
- Caroline Ovaert
- Department of Pediatric and Congenital Cardiology, Timone Enfant, AP-HM, Marseille, France.,Faculté de Médecine, Inserm, GMGF, UMR_S910, Aix Marseille Université, Marseille, France
| | - Tiffany Busa
- Faculté de Médecine, Inserm, GMGF, UMR_S910, Aix Marseille Université, Marseille, France.,Department of Clinical Genetics, Timone Enfant, AP-HM, Marseille, France
| | - Emilie Faure
- Faculté de Médecine, Inserm, GMGF, UMR_S910, Aix Marseille Université, Marseille, France
| | - Chantal Missirian
- Faculté de Médecine, Inserm, GMGF, UMR_S910, Aix Marseille Université, Marseille, France.,Department of Clinical Genetics, Timone Enfant, AP-HM, Marseille, France
| | - Nicole Philip
- Faculté de Médecine, Inserm, GMGF, UMR_S910, Aix Marseille Université, Marseille, France.,Department of Clinical Genetics, Timone Enfant, AP-HM, Marseille, France
| | - Florent Paoli
- Department of Pediatric and Congenital Cardiology, Timone Enfant, AP-HM, Marseille, France
| | - Mathieu Milh
- Faculté de Médecine, Inserm, GMGF, UMR_S910, Aix Marseille Université, Marseille, France.,Department of Pediatric Neurology, Timone Enfant, AP-HM, Marseille, France
| | - Loic Macé
- Department of Pediatric and Congenital Cardiology, Timone Enfant, AP-HM, Marseille, France
| | - Stephane Zaffran
- Faculté de Médecine, Inserm, GMGF, UMR_S910, Aix Marseille Université, Marseille, France
| |
Collapse
|
86
|
Seo S, Chen L, Liu W, Zhao D, Schultz KM, Sasman A, Liu T, Zhang HF, Gage PJ, Kume T. Foxc1 and Foxc2 in the Neural Crest Are Required for Ocular Anterior Segment Development. Invest Ophthalmol Vis Sci 2017; 58:1368-1377. [PMID: 28253399 PMCID: PMC5361455 DOI: 10.1167/iovs.16-21217] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Purpose The large Forkhead (Fox) transcription factor family has essential roles in development, and mutations cause a wide range of ocular and nonocular disease. One member, Foxc2 is expressed in neural crest (NC)-derived periocular mesenchymal cells of the developing murine eye; however, its precise role in the development, establishment, and maintenance of the ocular surface has yet to be investigated. Methods To specifically delete Foxc2 from NC-derived cells, conditional knockout mice for Foxc2 (NC-Foxc2−/−) were generated by crossing Foxc2F mice with Wnt1-Cre mice. Similarly, we also generated compound NC-specific mutations of Foxc2 and a closely related gene, Foxc1 (NC-Foxc1−/−;NC-Foxc2−/−) in mice. Results Neural crest-Foxc2−/− mice show abnormal thickness in the peripheral-to-central corneal stroma and limbus and displaced pupils with irregular iris. The neural crest-specific mutation in Foxc2 also leads to ectopic neovascularization in the cornea, as well as impaired ocular epithelial cell identity and corneal conjunctivalization. Compound, NC-specific Foxc1; Foxc2 homozygous mutant mice have more severe defects in structures of the ocular surface, such as the cornea and eyelids, accompanied by significant declines in the expression of another key developmental factor, Pitx2, and its downstream effector Dkk2, which antagonizes canonical Wnt signaling. Conclusions The neural crest-Foxc2 mutation is associated with corneal conjunctivalization, ectopic corneal neovascularization, and disrupted ocular epithelial cell identity. Furthermore, Foxc2 and Foxc1 cooperatively function in NC-derived mesenchymal cells to ensure proper morphogenesis of the ocular surface via the regulation of Wnt signaling. Together, Foxc2 is required in the NC lineage for mesenchymal-epithelial interactions in corneal and ocular surface development.
