1
|
Cao S, Wu Y, Albert Reece E, Xu C, Shen WB, Kaushal S, Yang P. Functional cargos of exosomes derived from Flk-1 + vascular progenitors enable neurulation and ameliorate embryonic anomalies in diabetic pregnancy. Commun Biol 2022; 5:648. [PMID: 35778435 PMCID: PMC9249756 DOI: 10.1038/s42003-022-03614-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 06/21/2022] [Indexed: 11/27/2022] Open
Abstract
Various types of progenitors initiate individual organ formation and their crosstalk orchestrates morphogenesis for the entire embryo. Here we show that progenitor exosomal communication across embryonic organs occurs in normal development and is altered in embryos of diabetic pregnancy. Endoderm fibroblast growth factor 2 (FGF2) stimulates mesoderm Flk-1+ vascular progenitors to produce exosomes containing the anti-stress protein Survivin. These exosomes act on neural stem cells of the neuroepithelium to facilitate neurulation by inhibiting cellular stress and apoptosis. Maternal diabetes causes Flk-1+ progenitor dysfunction by suppressing FGF2 through DNA hypermethylation. Restoring endoderm FGF2 prevents diabetes-induced survivin reduction in Flk-1+ progenitor exosomes. Transgenic Survivin expression in Flk-1+ progenitors or in utero delivery of survivin-enriched exosomes restores cellular homeostasis and prevents diabetes-induced neural tube defects (NTDs), whereas inhibiting exosome production induces NTDs. Thus, functional inter-organ communication via Flk-1 exosomes is vital for neurulation and its disruption leads to embryonic anomalies.
Collapse
Affiliation(s)
- Songying Cao
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yanqing Wu
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Institute of Life Sciences, Wenzhou University, Zhejiang Province, 325035, Wenzhou, China
| | - E Albert Reece
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cheng Xu
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Wei-Bin Shen
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sunjay Kaushal
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
- Division of Cardiovascular-Thoracic Surgery, Ann & Robert H. Lurie Children's Hospital of Chicago, 225 E. Chicago Avenue, Chicago, IL, 60611, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.
- Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Amnion signals are essential for mesoderm formation in primates. Nat Commun 2021; 12:5126. [PMID: 34446705 PMCID: PMC8390679 DOI: 10.1038/s41467-021-25186-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 07/22/2021] [Indexed: 11/26/2022] Open
Abstract
Embryonic development is largely conserved among mammals. However, certain genes show divergent functions. By generating a transcriptional atlas containing >30,000 cells from post-implantation non-human primate embryos, we uncover that ISL1, a gene with a well-established role in cardiogenesis, controls a gene regulatory network in primate amnion. CRISPR/Cas9-targeting of ISL1 results in non-human primate embryos which do not yield viable offspring, demonstrating that ISL1 is critically required in primate embryogenesis. On a cellular level, mutant ISL1 embryos display a failure in mesoderm formation due to reduced BMP4 signaling from the amnion. Via loss of function and rescue studies in human embryonic stem cells we confirm a similar role of ISL1 in human in vitro derived amnion. This study highlights the importance of the amnion as a signaling center during primate mesoderm formation and demonstrates the potential of in vitro primate model systems to dissect the genetics of early human embryonic development. Human and murine embryonic development has disparities, highlighting the need for primate systems. Here, the authors construct a post-implantation transcriptional atlas from non-human primate embryos and show ISL1 controls a gene regulatory network in the amnion required for mesoderm formation.
Collapse
|
3
|
Abstract
The lateral plate mesoderm (LPM) forms the progenitor cells that constitute the heart and cardiovascular system, blood, kidneys, smooth muscle lineage and limb skeleton in the developing vertebrate embryo. Despite this central role in development and evolution, the LPM remains challenging to study and to delineate, owing to its lineage complexity and lack of a concise genetic definition. Here, we outline the processes that govern LPM specification, organization, its cell fates and the inferred evolutionary trajectories of LPM-derived tissues. Finally, we discuss the development of seemingly disparate organ systems that share a common LPM origin. Summary: The lateral plate mesoderm is the origin of several major cell types and organ systems in the vertebrate body plan. How this mesoderm territory emerges and partitions into its downstream fates provides clues about vertebrate development and evolution.
