51
|
Larabee SM, Coia H, Jones S, Cheung E, Gallicano GI. miRNA-17 members that target Bmpr2 influence signaling mechanisms important for embryonic stem cell differentiation in vitro and gastrulation in embryos. Stem Cells Dev 2014; 24:354-71. [PMID: 25209090 DOI: 10.1089/scd.2014.0051] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Body axes and germ layers evolve at gastrulation, and in mammals are driven by many genes; however, what orchestrates the genetic pathways during gastrulation remains elusive. Previously, we presented evidence that microRNA-17 (miRNA-17) family members, miR-17-5p, miR-20a, miR-93, and miR-106a were differentially expressed in mouse embryos and functioned to control differentiation of the stem cell population. Here, we identify function(s) that these miRNAs have during gastrulation. Fluorescent in situ hybridization miRNA probes reveal that these miRNAs are localized at the mid/posterior primitive streak (ps) in distinct populations of primitive ectoderm, mesendoderm, and mesoderm. Seven different miRNA prediction algorithms are identified in silico bone morphogenic protein receptor 2 (Bmpr2) as a target of these miRNAs. Bmpr2 is a member of the TGFβ pathway and invokes stage-specific changes during gastrulation. Recently, Bmpr2 was shown regulating cytoskeletal dynamics, cell movement, and invasion. Our previous and current data led to a hypothesis by which members of the miR-17 family influence gastrulation by suppressing Bmpr2 expression at the primitive streak. This suppression influences fate decisions of cells by affecting genes downstream of BMPR2 as well as mesoderm invasion through regulation of actin dynamics.
Collapse
Affiliation(s)
- Shannon M Larabee
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center , Washington, District of Columbia
| | | | | | | | | |
Collapse
|
52
|
Abstract
Current dogma is that mouse primordial germ cells (PGCs) segregate within the allantois, or source of the umbilical cord, and translocate to the gonads, differentiating there into sperm and eggs. In light of emerging data on the posterior embryonic-extraembryonic interface, and the poorly studied but vital fetal-umbilical connection, we have reviewed the past century of experiments on mammalian PGCs and their relation to the allantois. We demonstrate that, despite best efforts and valuable data on the pluripotent state, what is and is not a PGC in vivo is obscure. Furthermore, sufficient experimental evidence has yet to be provided either for an extragonadal origin of mammalian PGCs or for their segregation within the posterior region. Rather, most evidence points to an alternative hypothesis that PGCs in the mouse allantois are part of a stem/progenitor cell pool that exhibits all known PGC "markers" and that builds/reinforces the fetal-umbilical interface, common to amniotes. We conclude by suggesting experiments to distinguish the mammalian germ line from the soma.
Collapse
|
53
|
Kandyba E, Hazen VM, Kobielak A, Butler SJ, Kobielak K. Smad1 and 5 but not Smad8 establish stem cell quiescence which is critical to transform the premature hair follicle during morphogenesis toward the postnatal state. Stem Cells 2014; 32:534-47. [PMID: 24023003 DOI: 10.1002/stem.1548] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Accepted: 08/05/2013] [Indexed: 11/11/2022]
Abstract
Hair follicles (HFs) are regenerative miniorgans that offer a highly informative model system to study the regulatory mechanisms of hair follicle stem cells (hfSCs) homeostasis and differentiation. Bone morphogenetic protein (BMP) signaling is key in both of these processes, governing hfSCs quiescence in the bulge and differentiation of matrix progenitors. However, whether canonical or noncanonical pathways of BMP signaling are responsible for these processes remains unresolved. Here, we conditionally ablated two canonical effectors of BMP signaling, Smad1 and Smad5 during hair morphogenesis and postnatal cycling in mouse skin. Deletion of Smad1 and Smad5 (dKO) in the epidermis during morphogenesis resulted in neonatal lethality with lack of visible whiskers. Interestingly, distinct patterns of phospho-Smads (pSmads) activation were detected with pSmad8 restricted to epidermis and pSmad1 and pSmad5 exclusively activated in HFs. Engraftment of dKO skin revealed retarded hair morphogenesis and failure to differentiate into visible hair. The formation of the prebulge and bulge reservoir for quiescent hfSCs was precluded in dKO HFs which remained in prolonged anagen. Surprisingly, in postnatal telogen HFs, pSmad8 expression was no longer limited to epidermis and was also present in dKO bulge hfSCs and matrix progenitors. Although pSmad8 activity alone could not prevent dKO hfSCs precocious anagen activation, it sustained efficient postnatal differentiation and regeneration of visible hairs. Together, our data suggest a pivotal role for canonical BMP signaling demonstrating distinguished nonoverlapping function of pSmad8 with pSmad1 and pSmad5 in hfSCs regulation and hair morphogenesis but a redundant role in adult hair progenitors differentiation.
Collapse
Affiliation(s)
- Eve Kandyba
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research and Department of Biochemistry and Molecular Biology, USC Norris Cancer Center, University of Southern California, Los Angeles, California, USA; Department of Pathology, Department of Biochemistry and Molecular Biology, USC Norris Cancer Center, University of Southern California, Los Angeles, California, USA
| | | | | | | | | |
Collapse
|
54
|
Lee HC, Lin YZ, Lai YT, Huang WJ, Hu JR, Tsai JN, Tsai HJ. Glycogen synthase kinase 3 beta in somites plays a role during the angiogenesis of zebrafish embryos. FEBS J 2014; 281:4367-83. [PMID: 25056693 DOI: 10.1111/febs.12942] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 07/15/2014] [Accepted: 07/21/2014] [Indexed: 12/12/2022]
Abstract
Glycogen synthase kinase 3 beta (Gsk3b) acts as a negative modulator in endothelial cells through the Wnt/β-catenin/PI3K/AKT/Gsk3b axis in cancer-induced angiogenesis. However, the function of Gsk3b during embryonic angiogenesis remains unclear. Here, either gsk3b knockdown by morpholino or Gsk3b loss of activity by LiCl treatment had serious phenotypic consequences, such as defects in the positioning and patterning of intersegmental blood vessels and reduction of vegfaa121 and vegfaa165 transcripts. In embryos treated with the phosphatidylinositol 3-kinase inhibitor, angiogenesis was severely inhibited, along with reduced Wnt, phosphorylated AKT and phosphorylated Gsk3b, suggesting that the remaining Gsk3b in somites could still degrade β-catenin, resulting in decreased vascular endothelial growth factor Aa(VegfAa) expression. However, in gsk3b-mRNA-overexpressed embryos, intersegmental vessels ectopically sprouted by the increase in phosphorylated-Gsk3b which prevented the degradation of β-catenin and promoted the increase in phosphorylated AKT activity, thus increasing VegfAa expression in somites. Interestingly, the Gsk3b-dependent cross-talk between PI3K/AKT and Wnt/β-catenin suggests that Wnt/β-catenin and PI3K/AKT interaction controls embryonic angiogenesis by a positive feedback loop rather than a hierarchical framework such as that found in cancer-induced angiogenesis. Thus, both active and inactive forms of Gsk3b mediate the cooperative signaling between Wnt/β-catenin and PI3K/AKT to control VegfAa expression in somites during angiogenesis in zebrafish embryos.
Collapse
Affiliation(s)
- Hung-Chieh Lee
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
55
|
An updated view on the differentiation of stem cells into endothelial cells. SCIENCE CHINA-LIFE SCIENCES 2014; 57:763-73. [DOI: 10.1007/s11427-014-4712-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 06/16/2014] [Indexed: 12/16/2022]
|
56
|
BMP signaling balances murine myeloid potential through SMAD-independent p38MAPK and NOTCH pathways. Blood 2014; 124:393-402. [PMID: 24894772 DOI: 10.1182/blood-2014-02-556993] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Bone morphogenetic protein (BMP) signaling regulates early hematopoietic development, proceeding from mesoderm patterning through the progressive commitment and differentiation of progenitor cells. The BMP pathway signals largely through receptor-mediated activation of Mothers Against Decapentaplegic homolog (SMAD) proteins, although alternate pathways are modulated through various components of mitogen-activated protein kinase (MAPK) signaling. Using a conditional, short hairpin RNA (shRNA)-based knockdown system in the context of differentiating embryonic stem cells (ESCs), we demonstrated previously that Smad1 promotes hemangioblast specification, but then subsequently restricts primitive progenitor potential. Here we show that co-knockdown of Smad5 restores normal progenitor potential of Smad1-depleted cells, suggesting opposing functions for Smad1 and Smad5. This balance was confirmed by cotargeting Smad1/5 with a specific chemical antagonist, LDN193189 (LDN). However, we discovered that LDN treatment after hemangioblast commitment enhanced primitive myeloid potential. Moreover, inhibition with LDN (but not SMAD depletion) increased expression of Delta-like ligands Dll1 and Dll3 and NOTCH activity; abrogation of NOTCH activity restored LDN-enhanced myeloid potential back to normal, corresponding with expression levels of the myeloid master regulator, C/EBPα. LDN but not SMAD activity was also associated with activation of the p38MAPK pathway, and blocking this pathway was sufficient to enhance myelopoiesis. Therefore, NOTCH and p38MAPK pathways balance primitive myeloid progenitor output downstream of the BMP pathway.
Collapse
|
57
|
Jiang X, Sung YK, Tian W, Qian J, Semenza GL, Nicolls MR. Graft microvascular disease in solid organ transplantation. J Mol Med (Berl) 2014; 92:797-810. [PMID: 24880953 PMCID: PMC4118041 DOI: 10.1007/s00109-014-1173-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 04/29/2014] [Accepted: 05/14/2014] [Indexed: 02/07/2023]
Abstract
Alloimmune inflammation damages the microvasculature of solid organ transplants during acute rejection. Although immunosuppressive drugs diminish the inflammatory response, they do not directly promote vascular repair. Repetitive microvascular injury with insufficient regeneration results in prolonged tissue hypoxia and fibrotic remodeling. While clinical studies show that a loss of the microvascular circulation precedes and may act as an initiating factor for the development of chronic rejection, preclinical studies demonstrate that improved microvascular perfusion during acute rejection delays and attenuates tissue fibrosis. Therefore, preservation of a functional microvasculature may represent an effective therapeutic strategy for preventing chronic rejection. Here, we review recent advances in our understanding of the role of the microvasculature in the long-term survival of transplanted solid organs. We also highlight microvessel-centered therapeutic strategies for prolonging the survival of solid organ transplants.