Collapse
Affiliation(s)
- Seungwoon Seo
- Feinberg Cardiovascular Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States 2Department of Life Science, Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Lisheng Chen
- Department of Ophthalmology and Visual Science, University of Michigan, Ann Arbor, Michigan, United States
| | - Wenzhong Liu
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, United States
| | - Demin Zhao
- Feinberg Cardiovascular Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Kathryn M Schultz
- Feinberg Cardiovascular Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Amy Sasman
- Feinberg Cardiovascular Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Ting Liu
- Feinberg Cardiovascular Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Hao F Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois, United States
| | - Philip J Gage
- Department of Ophthalmology and Visual Science, University of Michigan, Ann Arbor, Michigan, United States
| | - Tsutomu Kume
- Feinberg Cardiovascular Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| |
Collapse
|
87
|
Han B, Bhowmick N, Qu Y, Chung S, Giuliano AE, Cui X. FOXC1: an emerging marker and therapeutic target for cancer. Oncogene 2017; 36:3957-3963. [PMID: 28288141 PMCID: PMC5652000 DOI: 10.1038/onc.2017.48] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/03/2017] [Accepted: 02/04/2017] [Indexed: 02/07/2023]
Abstract
The Forkhead box C1 (FOXC1) transcription factor is involved in normal embryonic development and regulates the development and function of many organs. Most recently, a large body of literature has shown that FOXC1 plays a critical role in tumor development and metastasis. Clinical studies have demonstrated that elevated FOXC1 expression is associated with poor prognosis in many cancer subtypes, such as basal-like breast cancer (BLBC). FOXC1 is highly and specifically expressed in BLBC as opposed to other breast cancer subtypes. Its functions in breast cancer have been extensively explored. This review will summarize current knowledge on the function and regulation of FOXC1 in tumor development and progression with a focus on BLBC as well as the implications of these new findings in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- B Han
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - N Bhowmick
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Y Qu
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - S Chung
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - A E Giuliano
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - X Cui
- Department of Surgery, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
88
|
Xie HM, Werner P, Stambolian D, Bailey-Wilson JE, Hakonarson H, White PS, Taylor DM, Goldmuntz E. Rare copy number variants in patients with congenital conotruncal heart defects. Birth Defects Res 2017; 109:271-295. [PMID: 28398664 PMCID: PMC5407323 DOI: 10.1002/bdra.23609] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/22/2016] [Accepted: 11/30/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Previous studies using different cardiac phenotypes, technologies and designs suggest a burden of large, rare or de novo copy number variants (CNVs) in subjects with congenital heart defects. We sought to identify disease-related CNVs, candidate genes, and functional pathways in a large number of cases with conotruncal and related defects that carried no known genetic syndrome. METHODS Cases and control samples were divided into two cohorts and genotyped to assess each subject's CNV content. Analyses were performed to ascertain differences in overall CNV prevalence and to identify enrichment of specific genes and functional pathways in conotruncal cases relative to healthy controls. RESULTS Only findings present in both cohorts are presented. From 973 total conotruncal cases, a burden of rare CNVs was detected in both cohorts. Candidate genes from rare CNVs found in both cohorts were identified based on their association with cardiac development or disease, and/or their reported disruption in published studies. Functional and pathway analyses revealed significant enrichment of terms involved in either heart or early embryonic development. CONCLUSION Our study tested one of the largest cohorts specifically with cardiac conotruncal and related defects. These results confirm and extend previous findings that CNVs contribute to disease risk for congenital heart defects in general and conotruncal defects in particular. As disease heterogeneity renders identification of single recurrent genes or loci difficult, functional pathway and gene regulation network analyses appear to be more informative. Birth Defects Research 109:271-295, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hongbo M Xie
- The Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Petra Werner
- Division of Cardiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Dwight Stambolian
- Department of Ophthalmology and Human Genetics, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joan E Bailey-Wilson
- Statistical Genetics Section, National Human Genome Research Institute, National Institutes of Health, Baltimore, Maryland
| | - Hakon Hakonarson
- The Center for Applied Genomics, Department of Pediatrics, The Children's Hospital of Philadelphia, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Peter S White
- Division of Biomedical Informatics, Cincinnati Children's Hospital, Department of Biomedical Informatics, University of Cincinnati, Cincinnati, Ohio
| | - Deanne M Taylor
- The Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Elizabeth Goldmuntz
- Division of Cardiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
89
|
Motojima M, Kume T, Matsusaka T. Foxc1 and Foxc2 are necessary to maintain glomerular podocytes. Exp Cell Res 2017; 352:265-272. [PMID: 28223138 DOI: 10.1016/j.yexcr.2017.02.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/07/2017] [Accepted: 02/11/2017] [Indexed: 12/11/2022]
Abstract
Foxc1 and Foxc2 (Foxc1/2) are transcription factors involved in many biological processes. In adult kidneys, expression of Foxc1/2 is confined to the glomerular epithelial cells, i.e., podocytes. To bypass embryonic lethality of Foxc1/2 null mice, mice ubiquitously expressing inducible-Cre (ROSA26-CreERT2) or mice expressing Cre in podocytes (Nephrin-Cre) were mated with floxed-Foxc1 and floxed-Foxc2 mice. The CreERT2 was activated in adult mice by administrations of tamoxifen. Eight weeks after tamoxifen treatment, ROSA26-CreERT2; Foxc1+/flox; Foxc2flox/flox mice developed microalbuminuria, while ROSA26-Cre ERT2; Foxc1flox/flox; Foxc2+/flox mice had no microalbuminuria. The kidneys of conditional-Foxc1/2 null mice showed proteinaceous casts, protein reabsorption droplets in tubules and huge vacuoles in podocytes, indicating severe podocyte injury and massive proteinuria. Comparison of gene expression profiles revealed that Foxc1/2 maintain expression of genes necessary for podocyte function such as podocin and Cxcl12. In addition, mice with an innate podocyte-specific deletion of Foxc1/2 by Nephrin-Cre develop similar podocyte injury. These results demonstrate dose-dependence of Foxc1/2 gene in maintaining the podocyte with a more critical role for Foxc2 than Foxc1 and a critical role of Foxc1/2 in regulating expression of genes that maintain podocyte integrity.