Collapse
Affiliation(s)
- Karin D Prummel
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA.,Department of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Susan Nieuwenhuize
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA.,Department of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | - Christian Mosimann
- University of Colorado School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA .,Department of Molecular Life Sciences, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| |
Collapse
|
4
|
Yu SB, Umair Z, Kumar S, Lee U, Lee SH, Kim JI, Kim S, Park JB, Lee JY, Kim J. xCyp26c Induced by Inhibition of BMP Signaling Is Involved in Anterior-Posterior Neural Patterning of Xenopus laevis. Mol Cells 2016; 39:352-7. [PMID: 26923193 PMCID: PMC4844943 DOI: 10.14348/molcells.2016.0006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 01/15/2016] [Accepted: 01/18/2016] [Indexed: 01/08/2023] Open
Abstract
Vertebrate neurogenesis requires inhibition of endogenous bone morphogenetic protein (BMP) signals in the ectoderm. Blocking of BMPs in animal cap explants causes the formation of anterior neural tissues as a default fate. To identify genes involved in the anterior neural specification, we analyzed gene expression profiles using a Xenopus Affymetrix Gene Chip after BMP-4 inhibition in animal cap explants. We found that the xCyp26c gene, encoding a retinoic acid (RA) degradation enzyme, was upregulated following inhibition of BMP signaling in early neuroectodermal cells. Whole-mount in situ hybridization analysis showed that xCyp26c expression started in the anterior region during the early neurula stage. Overexpression of xCyp26c weakly induced neural genes in animal cap explants. xCyp26c abolished the expression of all trans-/cis-RA-induced posterior genes, but not basic FGF-induced posterior genes. Depletion of xCyp26c by morpholino-oligonucleotides suppressed the normal formation of the axis and head, indicating that xCyp26c plays a critical role in the specification of anterior neural tissue in whole embryos. In animal cap explants, however, xCyp26c morpholinos did not alter anterior-to-posterior neural tissue formation. Together, these results suggest that xCyp26c plays a specific role in anterior-posterior (A-P) neural patterning of Xenopus embryos.
Collapse
Affiliation(s)
- Saet-Byeol Yu
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Kangwon 200-702,
Korea
| | - Zobia Umair
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Kangwon 200-702,
Korea
| | - Shiv Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Kangwon 200-702,
Korea
| | - Unjoo Lee
- Department of Electrical Engineering, Hallym University, Kangwon200-702,
Korea
| | - Seung-Hwan Lee
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Kangwon 200-702,
Korea
| | - Jong-Il Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 110-799,
Korea
| | - SungChan Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Kangwon 200-702,
Korea
| | - Jae-Bong Park
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Kangwon 200-702,
Korea
| | - Jae-Yong Lee
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Kangwon 200-702,
Korea
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Kangwon 200-702,
Korea
| |
Collapse
|
5
|
Chen C, Jin J, Lee GA, Silva E, Donoghue M. Cross-species functional analyses reveal shared and separate roles for Sox11 in frog primary neurogenesis and mouse cortical neuronal differentiation. Biol Open 2016; 5:409-17. [PMID: 26962049 PMCID: PMC4890661 DOI: 10.1242/bio.015404] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
A well-functioning brain requires production of the correct number and types of cells during development; cascades of transcription factors are essential for cellular coordination. Sox proteins are transcription factors that affect various processes in the development of the nervous system. Sox11, a member of the SoxC family, is expressed in differentiated neurons and supports neuronal differentiation in several systems. To understand how generalizable the actions of Sox11 are across phylogeny, its function in the development of the frog nervous system and the mouse cerebral cortex were compared. Expression of Sox11 is largely conserved between these species; in the developing frog, Sox11 is expressed in the neural plate, neural tube and throughout the segmented brain, while in the mouse cerebral cortex, Sox11 is expressed in differentiated zones, including the preplate, subplate, marginal zone and cortical plate. In both frog and mouse, data demonstrate that Sox11 supports a role in promoting neuronal differentiation, with Sox11-positive cells expressing pan-neural markers and becoming morphologically complex. However, frog and mouse Sox11 cannot substitute for one another; a functional difference likely reflected in sequence divergence. Thus, Sox11 appears to act similarly in subserving neuronal differentiation but is species-specific in frog neural development and mouse corticogenesis. Summary: Sox11 acts to designate neurons in both mouse and frog brains, but orthologs are not functionally redundant. These data show evolutionary conservation of Sox11 function with molecular divergence.