Collapse
Affiliation(s)
- Xinguo Jiang
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA,
| | | | | | | | | | | |
Collapse
|
58
|
Flores LE, Hildebrandt TB, Kühl AA, Drews B. Early detection and staging of spontaneous embryo resorption by ultrasound biomicroscopy in murine pregnancy. Reprod Biol Endocrinol 2014; 12:38. [PMID: 24886361 PMCID: PMC4037759 DOI: 10.1186/1477-7827-12-38] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 04/26/2014] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Embryo resorption is a major problem in human medicine, agricultural animal production and in conservation breeding programs. Underlying mechanisms have been investigated in the well characterised mouse model. However, post mortem studies are limited by the rapid disintegration of embryonic structures. A method to reliably identify embryo resorption in alive animals has not been established yet. In our study we aim to detect embryos undergoing resorption in vivo at the earliest possible stage by ultra-high frequency ultrasound. METHODS In a longitudinal study, we monitored 30 pregnancies of wild type C57BI/6 mice using ultra-high frequency ultrasound (30-70 MHz), so called ultrasound biomicroscopy (UBM). We compared the sonoembryology of mouse conceptuses under spontaneous resorption and neighbouring healthy conceptuses and correlated the live ultrasound data with the respective histology. RESULTS The process of embryo resorption comprised of four stages: first, the conceptus exhibited growth retardation, second, bradycardia and pericardial edema were observed, third, further development ceased and the embryo died, and finally embryo remnants were resorbed by maternal immune cells. In early gestation (day 7 and 8), growth retardation was characterized by a small embryonic cavity. The embryo and its membranes were ill defined or did not develop at all. The echodensity of the embryonic fluid increased and within one to two days, the embryo and its cavity disappeared and was transformed into echodense tissue surrounded by fluid filled caverns. In corresponding histologic preparations, fibrinoid material interspersed with maternal granulocytes and lacunae filled with maternal blood were observed. In later stages (day 9-11) resorption prone embryos were one day behind in their development compared to their normal siblings. The space between Reichert's membrane and inner yolk sac membrane was enlarged The growth retarded embryos exhibited bradycardia and ultimately cessation of heart beat. Corresponding histology showed apoptotic cells in the embryo while the placenta was still intact. In the subsequent resorption process first the embryo and then its membranes disappeared. CONCLUSIONS Our results provide a temporal time course of embryo resorption. With this method, animals exhibiting embryo resorption can be targeted, enabling the investigation of underlying mechanisms before the onset of total embryo disintegration.
Collapse
Affiliation(s)
- Luis E Flores
- Department Reproduction Management, Leibniz Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Str. 17, 10315 Berlin, Germany
| | - Thomas B Hildebrandt
- Department Reproduction Management, Leibniz Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Str. 17, 10315 Berlin, Germany
| | - Anja A Kühl
- Charité – Department of Medicine I for Gastroenterology, Infectious Disease and Rheumatology, Research Center ImmunoSciences / Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Barbara Drews
- Department Reproduction Management, Leibniz Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Str. 17, 10315 Berlin, Germany
| |
Collapse
|
59
|
Kumar A, Lualdi M, Loncarek J, Cho YW, Lee JE, Ge K, Kuehn MR. Loss of function of mouse Pax-Interacting Protein 1-associated glutamate rich protein 1a (Pagr1a) leads to reduced Bmp2 expression and defects in chorion and amnion development. Dev Dyn 2014; 243:937-47. [PMID: 24633704 DOI: 10.1002/dvdy.24125] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 03/07/2014] [Accepted: 03/09/2014] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Human PAX-Interacting Protein 1 (PAXIP1)-associated glutamate rich protein 1 (PAGR1, also known as PA1) originally was discovered as part of a complex containing PAXIP1 and histone H3K4 methyltransferases MLL3 and MLL4, suggesting a role in epigenetic gene regulation. Further in vitro studies suggested additional functions in DNA damage repair and transcription. However, in vivo analysis of PAGR1 function has been lacking. RESULTS Here we show that expression of the cognate mouse gene Pagr1a is found predominately in the extraembryonic and chorionic ectoderm from pregastrulation stages and is up-regulated within the embryo proper after gastrulation. Embryos with a germ line deletion of Pagr1a establish the anterior-posterior axis, and show normal neuroectodermal, mesodermal, and endodermal patterning, but fail to develop beyond the four- to five-somite stage or to undergo axial rotation. Pagr1a(-) (/) (-) embryos also show abnormal development of extraembryonic tissues with defects seen in the amnion, chorion and visceral yolk sac. At the molecular level, Pagr1a(-) (/) (-) embryos have reduced expression of BMP2, a known regulator of extraembryonic development. CONCLUSIONS Loss of mouse Pagr1a function leads to defective extraembryonic development, likely due at least in part to altered BMP signaling, contributing to developmental arrest.
Collapse
Affiliation(s)
- Amit Kumar
- Laboratory of Protein Dynamics and Signaling, National Cancer Institute, National Institutes of Health, Frederick, Maryland
| | | | | | | | | | | | | |
Collapse
|
60
|
Wei CY, Wang HP, Zhu ZY, Sun YH. Transcriptional factors smad1 and smad9 act redundantly to mediate zebrafish ventral specification downstream of smad5. J Biol Chem 2014; 289:6604-6618. [PMID: 24488494 DOI: 10.1074/jbc.m114.549758] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) are multifunctional growth factors that play crucial roles during embryonic development and cell fate determination. Nuclear transduction of BMP signals requires the receptor type Smad proteins, Smad1, Smad5, and Smad9. However, how these Smad proteins cooperate in vivo to regulate various developmental processes is largely unknown. In zebrafish, it was widely believed that the maternally expressed smad5 is essential for dorso-ventral (DV) patterning, and the zygotically transcribed smad1 is not required for normal DV axis establishment. In the present study, we have identified zygotically expressed smad9, which cooperates with smad1 downstream of smad5, to mediate zebrafish early DV patterning in a functional redundant manner. Although knockdown of smad1 or smad9 alone does not lead to visible dorsalization, double knockdown strongly dorsalizes zebrafish embryos, which cannot be efficiently rescued by smad5 overexpression, whereas the dorsalization induced by smad5 knockdown can be fully rescued by overexpression of smad1 or smad9. We have further revealed that the transcription initiations of smad1 and smad9 are repressed by each other, that they are direct transcriptional targets of Smad5, and that smad9, like smad1, is required for myelopoiesis. In conclusion, our study uncovers that smad1 and smad9 act redundantly to each other downstream of smad5 to mediate ventral specification and to regulate embryonic myelopoiesis.
Collapse
Affiliation(s)
- Chang-Yong Wei
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, 7 Donghu South Road, Wuhan 430072, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Hou-Peng Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, 7 Donghu South Road, Wuhan 430072, China
| | - Zuo-Yan Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, 7 Donghu South Road, Wuhan 430072, China
| | - Yong-Hua Sun
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, 7 Donghu South Road, Wuhan 430072, China.
| |
Collapse
|
61
|
Jorgensen JS. Defining the neighborhoods that escort the oocyte through its early life events and into a functional follicle. Mol Reprod Dev 2013; 80:960-76. [PMID: 24105719 PMCID: PMC3980676 DOI: 10.1002/mrd.22232] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 08/15/2013] [Indexed: 01/19/2023]
Abstract
The ovary functions to chaperone the most precious cargo for female individuals, the oocyte, thereby allowing the passage of genetic material to subsequent generations. Within the ovary, single oocytes are surrounded by a legion of granulosa cells inside each follicle. These two cell types depend upon one another to support follicle formation and oocyte survival. The infrastructure and events that work together to ultimately form these functional follicles within the ovary are unprecedented, given that the oocyte originates as a cell like all other neighboring cells within the embryo prior to gastrulation. This review discusses the journey of the germ cell in the context of the developing female mouse embryo, with a focus on specific signaling events and cell-cell interactions that escort the primordial germ cell as it is specified into the germ cell fate, migrates through the hindgut into the gonad, differentiates into an oocyte, and culminates upon formation of the primordial and then primary follicle.
Collapse
Affiliation(s)
- Joan S Jorgensen
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
62
|
Anti-Müllerian hormone recruits BMPR-IA in immature granulosa cells. PLoS One 2013; 8:e81551. [PMID: 24312319 PMCID: PMC3842941 DOI: 10.1371/journal.pone.0081551] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 10/23/2013] [Indexed: 01/24/2023] Open
Abstract
Anti-Müllerian hormone (AMH) is a member of the TGF-β superfamily secreted by the gonads of both sexes. This hormone is primarily known for its role in the regression of the Müllerian ducts in male fetuses. In females, AMH is expressed in granulosa cells of developing follicles. Like other members of the TGF-β superfamily, AMH transduces its signal through two transmembrane serine/threonine kinase receptors including a well characterized type II receptor, AMHR-II. The complete signalling pathway of AMH involving Smads proteins and the type I receptor is well known in the Müllerian duct and in Sertoli and Leydig cells but not in granulosa cells. In addition, few AMH target genes have been identified in these cells. Finally, while several co-receptors have been reported for members of the TGF-β superfamily, none have been described for AMH. Here, we have shown that none of the Bone Morphogenetic Proteins (BMPs) co-receptors, Repulsive guidance molecules (RGMs), were essential for AMH signalling. We also demonstrated that the main Smad proteins used by AMH in granulosa cells were Smad 1 and Smad 5. Like for the other AMH target cells, the most important type I receptor for AMH in these cells was BMPR-IA. Finally, we have identified a new AMH target gene, Id3, which could be involved in the effects of AMH on the differentiation of granulosa cells and its other target cells.