Collapse
Affiliation(s)
- Masaru Motojima
- Department of Clinical Pharmacology, Tokai University School of Medicine, Isehara, Kanagawa 259-1193, Japan.
| | - Tsutomu Kume
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | - Taiji Matsusaka
- Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa 259-1193, Japan
| |
Collapse
|
90
|
Amin MB, Miura N, Uddin MKM, Islam MJ, Yoshida N, Iseki S, Kume T, Trainor PA, Saitsu H, Aoto K. Foxc2 CreERT2 knock-in mice mark stage-specific Foxc2-expressing cells during mouse organogenesis. Congenit Anom (Kyoto) 2017; 57:24-31. [PMID: 27783871 DOI: 10.1111/cga.12198] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/18/2016] [Accepted: 10/19/2016] [Indexed: 11/28/2022]
Abstract
Foxc2, a member of the winged helix transcription factor family, is essential for eye, calvarial bone, cardiovascular and kidney development in mice. Nevertheless, how Foxc2-expressing cells and their descendent cells contribute to the development of these tissues and organs has not been elucidated. Here, we generated a Foxc2 knock-in (Foxc2CreERT2 ) mouse, in which administration of estrogen receptor antagonist tamoxifen induces nuclear translocation of Cre recombinase in Foxc2-expressing cells. By crossing with ROSA-LacZ reporter mice (Foxc2CreERT2 ; R26R), the fate of Foxc2 positive (Foxc2+ ) cells was analyzed through LacZ staining at various embryonic stages. We found Foxc2+ cell descendants in the supraoccipital and exoccipital bone in E18.5 embryos, when tamoxifen was administered at embryonic day (E) 8.5. Furthermore, Foxc2+ descendant cranial neural crest cells at E8-10 were restricted to the corneal mesenchyme, while Foxc2+ cell derived cardiac neural crest cells at E6-12 were found in the aorta, pulmonary trunk and valves, and endocardial cushions. Foxc2+ cell descendant contributions to the glomerular podocytes in the kidney were also observed following E6.5 tamoxifen treatment. Our results are consistent with previous reports of Foxc2 expression during early embryogenesis and the Foxc2CreERT2 mouse provides a tool to investigate spatiotemporal roles of Foxc2 and contributions of Foxc2+ expressing cells during mouse embryogenesis.