Collapse
Affiliation(s)
- Chao Chen
- Department of Biology, Georgetown University, 37th and O Street NW, Washington, DC 20057, USA
| | - Jing Jin
- Department of Biology, Georgetown University, 37th and O Street NW, Washington, DC 20057, USA
| | - Garrett A Lee
- Department of Biology, Georgetown University, 37th and O Street NW, Washington, DC 20057, USA
| | - Elena Silva
- Department of Biology, Georgetown University, 37th and O Street NW, Washington, DC 20057, USA
| | - Maria Donoghue
- Department of Biology, Georgetown University, 37th and O Street NW, Washington, DC 20057, USA
| |
Collapse
|
6
|
Pillai-Kastoori L, Wen W, Morris AC. Keeping an eye on SOXC proteins. Dev Dyn 2015; 244:367-376. [PMID: 25476579 PMCID: PMC4344926 DOI: 10.1002/dvdy.24235] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/10/2014] [Accepted: 11/28/2014] [Indexed: 12/17/2022] Open
Abstract
The formation of a mature, functional eye requires a complex series of cell proliferation, migration, induction among different germinal layers, and cell differentiation. These processes are regulated by extracellular cues such as the Wnt/BMP/Hh/Fgf signaling pathways, as well as cell intrinsic transcription factors that specify cell fate. In this review article, we provide an overview of stages of embryonic eye morphogenesis, extrinsic and intrinsic factors that are required for each stage, and pediatric ocular diseases that are associated with defective eye development. In addition, we focus on recent findings about the roles of the SOXC proteins in regulating vertebrate ocular development and implicating SOXC mutations in human ocular malformations.
Collapse
Affiliation(s)
| | - Wen Wen
- Department of Biology, University of Kentucky, Lexington, Kentucky, USA
| | - Ann C. Morris
- Department of Biology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
7
|
Ninomiya N, Michiue T, Asashima M, Kurisaki A. BMP signaling regulates the differentiation of mouse embryonic stem cells into lung epithelial cell lineages. In Vitro Cell Dev Biol Anim 2013; 49:230-7. [PMID: 23468359 DOI: 10.1007/s11626-013-9589-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 02/01/2013] [Indexed: 10/27/2022]
Abstract
Somatic stem/progenitor cells are known to be present in most adult tissues. However, those in the lung have limited abilities for tissue regeneration after serious damage as a result of chronic disease. Therefore, regenerative medicine using exogenous stem cells has been suggested for the treatment of progressive lung diseases such as chronic obstructive pulmonary disease and pulmonary fibrosis. Embryonic stem (ES) cells and induced pluripotent stem cells, with their potent differentiation abilities, are promising sources for the generation of various tissue cells. In this study, we investigated the effects of various differentiation-inducing growth factors on the differentiation of lung cells from ES cells in vitro. Several factors, including activin, nodal, and noggin, significantly promoted the induction of Nkx2.1-positive lung progenitor cells when cells were cultured as embryoid bodies. Bone morphogenetic protein (BMP) 4 signaling controls the lineage commitment of lung cells along the proximal-distal axis. BMP4 promotes the induction of distal cell lineages of alveolar bud, such as Clara cells and mucus-producing goblet cells. These results suggest that several developmentally essential factors, including nodal/activin and BMP signaling, are important in the control of the differentiation of lung epithelial cells from mouse ES cells in vitro.