Collapse
|
63
|
Abstract
Thalidomide remains one of the world’s most notorious drugs due to the severe birth defects it induced in children between 1957 and 1962. Yet, to some this drug is a lifesaver, as it now enjoys renaissance in the treatment for a wide range of conditions including leprosy, multiple myeloma, Behcet’s disease, and some cancers. However, thalidomide has also been linked to causing a new generation of thalidomide survivors in Brazil, where the drug is used to treat leprosy. Surprisingly how thalidomide causes birth defects and how it acts in the treatment of clinical conditions are still far from clear. In the past decade great strides in our understanding of the actions of the drug, as well as molecular targets, have been made. The purpose of this review is to look at the recent work carried out into understanding how thalidomide causes birth defects, it’s molecular targets and the challenges that remain to be elucidated. These challenges include identifying clinically relevant but nonteratogenic forms of the drug, and the mechanisms underlying phocomelia and species specificity.
Collapse
|
64
|
McCormack N, O'Dea S. Regulation of epithelial to mesenchymal transition by bone morphogenetic proteins. Cell Signal 2013; 25:2856-62. [PMID: 24044921 DOI: 10.1016/j.cellsig.2013.09.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 09/02/2013] [Accepted: 09/06/2013] [Indexed: 02/08/2023]
Abstract
Epithelial to mesenchymal transition (EMT) is a process in which fully differentiated epithelial cells lose many of their epithelial characteristics and adopt features typical of mesenchymal cells, thus allowing cells to become migratory and invasive. EMT is a critical process in development and its role in cancer and fibrosis is becoming increasingly recognised. It is also becoming apparent that EMT is not just restricted to embryonic development and disease in adults, but in fact may be an important process for the maintenance and regeneration of adult tissue architecture. While transforming growth factor-β (TGF-β) is considered a prototypic inducer of EMT, relatively little is known about other signalling molecules that regulate EMT. Bone morphogenic proteins (BMPs) are members of the TGF-β superfamily and 20 different human BMPs have been identified. Originally named for their effects on bone, these proteins are now considered to be key morphogenetic signals that orchestrate tissue architecture throughout the body. BMP2, -4 and -7 are the best studied to date. There are disparate reports of the roles of BMPs in EMT during development, cancer and fibrosis. Here, we present an overview of this literature as well as the emerging role of EMT in tissue regeneration and the involvement of BMPs in regulating this process.
Collapse
Affiliation(s)
- Natasha McCormack
- Institute of Immunology, National University of Ireland Maynooth, Ireland.
| | | |
Collapse
|
65
|
Garside VC, Chang AC, Karsan A, Hoodless PA. Co-ordinating Notch, BMP, and TGF-β signaling during heart valve development. Cell Mol Life Sci 2013; 70:2899-917. [PMID: 23161060 PMCID: PMC4996658 DOI: 10.1007/s00018-012-1197-9] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 10/12/2012] [Accepted: 10/15/2012] [Indexed: 12/22/2022]
Abstract
Congenital heart defects affect approximately 1-5 % of human newborns each year, and of these cardiac defects 20-30 % are due to heart valve abnormalities. Recent literature indicates that the key factors and pathways that regulate valve development are also implicated in congenital heart defects and valve disease. Currently, there are limited options for treatment of valve disease, and therefore having a better understanding of valve development can contribute critical insight into congenital valve defects and disease. There are three major signaling pathways required for early specification and initiation of endothelial-to-mesenchymal transformation (EMT) in the cardiac cushions: BMP, TGF-β, and Notch signaling. BMPs secreted from the myocardium set up the environment for the overlying endocardium to become activated; Notch signaling initiates EMT; and both BMP and TGF-β signaling synergize with Notch to promote the transition of endothelia to mesenchyme and the mesenchymal cell invasiveness. Together, these three essential signaling pathways help form the cardiac cushions and populate them with mesenchyme and, consequently, set off the cascade of events required to develop mature heart valves. Furthermore, integration and cross-talk between these pathways generate highly stratified and delicate valve leaflets and septa of the heart. Here, we discuss BMP, TGF-β, and Notch signaling pathways during mouse cardiac cushion formation and how they together produce a coordinated EMT response in the developing mouse valves.
Collapse
Affiliation(s)
- Victoria C. Garside
- Terry Fox Laboratory, BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC V5Z 1L3 Canada
- Cell and Developmental Biology Program, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
| | - Alex C. Chang
- Michael Smith Genome Sciences Centre, BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC V5Z 1L3 Canada
| | - Aly Karsan
- Michael Smith Genome Sciences Centre, BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC V5Z 1L3 Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
| | - Pamela A. Hoodless
- Terry Fox Laboratory, BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC V5Z 1L3 Canada
- Cell and Developmental Biology Program, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
| |
Collapse
|
66
|
Hegarty SV, O'Keeffe GW, Sullivan AM. BMP-Smad 1/5/8 signalling in the development of the nervous system. Prog Neurobiol 2013; 109:28-41. [PMID: 23891815 DOI: 10.1016/j.pneurobio.2013.07.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 07/16/2013] [Accepted: 07/16/2013] [Indexed: 02/07/2023]
Abstract
The transcription factors, Smad1, Smad5 and Smad8, are the pivotal intracellular effectors of the bone morphogenetic protein (BMP) family of proteins. BMPs and their receptors are expressed in the nervous system (NS) throughout its development. This review focuses on the actions of Smad 1/5/8 in the developing NS. The mechanisms by which these Smad proteins regulate the induction of the neuroectoderm, the central nervous system (CNS) primordium, and finally the neural crest, which gives rise to the peripheral nervous system (PNS), are reviewed herein. We describe how, following neural tube closure, the most dorsal aspect of the tube becomes a signalling centre for BMPs, which directs the pattern of the development of the dorsal spinal cord (SC), through the action of Smad1, Smad5 and Smad8. The direct effects of Smad 1/5/8 signalling on the development of neuronal and non-neuronal cells from various neural progenitor cell populations are then described. Finally, this review discusses the neurodevelopmental abnormalities associated with the knockdown of Smad 1/5/8.
Collapse
Affiliation(s)
- Shane V Hegarty
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
| | | | | |
Collapse
|
67
|
Abstract
The circulatory system is the first organ system to develop in the vertebrate embryo and is critical throughout gestation for the delivery of oxygen and nutrients to, as well as removal of metabolic waste products from, growing tissues. Endothelial cells, which constitute the luminal layer of all blood and lymphatic vessels, emerge de novo from the mesoderm in a process known as vasculogenesis. The vascular plexus that is initially formed is then remodeled and refined via proliferation, migration, and sprouting of endothelial cells to form new vessels from preexisting ones during angiogenesis. Mural cells are also recruited by endothelial cells to form the surrounding vessel wall. During this vascular remodeling process, primordial endothelial cells are specialized to acquire arterial, venous, and blood-forming hemogenic phenotypes and functions. A subset of venous endothelium is also specialized to become lymphatic endothelium later in development. The specialization of all endothelial cell subtypes requires extrinsic signals and intrinsic regulatory events, which will be discussed in this review.
Collapse
Affiliation(s)
- Kathrina L Marcelo
- Interdepartmental Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA
| | | | | |
Collapse
|
68
|
Gavrilov S, Lacy E. Genetic dissection of ventral folding morphogenesis in mouse: embryonic visceral endoderm-supplied BMP2 positions head and heart. Curr Opin Genet Dev 2013; 23:461-9. [PMID: 23706163 DOI: 10.1016/j.gde.2013.04.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 04/03/2013] [Indexed: 01/08/2023]
Abstract
Ventral folding morphogenesis (VFM), a vital morphogenetic process in amniotes, mediates gut endoderm internalization, linear heart tube formation, ventral body wall closure and encasement of the fetus in extraembryonic membranes. Aberrant VFM underlies a number of birth defects such as gastroschisis and ectopia cordis in human and misplacement of head and heart in mouse. Recent cell lineage-specific mouse mutant analyses identified the Bone Morphogenetic Protein (BMP) pathway and Anterior Visceral Endoderm (AVE) as key regulators of anterior VFM. Loss of BMP2 expression solely from embryonic visceral endoderm (EmVE) and the AVE blocks formation of foregut invagination, and simultaneously, aberrantly positions the heart anterior/dorsal to the head, suggesting a mechanistic link between foregut and head/heart morphogenesis.
Collapse
Affiliation(s)
- Svetlana Gavrilov
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY 10065, USA
| | | |
Collapse
|
69
|
Islam SS, Mokhtari RB, Kumar S, Maalouf J, Arab S, Yeger H, Farhat WA. Spatio-temporal distribution of Smads and role of Smads/TGF-β/BMP-4 in the regulation of mouse bladder organogenesis. PLoS One 2013; 8:e61340. [PMID: 23620745 PMCID: PMC3631207 DOI: 10.1371/journal.pone.0061340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 03/07/2013] [Indexed: 11/24/2022] Open
Abstract
Although Shh, TGF-β and BMP-4 regulate radial patterning of the bladder mesenchyme and smooth muscle differentiation, it is not known what transcription factors, local environmental cues or signaling cascades mediate bladder smooth muscle differentiation. We investigated the expression patterns of signaling mediated by Smad2 and Smad3 in the mouse embryonic bladder from E12.5 to E16.5 by using qRT-PCR, in situ hybridization and antibodies specifically recognizing individual Smad proteins. The role of Smad2 and Smad3 during smooth muscle formation was examined by disrupting the Smad2/3 signaling pathway using TβR1 inhibitor SB-431542 in organ culture system. qRT-PCR results showed that R-Smads, Co-Smad and I-Smads were all expressed during bladder development. RNA ISH for BMP-4 and immunostaining of TGF-β1 showed that BMP-4 and TGF-β1 were expressed in the transitional epithelium, lamina propia and muscularis mucosa. Smad1, Smad5 and Smad8 were first expressed in the bladder epithelium and continued to be expressed in the transitional epithelium, muscularis mesenchyme and lamina propia as the bladder developed. Smad2, Smad3 and Smad4 were first detected in the bladder epithelium and subsequently were expressed in the muscularis mesenchyme and lamina propia. Smad6 and Smad7 showed overlapping expression with R-Smads, which are critical for bladder development. In bladder explants (E12.5 to E16.5) culture, Smad2 and Smad3 were found localized within the nuclei, suggesting critical transcriptional regulatory effects during bladder development. E12.5 to E16.5 bladders were cultured with and without TβR1 inhibitor SB-431542 and assessed by qRT-PCR and immunofluorescence. After three days in culture in SB-431542, α-SMA, Smad2 and Smad3 expressions were significantly decreased compared with controls, however, with no significant changes in the expression of smooth muscle myosin heavy chain (SM-Myh. Based on the Smad expression patterns, we suggest that individual or combinations of Smads may be necessary during mouse bladder organogenesis and may be critical mediators for bladder smooth muscle differentiation.