Collapse
Affiliation(s)
- Mohammed Badrul Amin
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Naoyuki Miura
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | | | - Nobuaki Yoshida
- Laboratory of Developmental Genetics, Center for Experimental Medicine and Systems Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Sachiko Iseki
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tsutomu Kume
- Erin Lambers, Feinberg Cardiovascular Research Institute, Department of Medicine, Northwestern University School of Medicine, Chicago, Illinois, USA
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, Missouri, USA.,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Missouri, USA
| | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kazushi Aoto
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
91
|
Lee S, Park C, Han JW, Kim JY, Cho K, Kim EJ, Kim S, Lee SJ, Oh SY, Tanaka Y, Park IH, An HJ, Shin CM, Sharma S, Yoon YS. Direct Reprogramming of Human Dermal Fibroblasts Into Endothelial Cells Using ER71/ETV2. Circ Res 2016; 120:848-861. [PMID: 28003219 DOI: 10.1161/circresaha.116.309833] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 12/15/2016] [Accepted: 12/21/2016] [Indexed: 12/14/2022]
Abstract
RATIONALE Direct conversion or reprogramming of human postnatal cells into endothelial cells (ECs), bypassing stem or progenitor cell status, is crucial for regenerative medicine, cell therapy, and pathophysiological investigation but has remained largely unexplored. OBJECTIVE We sought to directly reprogram human postnatal dermal fibroblasts to ECs with vasculogenic and endothelial transcription factors and determine their vascularizing and therapeutic potential. METHODS AND RESULTS We utilized various combinations of 7 EC transcription factors to transduce human postnatal dermal fibroblasts and found that ER71/ETV2 (ETS variant 2) alone best induced endothelial features. KDR+ (kinase insert domain receptor) cells sorted at day 7 from ER71/ETV2-transduced human postnatal dermal fibroblasts showed less mature but enriched endothelial characteristics and thus were referred to as early reprogrammed ECs (rECs), and did not undergo maturation by further culture. After a period of several weeks' transgene-free culture followed by transient reinduction of ER71/ETV2, early rECs matured during 3 months of culture and showed reduced ETV2 expression, reaching a mature phenotype similar to postnatal human ECs. These were termed late rECs. While early rECs exhibited an immature phenotype, their implantation into ischemic hindlimbs induced enhanced recovery from ischemia. These 2 rECs showed clear capacity for contributing to new vessel formation through direct vascular incorporation in vivo. Paracrine or proangiogenic effects of implanted early rECs played a significant role in repairing hindlimb ischemia. CONCLUSIONS This study for the first time demonstrates that ER71/ETV2 alone can directly reprogram human postnatal cells to functional, mature ECs after an intervening transgene-free period. These rECs could be valuable for cell therapy, personalized disease investigation, and exploration of the reprogramming process.
Collapse
Affiliation(s)
- Sangho Lee
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Changwon Park
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Ji Woong Han
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Ju Young Kim
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Kyuwon Cho
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Eun Jae Kim
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Sangsung Kim
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Shin-Jeong Lee
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Se Yeong Oh
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Yoshiaki Tanaka
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - In-Hyun Park
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Hyo Jae An
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Claire Min Shin
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Shraya Sharma
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| | - Young-Sup Yoon
- From the Department of Pharmacology, College of Medicine, University of Illinois at Chicago (C.P., E.J.K.); Department of Pediatrics, Children's Heart Research and Outcomes Center, Emory University School of Medicine, Atlanta, GA (C.P., J.Y.K., S.Y.O.); Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (S.L., J.W.H., K.C., S.K., H.J.A., C.M.S., S.S., Y.-s.Y.); Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT (Y.T., I.-H.P.); and Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea (S.-J.L., Y.-s.Y.)
| |
Collapse
|
92
|
Ko FC, Martins JS, Reddy P, Bragdon B, Hussein AI, Gerstenfeld LC, Demay MB. Acute Phosphate Restriction Impairs Bone Formation and Increases Marrow Adipose Tissue in Growing Mice. J Bone Miner Res 2016; 31:2204-2214. [PMID: 27324177 DOI: 10.1002/jbmr.2891] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/06/2016] [Accepted: 06/13/2016] [Indexed: 01/28/2023]
Abstract
Phosphate plays a critical role in chondrocyte maturation and skeletal mineralization. Studies examining the consequences of dietary phosphate restriction in growing mice demonstrated not only the development of rickets, but also a dramatic decrease in bone accompanied by increased marrow adipose tissue (MAT). Thus studies were undertaken to determine the effects of dietary phosphate restriction on bone formation and bone marrow stromal cell (BMSC) differentiation. Acute phosphate restriction of 28-day-old mice profoundly inhibited bone formation within 48 hours. It also resulted in increased mRNA expression of the early osteolineage markers Sox9 and Runt-related transcription factor 2 (Runx2), accompanied by decreased expression of the late osteolineage markers Osterix and Osteocalcin in BMSCs and osteoblasts, suggesting that phosphate restriction arrests osteoblast differentiation between Runx2 and Osterix. Increased expression of PPARγ and CEBPα, key regulators of adipogenic differentiation, was observed within 1 week of dietary phosphate restriction and was followed by a 13-fold increase in MAT at 3 weeks of phosphate restriction. In vitro phosphate restriction