Collapse
Affiliation(s)
- Naoto Ninomiya
- Department of Life Sciences (Biology), Graduate School of Arts and Sciences, The University of Tokyo, Meguro, Tokyo, Japan
| | | | | | | |
Collapse
|
8
|
Maina JN. Comparative molecular developmental aspects of the mammalian- and the avian lungs, and the insectan tracheal system by branching morphogenesis: recent advances and future directions. Front Zool 2012; 9:16. [PMID: 22871018 PMCID: PMC3502106 DOI: 10.1186/1742-9994-9-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 06/18/2012] [Indexed: 02/07/2023] Open
Abstract
Gas exchangers fundamentally form by branching morphogenesis (BM), a mechanistically profoundly complex process which derives from coherent expression and regulation of multiple genes that direct cell-to-cell interactions, differentiation, and movements by signaling of various molecular morphogenetic cues at specific times and particular places in the developing organ. Coordinated expression of growth-instructing factors determines sizes and sites where bifurcation occurs, by how much a part elongates before it divides, and the angle at which branching occurs. BM is essentially induced by dualities of factors where through feedback- or feed forward loops agonists/antagonists are activated or repressed. The intricate transactions between the development orchestrating molecular factors determine the ultimate phenotype. From the primeval time when the transformation of unicellular organisms to multicellular ones occurred by systematic accretion of cells, BM has been perpetually conserved. Canonical signalling, transcriptional pathways, and other instructive molecular factors are commonly employed within and across species, tissues, and stages of development. While much still remain to be elucidated and some of what has been reported corroborated and reconciled with rest of existing data, notable progress has in recent times been made in understanding the mechanism of BM. By identifying and characterizing the morphogenetic drivers, and markers and their regulatory dynamics, the elemental underpinnings of BM have been more precisely explained. Broadening these insights will allow more effective diagnostic and therapeutic interventions of developmental abnormalities and pathologies in pre- and postnatal lungs. Conservation of the molecular factors which are involved in the development of the lung (and other branched organs) is a classic example of nature's astuteness in economically utilizing finite resources. Once purposefully formed, well-tested and tried ways and means are adopted, preserved, and widely used to engineer the most optimal phenotypes. The material and time costs of developing utterly new instruments and routines with every drastic biological change (e.g. adaptation and speciation) are circumvented. This should assure the best possible structures and therefore functions, ensuring survival and evolutionary success.
Collapse
Affiliation(s)
- John N Maina
- Department of Zoology, University of Johannesburg, Auckland Park 2006, P,O, Box 524, Johannesburg, South Africa.
| |
Collapse
|
9
|
Lee SY, Lim SK, Cha SW, Yoon J, Lee SH, Lee HS, Park JB, Lee JY, Kim SC, Kim J. Inhibition of FGF signaling converts dorsal mesoderm to ventral mesoderm in early Xenopus embryos. Differentiation 2011; 82:99-107. [PMID: 21684060 DOI: 10.1016/j.diff.2011.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Revised: 05/20/2011] [Accepted: 05/24/2011] [Indexed: 11/16/2022]
Abstract
In early vertebrate development, mesoderm induction is a crucial event regulated by several factors including the activin, BMP and FGF signaling pathways. While the requirement of FGF in Nodal/activin-induced mesoderm formation has been reported, the fate of the tissue modulated by these signals is not fully understood. Here, we examined the fate of tissues when exogenous activin was added and FGF signaling was inhibited in animal cap explants of Xenopus embryos. Activin-induced dorsal mesoderm was converted to ventral mesoderm by inhibition of FGF signaling. We also found that inhibiting FGF signaling in the dorsal marginal zone, in vegetal-animal cap conjugates or in the presence of the activin signaling component Smad2, converted dorsal mesoderm to ventral mesoderm. The expression and promoter activities of a BMP responsive molecule, PV.1 and a Spemann organizer, noggin, were investigated while FGF signaling was inhibited. PV.1 expression increased, while noggin decreased. In addition, inhibiting BMP-4 signaling abolished ventral mesoderm formation induced by exogenous activin and FGF inhibition. Taken together, these results suggest that the formation of dorso-ventral mesoderm in early Xenopus embryos is regulated by a combination of FGF, activin and BMP signaling.