Collapse
Affiliation(s)
- Syed S. Islam
- Developmental and Stem Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Reza Bayat Mokhtari
- Department of Pediatric Laboratory Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Sushil Kumar
- Physiology and Experimental Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | | | - Sara Arab
- University of Toronto, Toronto, ON, Canada
| | - Herman Yeger
- Department of Pediatric Laboratory Medicine, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Walid A. Farhat
- Developmental and Stem Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| |
Collapse
|
70
|
Bone morphogenetic protein type I receptor antagonists decrease growth and induce cell death of lung cancer cell lines. PLoS One 2013; 8:e61256. [PMID: 23593444 PMCID: PMC3625205 DOI: 10.1371/journal.pone.0061256] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 03/11/2013] [Indexed: 11/19/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are highly conserved morphogens that are essential for normal development. BMP-2 is highly expressed in the majority of non-small cell lung carcinomas (NSCLC) but not in normal lung tissue or benign lung tumors. The effects of the BMP signaling cascade on the growth and survival of cancer cells is poorly understood. We show that BMP signaling is basally active in lung cancer cell lines, which can be effectively inhibited with selective antagonists of the BMP type I receptors. Lung cancer cell lines express alk2, alk3, and alk6 and inhibition of a single BMP receptor was not sufficient to decrease signaling. Inhibition of more than one type I receptor was required to decrease BMP signaling in lung cancer cell lines. BMP receptor antagonists and silencing of BMP type I receptors with siRNA induced cell death, inhibited cell growth, and caused a significant decrease in the expression of inhibitor of differentiation (Id1, Id2, and Id3) family members, which are known to regulate cell growth and survival in many types of cancers. BMP receptor antagonists also decreased clonogenic cell growth. Knockdown of Id3 significantly decreased cell growth and induced cell death of lung cancer cells. H1299 cells stably overexpressing Id3 were resistant to growth suppression and induction of cell death induced by the BMP antagonist DMH2. These studies suggest that BMP signaling promotes cell growth and survival of lung cancer cells, which is mediated through its regulation of Id family members. Selective antagonists of the BMP type I receptors represents a potential means to pharmacologically treat NSCLC and other carcinomas with an activated BMP signaling cascade.
Collapse
|
71
|
Lebrun JJ. The Dual Role of TGFβ in Human Cancer: From Tumor Suppression to Cancer Metastasis. ISRN MOLECULAR BIOLOGY 2012; 2012:381428. [PMID: 27340590 PMCID: PMC4899619 DOI: 10.5402/2012/381428] [Citation(s) in RCA: 254] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 10/12/2012] [Indexed: 12/31/2022]
Abstract
The transforming growth factor-beta (TGFβ) superfamily encompasses widespread and evolutionarily conserved polypeptide growth factors that regulate and orchestrate growth and differentiation in all cell types and tissues. While they regulate asymmetric cell division and cell fate determination during early development and embryogenesis, TGFβ family members play a major regulatory role in hormonal and immune responses, cell growth, cell death and cell immortalization, bone formation, tissue remodeling and repair, and erythropoiesis throughout adult life. The biological and physiological functions of TGFβ, the founding member of this family, and its receptors are of central importance to human diseases, particularly cancer. By regulating cell growth, death, and immortalization, TGFβ signaling pathways exert tumor suppressor effects in normal cells and early carcinomas. Thus, it is not surprising that a high number of human tumors arise due to mutations or deletions in the genes coding for the various TGFβ signaling components. As tumors develop and progress, these protective and cytostatic effects of TGFβ are often lost. TGFβ signaling then switches to promote cancer progression, invasion, and tumor metastasis. The molecular mechanisms underlying this dual role of TGFβ in human cancer will be discussed in depth in this paper, and it will highlight the challenge and importance of developing novel therapeutic strategies specifically aimed at blocking the prometastatic arm of the TGFβ signaling pathway without affecting its tumor suppressive effects.
Collapse
Affiliation(s)
- Jean-Jacques Lebrun
- Division of Medical Oncology, Department of Medicine, Royal Victoria Hospital, McGill University Health Center, Montreal, QC, Canada H3A 1A1
| |
Collapse
|
72
|
Eom DS, Amarnath S, Agarwala S. Apicobasal polarity and neural tube closure. Dev Growth Differ 2012; 55:164-72. [PMID: 23277919 DOI: 10.1111/dgd.12030] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Revised: 11/10/2012] [Accepted: 11/12/2012] [Indexed: 01/30/2023]
Abstract
During development, a flat neural plate rolls up and closes to form a neural tube. This process, called neural tube closure, is complex and requires morphogenetic events to occur along multiple axes of the neural plate. Recent studies suggest that cell and tissue polarity play a major role in neural tube morphogenesis. While the planar cell polarity pathway is known to be involved in this process, a role for the apicobasal polarity pathway has only recently begun to be elucidated. These studies show that bone morphogenetic proteins can regulate the apicobasal polarity pathway in the neural plate in a cell cycle dependent manner. This dynamically modulates apical junctions in the neural plate, resulting in cell and tissue shape changes that help bend, shape and close the neural tube.
Collapse
Affiliation(s)
- Dae Seok Eom
- Institute for Cell and Molecular Biology, University of Texas at Austin, TX 78712, USA
| | | | | |
Collapse
|
73
|
Marks-Bluth J, Pimanda JE. Cell signalling pathways that mediate haematopoietic stem cell specification. Int J Biochem Cell Biol 2012; 44:2175-84. [DOI: 10.1016/j.biocel.2012.09.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 09/09/2012] [Accepted: 09/10/2012] [Indexed: 01/27/2023]
|
74
|
Pereira PNG, Dobreva MP, Maas E, Cornelis FM, Moya IM, Umans L, Verfaillie CM, Camus A, de Sousa Lopes SMC, Huylebroeck D, Zwijsen A. Antagonism of Nodal signaling by BMP/Smad5 prevents ectopic primitive streak formation in the mouse amnion. Development 2012; 139:3343-54. [PMID: 22912414 DOI: 10.1242/dev.075465] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The strength and spatiotemporal activity of Nodal signaling is tightly controlled in early implantation mouse embryos, including by autoregulation and feedback loops, and involves secreted and intracellular antagonists. These control mechanisms, which are established at the extra-embryonic/embryonic interfaces, are essential for anterior-posterior patterning of the epiblast and correct positioning of the primitive streak. Formation of an ectopic primitive streak, or streak expansion, has previously been reported in mutants lacking antagonists that target Nodal signaling. Here, we demonstrate that loss-of-function of a major bone morphogenetic protein (BMP) effector, Smad5, results in formation of an ectopic primitive streak-like structure in mutant amnion accompanied by ectopic Nodal expression. This suggests that BMP/Smad5 signaling contributes to negative regulation of Nodal. In cultured cells, we find that BMP-activated Smad5 antagonizes Nodal signaling by interfering with the Nodal-Smad2/4-Foxh1 autoregulatory pathway through the formation of an unusual BMP4-induced Smad complex containing Smad2 and Smad5. Quantitative expression analysis supports that ectopic Nodal expression in the Smad5 mutant amnion is induced by the Nodal autoregulatory loop and a slow positive-feedback loop. The latter involves BMP4 signaling and also induction of ectopic Wnt3. Ectopic activation of these Nodal feedback loops in the Smad5 mutant amnion results in the eventual formation of an ectopic primitive streak-like structure. We conclude that antagonism of Nodal signaling by BMP/Smad5 signaling prevents primitive streak formation in the amnion of normal mouse embryos.
Collapse
Affiliation(s)
- Paulo N G Pereira
- Laboratory of Developmental Signaling of the VIB11 Center for the Biology of Disease, VIB, and Center for Human Genetics, KU Leuven, B-3000 Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Kaimakis P, Crisan M, Dzierzak E. The biochemistry of hematopoietic stem cell development. Biochim Biophys Acta Gen Subj 2012; 1830:2395-403. [PMID: 23069720 DOI: 10.1016/j.bbagen.2012.10.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 09/14/2012] [Accepted: 10/04/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND The cornerstone of the adult hematopoietic system and clinical treatments for blood-related disease is the cohort of hematopoietic stem cells (HSC) that is harbored in the adult bone marrow microenvironment. Interestingly, this cohort of HSCs is generated only during a short window of developmental time. In mammalian embryos, hematopoietic progenitor and HSC generation occurs within several extra- and intraembryonic microenvironments, most notably from 'hemogenic' endothelial cells lining the major vasculature. HSCs are made through a remarkable transdifferentiation of endothelial cells to a hematopoietic fate that is long-lived and self-renewable. Recent studies are beginning to provide an understanding of the biochemical signaling pathways and transcription factors/complexes that promote their generation. SCOPE OF REVIEW The focus of this review is on the biochemistry behind the generation of these potent long-lived self-renewing stem cells of the blood system. Both the intrinsic (master transcription factors) and extrinsic regulators (morphogens and growth factors) that affect the generation, maintenance and expansion of HSCs in the embryo will be discussed. MAJOR CONCLUSIONS The generation of HSCs is a stepwise process involving many developmental signaling pathways, morphogens and cytokines. Pivotal hematopoietic transcription factors are required for their generation. Interestingly, whereas these factors are necessary for HSC generation, their expression in adult bone marrow HSCs is oftentimes not required. Thus, the biochemistry and molecular regulation of HSC development in the embryo are overlapping, but differ significantly from the regulation of HSCs in the adult. GENERAL SIGNIFICANCE HSC numbers for clinical use are limiting, and despite much research into the molecular basis of HSC regulation in the adult bone marrow, no panel of growth factors, interleukins and/or morphogens has been found to sufficiently increase the number of these important stem cells. An understanding of the biochemistry of HSC generation in the developing embryo provides important new knowledge on how these complex stem cells are made, sustained and expanded in the embryo to give rise to the complete adult hematopoietic system, thus stimulating novel strategies for producing increased numbers of clinically useful HSCs. This article is part of a Special Issue entitled Biochemistry of Stem Cells.