did not alter BMSC osteogenic or adipogenic colony formation, implicating aberrant paracrine or endocrine signaling in the in vivo phenotype. Because BMP signaling regulates the transition between Runx2 and Osterix, this pathway was interrogated. A dramatic decrease in pSmad1/5/9 immunoreactivity was observed in the osteoblasts of phosphate-restricted mice on day 31 (d31) and d35. This was accompanied by attenuated expression of the BMP target genes Id1, KLF10, and Foxc2, the latter of which promotes osteogenic and angiogenic differentiation while impairing adipogenesis. A decrease in expression of the Notch target gene Hey1, a BMP-regulated gene that governs angiogenesis, was also observed in phosphate-restricted mice, in association with decreased metaphyseal marrow vasculature. Whereas circulating phosphate levels are known to control growth plate maturation and skeletal mineralization, these studies reveal novel consequences of phosphate restriction in the regulation of bone formation and osteoblast differentiation. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Frank C Ko
- Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Janaina S Martins
- Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Pooja Reddy
- Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Beth Bragdon
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, USA
| | - Amira I Hussein
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, USA
| | - Louis C Gerstenfeld
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, USA
| | - Marie B Demay
- Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
93
|
Liu CF, Samsa WE, Zhou G, Lefebvre V. Transcriptional control of chondrocyte specification and differentiation. Semin Cell Dev Biol 2016; 62:34-49. [PMID: 27771362 DOI: 10.1016/j.semcdb.2016.10.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 10/18/2016] [Indexed: 12/20/2022]
Abstract
A milestone in the evolutionary emergence of vertebrates was the invention of cartilage, a tissue that has key roles in modeling, protecting and complementing the bony skeleton. Cartilage is elaborated and maintained by chondrocytes. These cells derive from multipotent skeletal progenitors and they perform highly specialized functions as they proceed through sequential lineage commitment and differentiation steps. They form cartilage primordia, the primary skeleton of the embryo. They then transform these primordia either into cartilage growth plates, temporary drivers of skeletal elongation and endochondral ossification, or into permanent tissues, namely articular cartilage. Chondrocyte fate decisions and differentiated activities are controlled by numerous extrinsic and intrinsic cues, and they are implemented at the gene expression level by transcription factors. The latter are the focus of this review. Meritorious efforts from many research groups have led over the last two decades to the identification of dozens of key chondrogenic transcription factors. These regulators belong to all types of transcription factor families. Some have master roles at one or several differentiation steps. They include SOX9 and RUNX2/3. Others decisively assist or antagonize the activities of these masters. They include TWIST1, SOX5/6, and MEF2C/D. Many more have tissue-patterning roles and regulate cell survival, proliferation and the pace of cell differentiation. They include, but are not limited to, homeodomain-containing proteins and growth factor signaling mediators. We here review current knowledge of all these factors, one superclass, class, and family at a time. We then compile all knowledge into transcriptional networks. We also identify remaining gaps in knowledge and directions for future research to fill these gaps and thereby provide novel insights into cartilage disease mechanisms and treatment options.
Collapse
Affiliation(s)
- Chia-Feng Liu
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, 44195, USA.
| | - William E Samsa
- Department of Orthopaedics, Case Western Reserve University, Cleveland, OH, USA
| | - Guang Zhou
- Department of Orthopaedics, Case Western Reserve University, Cleveland, OH, USA; Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA; Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA
| | - Véronique Lefebvre
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, 44195, USA.
| |
Collapse
|
94
|
Yeo HC, Ting S, Brena RM, Koh G, Chen A, Toh SQ, Lim YM, Oh SKW, Lee DY. Genome-Wide Transcriptome and Binding Sites Analyses Identify Early FOX Expressions for Enhancing Cardiomyogenesis Efficiency of hESC Cultures. Sci Rep 2016; 6:31068. [PMID: 27501774 PMCID: PMC4977469 DOI: 10.1038/srep31068] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 07/14/2016] [Indexed: 01/19/2023] Open
Abstract
The differentiation efficiency of human embryonic stem cells (hESCs) into heart muscle cells (cardiomyocytes) is highly sensitive to culture conditions. To elucidate the regulatory mechanisms involved, we investigated hESCs grown on three distinct culture platforms: feeder-free Matrigel, mouse embryonic fibroblast feeders, and Matrigel replated on feeders. At the outset, we profiled and quantified their differentiation efficiency, transcriptome, transcription factor binding sites and DNA-methylation. Subsequent genome-wide analyses allowed us to reconstruct the relevant interactome, thereby forming the regulatory basis for implicating the contrasting differentiation efficiency of the culture conditions. We hypothesized that the parental expressions of FOXC1, FOXD1 and FOXQ1 transcription factors (TFs) are correlative with eventual cardiomyogenic outcome. Through WNT induction of the FOX TFs, we observed the co-activation of WNT3 and EOMES which are potent inducers of mesoderm differentiation. The result strengthened our hypothesis on the regulatory role of the FOX TFs in enhancing mesoderm differentiation capacity of hESCs. Importantly, the final proportions of cells expressing cardiac markers were directly correlated to the strength of FOX inductions within 72 hours after initiation of differentiation across different cell lines and protocols. Thus, we affirmed the relationship between early FOX TF expressions and cardiomyogenesis efficiency.