Collapse
Affiliation(s)
- Sung-Young Lee
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, 200-702, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Yoon J, Kim JH, Lee OJ, Yu SB, Kim JI, Kim SC, Park JB, Lee JY, Kim J. xCITED2 Induces Neural Genes in Animal Cap Explants of Xenopus Embryos. Exp Neurobiol 2011; 20:123-9. [PMID: 22110370 PMCID: PMC3214773 DOI: 10.5607/en.2011.20.3.123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Accepted: 06/17/2011] [Indexed: 11/19/2022] Open
Abstract
Neural tissue is arisen from presumptive ectoderm via inhibition of bone morphogenetic protein (BMP) signaling during Xenopus early development. Previous studies demonstrate that ectopic expression of dominant negative BMP4 receptor (DNBR) produces neural tissue in animal cap explants (AC) and also increases the expression level of various genes involved in neurogenesis. To investigate detail mechanism of neurogenesis in transcriptional level, we analyzed RNAs increased by DNBR using total RNA sequencing analysis and identified several candidate genes. Among them, xCITED2 (Xenopus CBP/p300-interacting transcription activator) was induced 4.6 fold by DNBR and preferentially expressed in neural tissues at tadpole stage. Ectopic expression of xCITED2 induced anterior neural genes without mesoderm induction and reduced BMP downstream genes, an eye specific marker and posterior neural marker. Taken together, these results suggest that xCITED2 may have a role in the differentiation of anterior neural tissue during Xenopus early development.
Collapse
Affiliation(s)
- Jaeho Yoon
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon 200-702, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Lee SY, Yoon J, Lee HS, Hwang YS, Cha SW, Jeong CH, Kim JI, Park JB, Lee JY, Kim S, Park MJ, Dong Z, Kim J. The function of heterodimeric AP-1 comprised of c-Jun and c-Fos in activin mediated Spemann organizer gene expression. PLoS One 2011; 6:e21796. [PMID: 21829441 PMCID: PMC3146467 DOI: 10.1371/journal.pone.0021796] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 06/13/2011] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Activator protein-1 (AP-1) is a mediator of BMP or FGF signaling during Xenopus embryogenesis. However, specific role of AP-1 in activin signaling has not been elucidated during vertebrate development. METHODOLOGY/PRINCIPAL FINDINGS We provide new evidence showing that overexpression of heterodimeric AP-1 comprised of c-jun and c-fos (AP-1(c-Jun/c-Fos)) induces the expression of BMP-antagonizing organizer genes (noggin, chordin and goosecoid) that were normally expressed by high dose of activin. AP-1(c-Jun/c-Fos) enhanced the promoter activities of organizer genes but reduced that of PV.1, a BMP4-response gene. A loss of function study clearly demonstrated that AP-1(c-Jun/c-Fos) is required for the activin-induced organizer and neural gene expression. Moreover, physical interaction of AP-1(c-Jun/c-Fos) and Smad3 cooperatively enhanced the transcriptional activity of goosecoid via direct binding on this promoter. Interestingly, Smad3 mutants at c-Jun binding site failed in regulation of organizer genes, indicating that these physical interactions are specifically necessary for the expression of organizer genes. CONCLUSIONS/SIGNIFICANCE AP-1(c-Jun/c-Fos) plays a specific role in organizer gene expression in downstream of activin signal during early Xenopus embryogenesis.