Collapse
Affiliation(s)
- P Kaimakis
- Erasmus Medical Center, Erasmus MC Stem Cell Institute, Dept. of Cell Biology, PO Box 2040, 3000 CA Rotterdam, Netherlands
| | | | | |
Collapse
|
76
|
Eom DS, Amarnath S, Fogel JL, Agarwala S. Bone morphogenetic proteins regulate hinge point formation during neural tube closure by dynamic modulation of apicobasal polarity. ACTA ACUST UNITED AC 2012; 94:804-16. [PMID: 22865775 DOI: 10.1002/bdra.23052] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 05/23/2012] [Accepted: 06/07/2012] [Indexed: 01/23/2023]
Abstract
BACKGROUND A critical event in neural tube closure is the formation of median hinge points (MHPs) and dorsolateral hinge points (DLHPs). Together, they buckle the ventral midline and elevate and juxtapose the neural folds for proper neural tube closure. Dynamic cell behaviors occur at hinge points (HPs), but their molecular regulation is largely unexplored. Bone morphogenetic proteins (BMPs) have been implicated in a variety of neural tube closure defects, although the underlying mechanisms are poorly understood. METHODS In this study, we used in vivo electroporations, high-resolution microscopy, and biochemical analyses to explore the role of BMP signaling in chick midbrain neural tube closure. RESULTS We identified a cell-cycle-dependent BMP gradient in the midbrain neural plate, which results in low-level BMP activity at the MHP. We show that although BMP signaling does not have a role in midbrain cell-fate specification, its attenuation is necessary and sufficient for MHP formation and midbrain closure. BMP blockade induces MHP formation by regulating apical constriction and basal nuclear migration. Furthermore, BMP signaling is critically important for maintaining epithelial organization by biochemically interacting with apicobasal polarity proteins (e.g., PAR3). As a result, prolonged BMP blockade disrupts apical junctions, desegregating the apical (PAR3(+), ZO1(+)) and basolateral (LGL(+)) compartments. Direct apical LGL-GFP misexpression in turn is sufficient to induce ectopic HPs. CONCLUSIONS BMPs have a critical role in maintaining epithelial organization, a role that is conserved across species and tissue types. Its cell-cycle-dependent modulation in the neural plate dynamically regulates apicobasal polarity and helps to bend, shape, and close the neural tube.
Collapse
Affiliation(s)
- Dae Seok Eom
- Institute for Cell and Molecular Biology, University of Texas, Austin, TX 78712, USA
| | | | | | | |
Collapse
|
77
|
Todorović-Raković N, Vujasinović T, Abu Rabi Z. Selection of clinically useful angiogenesis-related biomarkers: an update. Int J Biol Markers 2012; 27:e65-e81. [PMID: 22307386 DOI: 10.5301/jbm.2012.8989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2011] [Indexed: 11/20/2022]
Abstract
Angiogenesis is a complex phenomenon that involves interaction between growth factors/cytokines and their receptors, and proteolytic enzymes and their inhibitors, which, in addition to and in accordance with their main roles, act together during this multistep process. Cancer angiogenesis is specific, because the same factors that enable angiogenesis are involved in the process of carcinogenesis. The aim of this review was to analyze the current knowledge regarding the significance of selected biomarkers in cancer angiogenesis, with emphasis on their prognostic value in the circulation.
Collapse
Affiliation(s)
- Nataša Todorović-Raković
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade - Serbia.
| | | | | |
Collapse
|
78
|
Tmem100, an ALK1 receptor signaling-dependent gene essential for arterial endothelium differentiation and vascular morphogenesis. Proc Natl Acad Sci U S A 2012; 109:12064-9. [PMID: 22783020 DOI: 10.1073/pnas.1207210109] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Members of the transforming growth factor-β superfamily play essential roles in various aspects of embryonic development and physiological organ function. Among them, bone morphogenetic protein (BMP) 9 and BMP10 regulate embryonic vascular development by activating their endothelial receptor ALK1 (activin receptor-like kinase 1, also called Acvrl1). ALK1-mediated intracellular signaling is implicated in the etiologies of human diseases, but their downstream functional proteins are largely unknown. In this study, we identified Tmem100, a gene encoding a previously uncharacterized intracellular transmembrane protein, to be an embryonic endothelium-enriched gene activated by BMP9 and BMP10 through the ALK1 receptor. Tmem100 null mice showed embryonic lethality due to impaired differentiation of arterial endothelium and defects of vascular morphogenesis, which phenocopied most of the vascular abnormalities observed with the Acvrl1/Alk1 deficiency. The activity of Notch- and Akt-mediated signaling, which is essential for vascular development, was down-regulated in Tmem100 null mice. Cre-mediated deletion of Tmem100 in endothelial cells was sufficient to recapitulate the null phenotypes. These data indicated that TMEM100 may play indispensable roles downstream of BMP9/BMP10-ALK1 signaling during endothelial differentiation and vascular morphogenesis.
Collapse
|
79
|
Periostin as a biomarker of the amniotic membrane. Stem Cells Int 2012; 2012:987185. [PMID: 22966238 PMCID: PMC3395182 DOI: 10.1155/2012/987185] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 04/26/2012] [Accepted: 05/09/2012] [Indexed: 01/25/2023] Open
Abstract
Tracing the precise developmental origin of amnion and amnion-derived stem cells is still challenging and depends chiefly on analyzing powerful genetic model amniotes like mouse. Profound understanding of the fundamental differences in amnion development in both the disc-shaped primate and human embryo and the cup-shaped mouse embryo is pivotal in particular when sampling amniotic membrane from nonprimate species for isolating candidate amniotic stem cells. The availability of molecular marker genes that are specifically expressed in the amniotic membrane and not in other extraembryonic membranes would be instrumental to validate unequivocally the starting material under investigation. So far such amniotic markers have not been reported. We postulated that bone morphogenetic protein (BMP) target genes are putative amniotic membrane markers mainly because deficiency in one of several components of the BMP signaling cascade in mice has been documented to result in defective development of the early amnion. Comparative gene expression analysis of acknowledged target genes for BMP in different extraembryonic tissues, combined with in situ hybridization, identified Periostin (Postn) mRNA enrichment in amnion throughout gestation. In addition, we identify and propose a combination of markers as transcriptional signature for the different extraembryonic tissues in mouse.
Collapse
|
80
|
Kruithof BPT, Duim SN, Moerkamp AT, Goumans MJ. TGFβ and BMP signaling in cardiac cushion formation: lessons from mice and chicken. Differentiation 2012; 84:89-102. [PMID: 22656450 DOI: 10.1016/j.diff.2012.04.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Revised: 03/28/2012] [Accepted: 04/04/2012] [Indexed: 02/01/2023]
Abstract
Cardiac cushion formation is crucial for both valvular and septal development. Disruption in this process can lead to valvular and septal malformations, which constitute the largest part of congenital heart defects. One of the signaling pathways that is important for cushion formation is the TGFβ superfamily. The involvement of TGFβ and BMP signaling pathways in cardiac cushion formation has been intensively studied using chicken in vitro explant assays and in genetically modified mice. In this review, we will summarize and discuss the role of TGFβ and BMP signaling components in cardiac cushion formation.
Collapse
Affiliation(s)
- Boudewijn P T Kruithof
- Department of Molecular Cell Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| | | | | | | |
Collapse
|
81
|
Hazen VM, Andrews MG, Umans L, Crenshaw EB, Zwijsen A, Butler SJ. BMP receptor-activated Smads confer diverse functions during the development of the dorsal spinal cord. Dev Biol 2012; 367:216-27. [PMID: 22609550 DOI: 10.1016/j.ydbio.2012.05.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 04/21/2012] [Accepted: 05/09/2012] [Indexed: 01/19/2023]
Abstract
Bone Morphogenetic Proteins (BMPs) have multiple activities in the developing spinal cord: they specify the identity of the dorsal-most neuronal populations and then direct the trajectories of dorsal interneuron (dI) 1 commissural axons. How are these activities decoded by dorsal neurons to result in different cellular outcomes? Our previous studies have shown that the diverse functions of the BMPs are mediated by the canonical family of BMP receptors and then regulated by specific inhibitory (I) Smads, which block the activity of a complex of Smad second messengers. However, the extent to which this complex translates the different activities of the BMPs in the spinal cord has remained unresolved. Here, we demonstrate that the receptor-activated (R) Smads, Smad1 and Smad5 play distinct roles mediating the abilities of the BMPs to direct cell fate specification and axon outgrowth. Smad1 and Smad5 occupy spatially distinct compartments within the spinal cord, with Smad5 primarily associated with neural progenitors and Smad1 with differentiated neurons. Consistent with this expression profile, loss of function experiments in mouse embryos reveal that Smad5 is required for the acquisition of dorsal spinal neuron identities whereas Smad1 is critical for the regulation of dI1 axon outgrowth. Thus the R-Smads, like the I-Smads, have discrete roles mediating BMP-dependent cellular processes during spinal interneuron development.