Collapse
Affiliation(s)
- Hock Chuan Yeo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore 138668, Singapore.,Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore
| | - Sherwin Ting
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore 138668, Singapore
| | - Romulo Martin Brena
- USC Epigenome Center, University of Southern California, Los Angeles, CA 90033, USA
| | - Geoffrey Koh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore 138668, Singapore
| | - Allen Chen
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore 138668, Singapore
| | - Siew Qi Toh
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore
| | - Yu Ming Lim
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore 138668, Singapore
| | - Steve Kah Weng Oh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore 138668, Singapore
| | - Dong-Yup Lee
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01, Singapore 138668, Singapore.,Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.,NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, 28 Medical Drive, Singapore 117456, Singapore
| |
Collapse
|
95
|
Fatima A, Wang Y, Uchida Y, Norden P, Liu T, Culver A, Dietz WH, Culver F, Millay M, Mukouyama YS, Kume T. Foxc1 and Foxc2 deletion causes abnormal lymphangiogenesis and correlates with ERK hyperactivation. J Clin Invest 2016; 126:2437-51. [PMID: 27214551 PMCID: PMC4922698 DOI: 10.1172/jci80465] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 04/05/2016] [Indexed: 01/12/2023] Open
Abstract
The lymphatic vasculature is essential for maintaining interstitial fluid homeostasis, and dysfunctional lymphangiogenesis contributes to various pathological processes, including inflammatory disease and tumor metastasis. Mutations in FOXC2 are dominantly associated with late-onset lymphedema; however, the precise role of FOXC2 and a closely related factor, FOXC1, in the lymphatic system remains largely unknown. Here we identified a molecular cascade by which FOXC1 and FOXC2 regulate ERK signaling in lymphatic vessel growth. In mice, lymphatic endothelial cell-specific (LEC-specific) deletion of Foxc1, Foxc2, or both resulted in increased LEC proliferation, enlarged lymphatic vessels, and abnormal lymphatic vessel morphogenesis. Compared with LECs from control animals, LECs from mice lacking both Foxc1 and Foxc2 exhibited aberrant expression of Ras regulators, and embryos with LEC-specific deletion of Foxc1 and Foxc2, alone or in combination, exhibited ERK hyperactivation. Pharmacological ERK inhibition in utero abolished the abnormally enlarged lymphatic vessels in FOXC-deficient embryos. Together, these results identify FOXC1 and FOXC2 as essential regulators of lymphangiogenesis and indicate a new potential mechanistic basis for lymphatic-associated diseases.
Collapse
Affiliation(s)
- Anees Fatima
- Feinberg Cardiovascular Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ying Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Yutaka Uchida
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Pieter Norden
- Feinberg Cardiovascular Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ting Liu
- Feinberg Cardiovascular Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Austin Culver
- Feinberg Cardiovascular Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - William H. Dietz
- Feinberg Cardiovascular Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Ford Culver
- Feinberg Cardiovascular Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Meredith Millay
- Feinberg Cardiovascular Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Yoh-suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Tsutomu Kume
- Feinberg Cardiovascular Research Institute, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
96
|
Sato A, Kamekura R, Kawata K, Kawada M, Jitsukawa S, Yamashita K, Sato N, Himi T, Ichimiya S. Novel Mechanisms of Compromised Lymphatic Endothelial Cell Homeostasis in Obesity: The Role of Leptin in Lymphatic Endothelial Cell Tube Formation and Proliferation. PLoS One 2016; 11:e0158408. [PMID: 27366905 PMCID: PMC4930203 DOI: 10.1371/journal.pone.0158408] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 06/15/2016] [Indexed: 11/19/2022] Open
Abstract
Leptin is a hormone produced by adipose tissue that regulates various physiological processes. Recent studies have shown that the level of circulating leptin is elevated in obese patients and have suggested a relationship between obesity and postoperative lymphedema. However, the mechanisms by which postoperative lymphedema develops in obese patients and the mechanisms by which leptin regulates lymphatic endothelial cell homeostasis such as tube formation and cell proliferation remain unknown. Here we report that leptin regulates tube formation and cell proliferation in human dermal lymphatic endothelial cells (HDLECs) by activation of the signal transducer and activator of transcription 3 pathway, which is downstream signaling of the leptin receptor. Additionally, we found that upregulation of suppressor of cytokine signaling 3 underlies the mechanisms by which a high dose of leptin inhibits cell proliferation and tube formation. Leptin also enhanced expression of the proinflammatory cytokine IL-6 in HDLECs. Interestingly, IL-6 rescues the compromised cell proliferation and tube formation caused by treatment with a high dose of leptin in an autocrine or paracrine manner. Taken together, our findings reveal a novel mechanism by which compromised HDLECs maintain their homeostasis during inflammation mediated by leptin and IL-6. Thus, regulating the level of leptin or IL-6 may be a viable strategy to reduce the incidence of postoperative lymphedema.