Collapse
Affiliation(s)
- V M Hazen
- Department of Biological Sciences, Neuroscience Graduate Program, University of Southern California, Los Angeles, CA90089, USA
| | | | | | | | | | | |
Collapse
|
82
|
BMP signaling in vascular diseases. FEBS Lett 2012; 586:1993-2002. [DOI: 10.1016/j.febslet.2012.04.030] [Citation(s) in RCA: 223] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 04/05/2012] [Accepted: 04/17/2012] [Indexed: 12/24/2022]
|
83
|
Fine-tune of intrinsic ERK activity by extrinsic BMP signaling in mouse embryonic stem cells. Protein Cell 2012; 3:401-4. [PMID: 22528752 DOI: 10.1007/s13238-012-2925-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Accepted: 03/26/2012] [Indexed: 10/28/2022] Open
Abstract
Embryonic stem (ES) cells hold great promise in regenerative medicine and it is an urgent task to understand the underlying molecular mechanisms that control ES cell fate choice between self-renewal and differentiation. In mouse ES cells, extrinsic leukemia inhibitory factor (LIF) and bone morphogenetic protein (BMP) signaling pathways play pivotal roles in maintaining the self-renewal status under serum and feeder free culture conditions. Intrinsic extracellular-signal regulated kinase (ERK) activity is also important in determining mouse ES cell fate-low ERK activity keeps mouse ES cell self-renewal while high ERK activity drives differentiation. We recently found that while LIF signaling augments ERK activity, BMP signaling inhibits ERK activity in mouse ES cells via direct upregulation of an ERK phosphatase-dual-specificity phosphatase 9. The cooperative effects of LIF and BMP signaling keep appropriate ERK activity and maintain mouse ES cell self-renewal (Li et al., 2012). These findings shed light on how extrinsic signals converge to intrinsic signaling molecules to regulate cell fate determination. This perspective summarizes our recent new findings and discusses the current unsolved questions and future directions.
Collapse
|
84
|
Rhee JM, Iannaccone PM. Mapping mouse hemangioblast maturation from headfold stages. Dev Biol 2012; 365:1-13. [PMID: 22426104 DOI: 10.1016/j.ydbio.2012.02.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 02/14/2012] [Accepted: 02/15/2012] [Indexed: 11/18/2022]
Abstract
The mouse posterior primitive streak at neural plate/headfold stages (NP/HF, ~7.5 dpc-8 dpc) represents an optimal window from which hemangioblasts can be isolated. We performed immunohistochemistry on this domain using established monoclonal antibodies for proteins that affect blood and endothelial fates. We demonstrate that HoxB4 and GATA1 are the first set of markers that segregate independently to endothelial or blood populations during NP/HF stages of mouse embryonic development. In a subset of cells, both proteins are co-expressed and immunoreactivities appear mutually excluded within nuclear spaces. We searched for this particular state at later sites of hematopoietic stem cell emergence, viz., the aorta-gonad-mesonephros (AGM) and the fetal liver at 10.5-11.5 dpc, and found that only a rare number of cells displayed this character. Based on this spatial-temporal argument, we propose that the earliest blood progenitors emerge either directly from the epiblast or through segregation within the allantoic core domain (ACD) through reduction of cell adhesion and pSmad1/5 nuclear signaling, followed by a stochastic decision toward a blood or endothelial fate that involves GATA1 and HoxB4, respectively. A third form in which binding distributions are balanced may represent a common condition shared by hemangioblasts and HSCs. We developed a heuristic model of hemangioblast maturation, in part, to be explicit about our assumptions.
Collapse
Affiliation(s)
- Jerry M Rhee
- Children's Memorial Research Center, Department of Pediatrics, Developmental Biology Program, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | | |
Collapse
|
85
|
Moya IM, Umans L, Maas E, Pereira PNG, Beets K, Francis A, Sents W, Robertson EJ, Mummery CL, Huylebroeck D, Zwijsen A. Stalk cell phenotype depends on integration of Notch and Smad1/5 signaling cascades. Dev Cell 2012; 22:501-14. [PMID: 22364862 DOI: 10.1016/j.devcel.2012.01.007] [Citation(s) in RCA: 183] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2010] [Revised: 11/02/2011] [Accepted: 01/16/2012] [Indexed: 12/11/2022]
Abstract
Gradients of vascular endothelial growth factor (VEGF) induce single endothelial cells to become leading tip cells of emerging angiogenic sprouts. Tip cells then suppress tip-cell features in adjacent stalk cells via Dll4/Notch-mediated lateral inhibition. We report here that Smad1/Smad5-mediated BMP signaling synergizes with Notch signaling during selection of tip and stalk cells. Endothelium-specific inactivation of Smad1/Smad5 in mouse embryos results in impaired Dll4/Notch signaling and increased numbers of tip-cell-like cells at the expense of stalk cells. Smad1/5 downregulation in cultured endothelial cells reduced the expression of several target genes of Notch and of other stalk-cell-enriched transcripts (Hes1, Hey1, Jagged1, VEGFR1, and Id1-3). Moreover, Id proteins act as competence factors for stalk cells and form complexes with Hes1, which augment Hes1 levels in the endothelium. Our findings provide in vivo evidence for a regulatory loop between BMP/TGFβ-Smad1/5 and Notch signaling that orchestrates tip- versus stalk-cell selection and vessel plasticity.
Collapse
Affiliation(s)
- Iván M Moya
- VIB11 Center for the Biology of Disease, Laboratory of Developmental Signaling, VIB and Center for Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Wong YL, Behringer RR, Kwan KM. Smad1/Smad5 signaling in limb ectoderm functions redundantly and is required for interdigital programmed cell death. Dev Biol 2012; 363:247-57. [PMID: 22240098 DOI: 10.1016/j.ydbio.2011.12.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 12/17/2011] [Accepted: 12/22/2011] [Indexed: 11/30/2022]
Abstract
Bone morphogenetic proteins (BMPs) are secreted signals that regulate apical ectodermal ridge (AER) functions and interdigital programmed cell death (PCD) of developing limb. However the identities of the intracellular mediators of these signals are unknown. To investigate the role of Smad proteins in BMP-regulated AER functions in limb development, we inactivated Smad1 and Smad5 selectively in AER and ventral ectoderm of developing limb, using Smad1 or/and Smad5 floxed alleles and an En1(Cre/+) knock-in allele. Single inactivation of either Smad1 or Smad5 did not result in limb abnormalities. However, the Smad1/Smad5 double mutants exhibited syndactyly due to a reduction in interdigital PCD and an increase in interdigital cell proliferation. Cell tracing experiments in the Smad1/Smad5 double mutants showed that ventral ectoderm became thicker and the descendents of ventral En1(Cre/+) expressing ectodermal cells were located at dorsal interdigital regions. At the molecular level, Fgf8 expression was prolonged in the interdigital ectoderm of embryonic day (E) 13 Smad1/Smad5 double mutants, suggesting that the ectopic Fgf8 expression may serve as a survival signal for interdigital epithelial and mesenchymal cells. Our result suggests that Smad1 and Smad5 are required and function redundantly as intracellular mediators for BMP signaling in the AER and ventral ectoderm. Smad1/Smad5 signaling in the AER and ventral ectoderm regulates interdigital tissue regression of developing limb. Our mutants with defects in interdigital PCD could also serve as a valuable model for investigation of PCD regulation machinery.
Collapse
Affiliation(s)
- Yuk Lau Wong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, PR China
| | | | | |
Collapse
|
87
|
Li X, Tripurani SK, James R, Pangas SA. Minimal Fertility Defects in Mice Deficient in Oocyte-Expressed Smad41. Biol Reprod 2012; 86:1-6. [DOI: 10.1095/biolreprod.111.094375] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
88
|
Le Dréau G, Garcia-Campmany L, Rabadán MA, Ferronha T, Tozer S, Briscoe J, Martí E. Canonical BMP7 activity is required for the generation of discrete neuronal populations in the dorsal spinal cord. Development 2011; 139:259-68. [PMID: 22159578 DOI: 10.1242/dev.074948] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BMP activity is essential for many steps of neural development, including the initial role in neural induction and the control of progenitor identities along the dorsal-ventral axis of the neural tube. Taking advantage of chick in ovo electroporation, we show a novel role for BMP7 at the time of neurogenesis initiation in the spinal cord. Using in vivo loss-of-function experiments, we show that BMP7 activity is required for the generation of three discrete subpopulations of dorsal interneurons: dI1-dI3-dI5. Analysis of the BMP7 mouse mutant shows the conservation of this activity in mammals. Furthermore, this BMP7 activity appears to be mediated by the canonical Smad pathway, as we demonstrate that Smad1 and Smad5 activities are similarly required for the generation of dI1-dI3-dI5. Moreover, we show that this role is independent of the patterned expression of progenitor proteins in the dorsal spinal cord, but depends on the BMP/Smad regulation of specific proneural proteins, thus narrowing this BMP7 activity to the time of neurogenesis. Together, these data establish a novel role for BMP7 in primary neurogenesis, the process by which a neural progenitor exits the cell cycle and enters the terminal differentiation pathway.
Collapse
Affiliation(s)
- Gwenvael Le Dréau
- Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, c/Baldiri i Reixac 20, Barcelona 08028, Spain
| | | | | | | | | | | | | |
Collapse
|
89
|
Conidi A, Cazzola S, Beets K, Coddens K, Collart C, Cornelis F, Cox L, Joke D, Dobreva MP, Dries R, Esguerra C, Francis A, Ibrahimi A, Kroes R, Lesage F, Maas E, Moya I, Pereira PNG, Stappers E, Stryjewska A, van den Berghe V, Vermeire L, Verstappen G, Seuntjens E, Umans L, Zwijsen A, Huylebroeck D. Few Smad proteins and many Smad-interacting proteins yield multiple functions and action modes in TGFβ/BMP signaling in vivo. Cytokine Growth Factor Rev 2011; 22:287-300. [PMID: 22119658 DOI: 10.1016/j.cytogfr.2011.11.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Signaling by the many ligands of the TGFβ family strongly converges towards only five receptor-activated, intracellular Smad proteins, which fall into two classes i.e. Smad2/3 and Smad1/5/8, respectively. These Smads bind to a surprisingly high number of Smad-interacting proteins (SIPs), many of which are transcription factors (TFs) that co-operate in Smad-controlled target gene transcription in a cell type and context specific manner. A combination of functional analyses in vivo as well as in cell cultures and biochemical studies has revealed the enormous versatility of the Smad proteins. Smads and their SIPs regulate diverse molecular and cellular processes and are also directly relevant to development and disease. In this survey, we selected appropriate examples on the BMP-Smads, with emphasis on Smad1 and Smad5, and on a number of SIPs, i.e. the CPSF subunit Smicl, Ttrap (Tdp2) and Sip1 (Zeb2, Zfhx1b) from our own research carried out in three different vertebrate models.
Collapse
Affiliation(s)
- Andrea Conidi
- Laboratory of Molecular Biology (Celgen) of Center for Human Genetics, University of Leuven, Campus Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Madabhushi M, Lacy E. Anterior visceral endoderm directs ventral morphogenesis and placement of head and heart via BMP2 expression. Dev Cell 2011; 21:907-19. [PMID: 22075149 PMCID: PMC3386144 DOI: 10.1016/j.devcel.2011.08.027] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 05/30/2011] [Accepted: 08/29/2011] [Indexed: 01/07/2023]
Abstract
In amniotes, ventral folding morphogenesis achieves gut internalization, linear heart tube formation, ventral body wall closure, and encasement of the fetus in extraembryonic membranes. Impairment of ventral morphogenesis results in human birth defects involving body wall, gut, and heart malformations and in mouse misplacement of head and heart. Absence of knowledge about genetic pathways and cell populations directing ventral folding in mammals has precluded systematic study of cellular mechanisms driving this vital morphogenetic process. We report tissue-specific mouse mutant analyses identifying the bone morphogenetic protein (BMP) pathway as a key regulator of ventral morphogenesis. BMP2 expressed in anterior visceral endoderm (AVE) signals to epiblast derivatives during gastrulation to orchestrate initial stages of ventral morphogenesis, including foregut development and positioning of head and heart. These findings identify unanticipated functions for the AVE in organizing the gastrulating embryo and indicate that visceral endoderm-expressed BMP2 coordinates morphogenetic cell behaviors in multiple epiblast lineages.
Collapse
Affiliation(s)
- Mary Madabhushi
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY 10065, USA
| | | |
Collapse
|
91
|
Armulik A, Genové G, Betsholtz C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell 2011; 21:193-215. [PMID: 21839917 DOI: 10.1016/j.devcel.2011.07.001] [Citation(s) in RCA: 1955] [Impact Index Per Article: 139.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pericytes, the mural cells of blood microvessels, have recently come into focus as regulators of vascular morphogenesis and function during development, cardiovascular homeostasis, and disease. Pericytes are implicated in the development of diabetic retinopathy and tissue fibrosis, and they are potential stromal targets for cancer therapy. Some pericytes are probably mesenchymal stem or progenitor cells, which give rise to adipocytes, cartilage, bone, and muscle. However, there is still confusion about the identity, ontogeny, and progeny of pericytes. Here, we review the history of these investigations, indicate emerging concepts, and point out problems and promise in the field of pericyte biology.
Collapse
Affiliation(s)
- Annika Armulik
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | | | | |
Collapse
|
92
|
Drake KM, Zygmunt D, Mavrakis L, Harbor P, Wang L, Comhair SA, Erzurum SC, Aldred MA. Altered MicroRNA processing in heritable pulmonary arterial hypertension: an important role for Smad-8. Am J Respir Crit Care Med 2011; 184:1400-8. [PMID: 21920918 DOI: 10.1164/rccm.201106-1130oc] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
RATIONALE Heritable pulmonary arterial hypertension (HPAH) is primarily caused by mutations of the bone morphogenetic protein (BMP) type-II receptor (BMPR2). Recent identification of mutations in the downstream mediator Smad-8 (gene, SMAD9) was surprising, because loss of Smad-8 function in canonical BMP signaling is largely compensated by Smad-1 and -5. We therefore hypothesized that noncanonical pathways may play an important role in PAH. OBJECTIVES To determine whether HPAH mutations disrupt noncanonical Smad-mediated microRNA (miR) processing. METHODS Expression of miR-21, miR-27a, and miR-100 was studied in pulmonary artery endothelial (PAEC) and pulmonary artery smooth muscle cells (PASMC) from explant lungs of patients with PAH. MEASUREMENTS AND MAIN RESULTS SMAD9 mutation completely abrogated miR induction, whereas canonical signaling was only reduced by one-third. miR-21 levels actually decreased, suggesting that residual canonical signaling uses up or degrades existing miR-21. BMPR2 mutations also led to loss of miR induction in two of three cases. HPAH cells proliferated faster than other PAH or controls. miR-21 and miR-27a each showed antiproliferative effects in PAEC and PASMC, and PAEC growth rate after BMP treatment correlated strongly with miR-21 fold-change. Overexpression of SMAD9 corrected miR processing and reversed the hyperproliferative phenotype. CONCLUSIONS HPAH-associated mutations engender a primary defect in noncanonical miR processing, whereas canonical BMP signaling is partially maintained. Smad-8 is essential for this miR pathway and its loss was not complemented by Smad-1 and -5; this may represent the first nonredundant role for Smad-8. Induction of miR-21 and miR-27a may be a critical component of BMP-induced growth suppression, loss of which likely contributes to vascular cell proliferation in HPAH.
Collapse
Affiliation(s)
- Kylie M Drake
- Genomic Medicine Institute, Cleveland Clinic, OH 44195, USA
| | | | | | | | | | | | | | | |
Collapse
|
93
|
Pereira PNG, Dobreva MP, Graham L, Huylebroeck D, Lawson KA, Zwijsen AN. Amnion formation in the mouse embryo: the single amniochorionic fold model. BMC DEVELOPMENTAL BIOLOGY 2011; 11:48. [PMID: 21806820 PMCID: PMC3163621 DOI: 10.1186/1471-213x-11-48] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 08/01/2011] [Indexed: 12/16/2022]
Abstract
BACKGROUND Despite the detailed knowledge obtained over the last decade on the molecular regulation of gastrulation in amniotes, the process of amnion development has been poorly described and illustrated in mice, and conflicting descriptions exist. Understanding the morphogenesis and development not only of the early mouse embryo, but also of its extraembryonic tissues, is crucial for correctly interpreting fate-mapping data and mouse mutants with gastrulation defects. Moreover, the recent isolation from amnion of cells with stem cell features further argues for a better understanding of the process of amnion formation. Here, we revisit the highly dynamic process of amnion formation in the mouse. Amnion development starts early during gastrulation and is intimately related to the formation of the exocoelom and the expansion of the amniotic fold. The authoritative description involves the fusion of two amniotic folds, a big posterior and a smaller anterior fold. We challenged this 'two amniotic folds' model by performing detailed histomorphological analyses of dissected, staged embryos and 3D reconstructions using historical sections. RESULTS A posterior fold of extraembryonic ectoderm and associated epiblast is formed early during gastrulation by accumulation of extraembryonic mesoderm posterior to the primitive streak. Previously called the "posterior amniotic fold", we rename it the "amniochorionic fold" (ACF) because it forms both amnion and chorion. Exocoelom formation within the ACF seems not to involve apoptosis within the mesoderm. The ACF and exocoelom expand without disrupting the anterior junction of epiblast, extraembryonic ectoderm and visceral endoderm. No separate anterior fold is formed; its absence was confirmed in 3D reconstructions. Amnion and chorion closure is eccentric, close to the anterior margin of the egg cylinder: we name it the "anterior separation point". CONCLUSIONS Here, we reconcile previous descriptions of amnion formation and provide new nomenclature, as well as an animation, that clarify and emphasize the arrangement of the tissues that contribute to amnion development and the dynamics of the process. According to our data, the amnion and the chorion are formed by a single amniochorionic fold initiated posteriorly. Finally, we give an overview on mutant mouse models with impaired amnion development.
Collapse
Affiliation(s)
- Paulo N G Pereira
- Laboratory of Developmental Signaling of the Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium.
| | | | | | | | | | | |
Collapse
|
94
|
Rosenfeld JA, Drautz JM, Clericuzio CL, Cushing T, Raskin S, Martin J, Tervo RC, Pitarque JA, Nowak DM, Karolak JA, Lamb AN, Schultz RA, Ballif BC, Bejjani BA, Gajecka M, Shaffer LG. Deletions and duplications of developmental pathway genes in 5q31 contribute to abnormal phenotypes. Am J Med Genet A 2011; 155A:1906-16. [PMID: 21744490 DOI: 10.1002/ajmg.a.34100] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Accepted: 04/15/2011] [Indexed: 02/04/2023]
Abstract
Although copy number changes of 5q31 have been rarely reported, deletions have been associated with some common characteristics, such as short stature, failure to thrive, developmental delay (DD)/intellectual disability (ID), club feet, dislocated hips, and dysmorphic features. We report on three individuals with deletions and two individuals with duplications at 5q31, ranging from 3.6 Mb to 8.1 Mb and 830 kb to 3.4 Mb in size, respectively. All five copy number changes are apparently de novo and involve several genes that are important in developmental pathways, including PITX1, SMAD5, and WNT8A. The individuals with deletions have characteristic features including DD, short stature, club feet, cleft or high palate, dysmorphic features, and skeletal anomalies. Haploinsufficiency of PITX1, a transcription factor important for limb development, is likely the cause for the club feet, skeletal anomalies, and cleft/high palate, while additional genes, including SMAD5 and WNT8A, may also contribute to additional phenotypic features. Two patients with deletions also presented with corneal anomalies. To identify a causative gene for the corneal anomalies, we sequenced candidate genes in a family with apparent autosomal dominant keratoconus with suggestive linkage to 5q31, but no mutations in candidate genes were found. The duplications are smaller than the deletions, and the patients with duplications have nonspecific features. Although development is likely affected by increased dosage of the genes in the region, the developmental disruption appears less severe than that seen with deletion.