Collapse
Affiliation(s)
- Akinori Sato
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
- Division of Breast Surgery, KKR Sapporo Medical Center Tonan Hospital, Sapporo, Japan
| | - Ryuta Kamekura
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Koji Kawata
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masaya Kawada
- Division of Breast Surgery, KKR Sapporo Medical Center Tonan Hospital, Sapporo, Japan
| | - Sumito Jitsukawa
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Keiji Yamashita
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Noriyuki Sato
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tetsuo Himi
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shingo Ichimiya
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- * E-mail:
| |
Collapse
|
97
|
Motojima M, Tanimoto S, Ohtsuka M, Matsusaka T, Kume T, Abe K. Characterization of Kidney and Skeleton Phenotypes of Mice Double Heterozygous for Foxc1 and Foxc2. Cells Tissues Organs 2016; 201:380-9. [PMID: 27193493 DOI: 10.1159/000445027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2016] [Indexed: 11/19/2022] Open
Abstract
Foxc1 and Foxc2 play key roles in mouse development. Foxc1 mutant mice develop duplex kidneys with double ureters, and lack calvarial and sternal bones. Foxc2 null mice have been reported to have glomerular abnormalities in the kidney and axial skeletal anomalies. Expression patterns of Foxc1 and Foxc2 overlap extensively and are believed to have interactive roles. However, cooperative roles of these factors in glomerular and skeletal development are unknown. Therefore, we examined the kidneys and skeleton of mice that were double heterozygous for Foxc1 and Foxc2. Double heterozygotes were generated by mating single heterozygotes for Foxc1 and Foxc2. Newborn double heterozygous mice showed many anomalies in the kidney and urinary tract resembling Foxc1 phenotypes, including duplex kidneys, double ureters, hydronephrosis and mega-ureter. Some mice had hydronephrosis alone. In addition to these macroscopic anomalies, some mice had abnormal glomeruli and disorganized glomerular capillaries observed in Foxc2 phenotypes. Interestingly, these mice also showed glomerular cysts not observed in the single-gene knockout of either Foxc1 or Foxc2 but observed in conditional knockout of Foxc2 in the kidney. Serial section analysis revealed that all cystic glomeruli were connected to proximal tubules, precluding the possibility of atubular glomeruli resulting in cyst formation. Dorsally opened vertebral arches and malformations of sternal bones in the double heterozygotes were phenotypes similar to Foxc1 null mice. Absent or split vertebral bodies in the double heterozygotes were phenotypes similar to Foxc2 null mice, whilst hydrocephalus noted in the Foxc1 phenotype was not observed. Thus, Foxc1 and Foxc2 have a role in kidney and axial skeleton development. These transcription factors might interact in the regulation of the embryogenesis of these organs.
Collapse
|
98
|
Lambers E, Arnone B, Fatima A, Qin G, Wasserstrom JA, Kume T. Foxc1 Regulates Early Cardiomyogenesis and Functional Properties of Embryonic Stem Cell Derived Cardiomyocytes. Stem Cells 2016; 34:1487-500. [DOI: 10.1002/stem.2301] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 12/21/2015] [Accepted: 12/23/2015] [Indexed: 01/16/2023]
Affiliation(s)
- Erin Lambers
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University; Chicago Illinois USA
| | - Baron Arnone
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University; Chicago Illinois USA
| | - Anees Fatima
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University; Chicago Illinois USA
| | - Gangjian Qin
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University; Chicago Illinois USA
| | - J. Andrew Wasserstrom
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University; Chicago Illinois USA
| | - Tsutomu Kume
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University; Chicago Illinois USA
| |
Collapse
|
99
|
Mayeuf-Louchart A, Montarras D, Bodin C, Kume T, Vincent SD, Buckingham M. Endothelial cell specification in the somite is compromised in Pax3-positive progenitors of Foxc1/2 conditional mutants, with loss of forelimb myogenesis. Development 2016; 143:872-9. [PMID: 26839363 DOI: 10.1242/dev.128017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/23/2016] [Indexed: 11/20/2022]
Abstract
Pax3 and Foxc2 have been shown genetically to mutually repress each other in the mouse somite. Perturbation of this balance in multipotent cells of the dermomyotome influences cell fate; upregulation of Foxc2 favours a vascular fate, whereas higher levels of Pax3 lead to myogenesis. Foxc1 has overlapping functions with Foxc2. In Foxc1/2 double-mutant embryos, somitogenesis is severely affected, precluding analysis of somite derivatives. We have adopted a conditional approach whereby mutations in Foxc1 and Foxc2 genes were targeted to Pax3-expressing cells. Inclusion of a conditional reporter allele in the crosses made it possible to follow cells that had expressed Pax3. At the forelimb level, endothelial and myogenic cells migrate from adjacent somites into the limb bud. This population of endothelial cells is compromised in the double mutant, whereas excessive production of myogenic cells is observed in the trunk. However, strikingly, myogenic progenitors fail to enter the limbs, leading to the absence of skeletal muscle. Pax3-positive migratory myogenic progenitors, marked by expression of Lbx1, are specified in the somite at forelimb level, but endothelial progenitors are absent. The myogenic progenitors do not die, but differentiate prematurely adjacent to the somite. We conclude that the small proportion of somite-derived endothelial cells in the limb is required for the migration of myogenic limb progenitors.