Collapse
|
95
|
Cook BD, Liu S, Evans T. Smad1 signaling restricts hematopoietic potential after promoting hemangioblast commitment. Blood 2011; 117:6489-97. [PMID: 21515822 PMCID: PMC3123019 DOI: 10.1182/blood-2010-10-312389] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Accepted: 04/13/2011] [Indexed: 11/20/2022] Open
Abstract
Bone morphogenetic protein (BMP) signaling regulates embryonic hematopoiesis via receptor-mediated activation of downstream SMAD proteins, including SMAD1. In previous work, we showed that Smad1 expression is sufficient to enhance commitment of mesoderm to hemangioblast fate. We also found indirect evidence to support a subsequent repressive function for Smad1 in hematopoiesis. To test this hypothesis directly, we developed a novel system allowing temporal control of Smad1 levels by conditional knockdown in embryonic stem cell derivatives. Depletion of Smad1 in embryoid body cultures before hemangioblast commitment limits hematopoietic potential because of a block in mesoderm development. Conversely, when Smad1 is depleted in FlK1(+) mesoderm, at a stage after hemangioblast commitment, the pool of hematopoietic progenitors is expanded. This involves enhanced expression levels for genes specific to hematopoiesis, including Gata1, Runx1 and Eklf, rather than factors required for earlier specification of the hemangioblast. The phenotype correlates with increased nuclear SMAD2 activity, indicating molecular cross-regulation between the BMP and TGF-β signaling pathways. Consistent with this mechanism, hematopoiesis was enhanced when Smad2 was directly expressed during this same developmental window. Therefore, this study reveals a temporally defined function for Smad1 in restricting the expansion of early hematopoietic progenitors.
Collapse
Affiliation(s)
- Brandoch D Cook
- Department of Surgery, Weill Cornell Medical College, 1300 York Ave, New York, NY 10065, USA
| | | | | |
Collapse
|
96
|
Liu W, Foley AC. Signaling pathways in early cardiac development. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2011; 3:191-205. [PMID: 20830688 DOI: 10.1002/wsbm.112] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cardiomyocyte differentiation is a complex multistep process requiring the proper temporal and spatial integration of multiple signaling pathways. Previous embryological and genetic studies have identified a number of signaling pathways that are critical to mediate the initial formation of the mesoderm and its allocation to the cardiomyocyte lineage. It has become clear that some of these signaling networks work autonomously, in differentiating myocardial cells whereas others work non-autonomously, in neighboring tissues, to regulate cardiac differentiation indirectly. Here, we provide an overview of three signaling networks that mediate cardiomyocyte specification and review recent insights into their specific roles in heart development. In addition, we demonstrate how systems level, 'omic approaches' and other high-throughput techniques such as small molecules screens are beginning to impact our understanding of cardiomyocyte specification and, to identify novel signaling pathways involved in this process. In particular, it now seems clear that at least one chemokine receptor CXCR4 is an important marker for cardiomyocyte progenitors and may play a functional role in their differentiation. Finally, we discuss some gaps in our current understanding of early lineage selection that could be addressed by various types of omic analysis.
Collapse
Affiliation(s)
- Wenrui Liu
- Greenberg Division of Cardiology, Department of Medicine, Weill Medical College of Cornell University, New York, NY, USA
| | | |
Collapse
|
97
|
Tian M, Neil JR, Schiemann WP. Transforming growth factor-β and the hallmarks of cancer. Cell Signal 2011; 23:951-62. [PMID: 20940046 PMCID: PMC3076078 DOI: 10.1016/j.cellsig.2010.10.015] [Citation(s) in RCA: 208] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 10/01/2010] [Indexed: 02/07/2023]
Abstract
Tumorigenesis is in many respects a process of dysregulated cellular evolution that drives malignant cells to acquire six phenotypic hallmarks of cancer, including their ability to proliferate and replicate autonomously, to resist cytostatic and apoptotic signals, and to induce tissue invasion, metastasis, and angiogenesis. Transforming growth factor-β (TGF-β) is a potent pleiotropic cytokine that functions as a formidable barrier to the development of cancer hallmarks in normal cells and tissues. Paradoxically, tumorigenesis counteracts the tumor suppressing activities of TGF-β, thus enabling TGF-β to stimulate cancer invasion and metastasis. Fundamental gaps exist in our knowledge of how malignant cells overcome the cytostatic actions of TGF-β, and of how TGF-β stimulates the acquisition of cancer hallmarks by developing and progressing human cancers. Here we review the molecular and cellular mechanisms that underlie the ability of TGF-β to mediate tumor suppression in normal cells, and conversely, to facilitate cancer progression and disease dissemination in malignant cells.
Collapse
Affiliation(s)
- Maozhen Tian
- Division of General Medical Sciences–Oncology, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - Jason R. Neil
- Department of Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - William P. Schiemann
- Division of General Medical Sciences–Oncology, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
98
|
Daane JM, Enders AC, Downs KM. Mesothelium of the murine allantois exhibits distinct regional properties. J Morphol 2011; 272:536-56. [PMID: 21284019 PMCID: PMC3078636 DOI: 10.1002/jmor.10928] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 10/18/2010] [Accepted: 10/25/2010] [Indexed: 11/06/2022]
Abstract
The rodent allantois is thought to be unique amongst mammals in not having an endodermal component. Here, we have investigated the mesothelium, or outer surface, of murine umbilical precursor tissue, the allantois (∼7.25-8.5 days postcoitum, dpc) to discover whether it exhibits the properties of an epithelium. A combination of morphology, challenge with biotinylated dextran amines (BDAs), and immunohistochemistry revealed that the mesothelium of the mouse allantois exhibits distinct regional properties. By headfold stages (∼7.75-8.0 dpc), distal mesothelium was generally squamous in shape, and highly permeable to BDA challenge, whereas ventral proximal mesothelium, referred to as "ventral cuboidal mesothelium" (VCM) for the characteristic cuboidal shape of its cells, was relatively impermeable. Although "dorsal cuboidal mesothelium" (DCM) resembled the VCM in cell shape, its permeability to BDA was intermediate between the other two regions. Results of immunostaining for Zonula Occludens-1 (ZO-1) and Epithelial-cadherin (E-cadherin), together with transmission electron microscopy (TEM), suggested that impermeability in the VCM may be due to greater cellular contact area between cells and close packing rather than to maturity of tight junctions, the latter of which, by comparison with the visceral yolk sac, appeared to be rare or absent from the allantoic surface. Both VCM and DCM exhibited an ultrastructure more favorable for protein synthesis than did the distal squamous mesothelium; however, at most stages, VCM exhibited robust afadin (AF-6), whereas the DCM uniquely contained alpha-4-integrin. These observations demonstrate that the allantoic mesothelium is not a conventional epithelium but possesses regional ultrastructural, functional and molecular differences that may play important roles in the correct deployment of the umbilical cord and its associated vascular, hematopoietic, and other cell types.
Collapse
Affiliation(s)
- Jacob M. Daane
- Department of Anatomy, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin 53706
| | - Allen C. Enders
- Department of Cell Biology and Human Anatomy, University of California-Davis, Davis, California 95616
| | - Karen M. Downs
- Department of Anatomy, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin 53706
| |
Collapse
|
99
|
Xie WB, Li Z, Miano JM, Long X, Chen SY. Smad3-mediated myocardin silencing: a novel mechanism governing the initiation of smooth muscle differentiation. J Biol Chem 2011; 286:15050-7. [PMID: 21402709 PMCID: PMC3083168 DOI: 10.1074/jbc.m110.202747] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 03/11/2011] [Indexed: 12/30/2022] Open
Abstract
Both TGF-β and myocardin (MYOCD) are important for smooth muscle cell (SMC) differentiation, but their precise role in regulating the initiation of SMC development is less clear. In TGF-β-induced SMC differentiation of pluripotent C3H10T1/2 progenitors, we found that TGF-β did not significantly induce Myocd mRNA expression until 18 h of stimulation. On the other hand, early SMC markers such as SM α-actin, SM22α, and SM calponin were detectable beginning 2 or 4 h after TGF-β treatment. These results suggest that Myocd expression is blocked during the initiation of TGF-β-induced SMC differentiation. Consistent with its endogenous expression, Myocd promoter activity was not elevated until 18 h following TGF-β stimulation. Surprisingly, Smad signaling was inhibitory to Myocd expression because blockade of Smad signaling enhanced Myocd promoter activity. Overexpression of Smad3, but not Smad2, inhibited Myocd promoter activity. Conversely, shRNA knockdown of Smad3 allowed TGF-β to activate the Myocd promoter in the initial phase of induction. Myocd was activated by PI3 kinase signaling and its downstream target Nkx2.5. Interestingly, Smad3 did not affect PI3 kinase activity. However, Smad3 physically interacted with Nkx2.5. This interaction blocked Nkx2.5 binding to the Myocd promoter in the early stage of TGF-β induction, leading to inhibition of Myocd mRNA expression. Moreover, Smad3 inhibited Nkx2.5-activated Myocd promoter activity in a dose-dependent manner. Taken together, our results reveal a novel mechanism for Smad3-mediated inhibition of Myocd in the initiation phase of SMC differentiation.
Collapse
Affiliation(s)
- Wei-Bing Xie
- From the Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia 30602 and
| | - Zuguo Li
- From the Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia 30602 and
| | - Joseph M. Miano
- the Aab Cardiovascular Research Institute, University of Rochester, Rochester, New York 14642
| | - Xiaochun Long
- the Aab Cardiovascular Research Institute, University of Rochester, Rochester, New York 14642
| | - Shi-You Chen
- From the Department of Physiology and Pharmacology, University of Georgia, Athens, Georgia 30602 and
| |
Collapse
|
100
|
Bone morphogenetic protein 2 improves patellar tendon healing by promoting migration and proliferation of tenocytes. ACTA ACUST UNITED AC 2011. [DOI: 10.1007/s11434-011-4438-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|