Collapse
Affiliation(s)
- Alicia Mayeuf-Louchart
- CNRS UMR 3738, Department of Developmental and Stem Cell Biology, Institut Pasteur, 28 Rue du Dr Roux, Paris 75015, France
| | - Didier Montarras
- CNRS UMR 3738, Department of Developmental and Stem Cell Biology, Institut Pasteur, 28 Rue du Dr Roux, Paris 75015, France
| | - Catherine Bodin
- CNRS UMR 3738, Department of Developmental and Stem Cell Biology, Institut Pasteur, 28 Rue du Dr Roux, Paris 75015, France
| | - Tsutomu Kume
- Feinberg Cardiovascular Research Institute, Northwestern University School of Medicine, Chicago, IL 60611, USA
| | - Stéphane D Vincent
- CNRS UMR 3738, Department of Developmental and Stem Cell Biology, Institut Pasteur, 28 Rue du Dr Roux, Paris 75015, France
| | - Margaret Buckingham
- CNRS UMR 3738, Department of Developmental and Stem Cell Biology, Institut Pasteur, 28 Rue du Dr Roux, Paris 75015, France
| |
Collapse
|
100
|
Sanchez-Castro M, Eldjouzi H, Charpentier E, Busson PF, Hauet Q, Lindenbaum P, Delasalle-Guyomarch B, Baudry A, Pichon O, Pascal C, Lefort B, Bajolle F, Pezard P, Schott JJ, Dina C, Redon R, Gournay V, Bonnet D, Le Caignec C. Search for Rare Copy-Number Variants in Congenital Heart Defects Identifies Novel Candidate Genes and a Potential Role for FOXC1 in Patients With Coarctation of the Aorta. ACTA ACUST UNITED AC 2016; 9:86-94. [DOI: 10.1161/circgenetics.115.001213] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 12/03/2015] [Indexed: 12/16/2022]
Abstract
Background—
Congenital heart defects are the most frequent malformations among newborns and a frequent cause of morbidity and mortality. Although genetic variation contributes to congenital heart defects, their precise molecular bases remain unknown in the majority of patients.
Methods and Results—
We analyzed, by high-resolution array comparative genomic hybridization, 316 children with sporadic, nonsyndromic congenital heart defects, including 76 coarctation of the aorta, 159 transposition of the great arteries, and 81 tetralogy of Fallot, as well as their unaffected parents. We identified by array comparative genomic hybridization, and validated by quantitative real-time polymerase chain reaction, 71 rare de novo (n=8) or inherited (n=63) copy-number variants (CNVs; 50 duplications and 21 deletions) in patients. We identified 113 candidate genes for congenital heart defects within these CNVs, including
BTRC
,
CHRNB3
,
CSRP2BP
,
ERBB2
,
ERMARD
,
GLIS3
,
PLN
,
PTPRJ
,
RLN3
, and
TCTE3
. No de novo CNVs were identified in patients with transposition of the great arteries in contrast to coarctation of the aorta and tetralogy of Fallot (
P
=0.002; Fisher exact test). A search for transcription factor binding sites showed that 93% of the rare CNVs identified in patients with coarctation of the aorta contained at least 1 gene with FOXC1-binding sites. This significant enrichment (
P
<0.0001; permutation test) was not observed for the CNVs identified in patients with transposition of the great arteries and tetralogy of Fallot. We hypothesize that these CNVs may alter the expression of genes regulated by FOXC1. Foxc1 belongs to the forkhead transcription factors family, which plays a critical role in cardiovascular development in mice.
Conclusions—
These data suggest that deregulation of
FOXC1
or its downstream genes play a major role in the pathogenesis of coarctation of the aorta in humans.
Collapse
|