Collapse
Affiliation(s)
- Sung-Young Lee
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| | - Jaeho Yoon
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
| | - Hyun-Shik Lee
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
- School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Yoo-Seok Hwang
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Sang-Wook Cha
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Chul-Ho Jeong
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Jong-Il Kim
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
| | - Jae-Yong Lee
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
| | - SungChan Kim
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
| | - Mae Ja Park
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| | - Jaebong Kim
- Department of Biochemistry, College of Medicine, Hallym University, ChunCheon, Kangwon-Do, Republic of Korea
| |
Collapse
|
12
|
Kim Y, Joshi SD, Messner WC, LeDuc PR, Davidson LA. Detection of dynamic spatiotemporal response to periodic chemical stimulation in a Xenopus embryonic tissue. PLoS One 2011; 6:e14624. [PMID: 21305055 PMCID: PMC3031512 DOI: 10.1371/journal.pone.0014624] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Accepted: 12/31/2010] [Indexed: 11/19/2022] Open
Abstract
Embryonic development is guided by a complex and integrated set of stimuli that results in collective system-wide organization that is both time and space regulated. These regulatory interactions result in the emergence of highly functional units, which are correlated to frequency-modulated stimulation profiles. We have determined the dynamic response of vertebrate embryonic tissues to highly controlled, time-varying localized chemical stimulation using a microfluidic system with feedback control. Our approach has enabled localized spatiotemporal manipulation of the steroid hormone dexamethasone (DEX) in Animal Cap (AC) tissues isolated from gastrulating Xenopus embryos. Using this approach we investigated cell-scale responses to precisely controlled stimulation by tracking the redistribution of a GFP-tagged DEX-reporter constructed from the human glucocorticoid receptor (GR). We exposed defined regions of a single AC explant to different stimulation conditions--continuous stimulation, periodic stimulation, and no stimulation. We observed collective behavior of the GR transport into the nucleus was first-order. Furthermore, the dynamic response was well-modeled by a first-order differential equation with a single time derivative. The model predicted that responses to periodic stimulations closely matched the results of the frequency-based experiments. We find that stimulation with localized bursts versus continuous stimulation can result in highly distinct responses. This finding is critical as controlled space and time exposure to growth factors is a hallmark of complex processes in embryonic development. These complex responses to cellular signaling and transport machinery were similar to emergent behaviors in other complex systems, suggesting that even within a complex embryonic tissue, the overall system can converge toward a predictive first-order response.
Collapse
Affiliation(s)
- YongTae Kim
- Departments of Mechanical Engineering, Biomedical Engineering, and Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
| | - Sagar D. Joshi
- Departments of Bioengineering and Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - William C. Messner
- Departments of Mechanical Engineering, Biomedical Engineering, and Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (WCM); (PRL); (LAD)
| | - Philip R. LeDuc
- Departments of Mechanical Engineering, Biomedical Engineering, and Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (WCM); (PRL); (LAD)
| | - Lance A. Davidson
- Departments of Bioengineering and Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (WCM); (PRL); (LAD)
| |
Collapse
|
13
|
Huang HY, Hu LL, Song TJ, Li X, He Q, Sun X, Li YM, Lu HJ, Yang PY, Tang QQ. Involvement of cytoskeleton-associated proteins in the commitment of C3H10T1/2 pluripotent stem cells to adipocyte lineage induced by BMP2/4. Mol Cell Proteomics 2010; 10:M110.002691. [PMID: 20713452 DOI: 10.1074/mcp.m110.002691] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The developmental pathway that gives rise to mature adipocytes involves two distinct stages: commitment and terminal differentiation. Although the important proteins/factors contributing to terminal adipocyte differentiation have been well defined, the proteins/factors in the commitment of mesenchymal stem cells to the adipocyte lineage cells have not. In this study, we applied proteomics analysis profiling to characterize differences between uncommitted C3H10T1/2 pluripotent stem cells and those that have been committed to the adipocyte lineage by BMP4 or BMP2 with the goal to identify such proteins/factors and to understand the molecular mechanisms that govern the earliest stages of adipocyte lineage commitment. Eight proteins were found to be up-regulated by BMP2, and 27 proteins were up-regulated by BMP4, whereas five unique proteins were up-regulated at least 10-fold by both BMP2/4, including three cytoskeleton-associated proteins (i.e. lysyl oxidase (LOX), translationally controlled tumor protein 1 (TPT1), and αB-crystallin). Western blotting further confirmed the induction of the expression of these cytoskeleton-associated proteins in the committed C3H10T1/2 induced by BMP2/4. Importantly, knockdown of LOX expression totally prevented the commitment, whereas knockdown of TPT1 and αB-crystallin expression partially inhibited the commitment. Several published reports suggest that cell shape can influence the differentiation of partially committed precursors of adipocytes, osteoblasts, and chondrocytes. We observed a dramatic change of cell shape during the commitment process, and we showed that knockdown of these cytoskeleton-associated proteins prevented the cell shape change and restored F-actin organization into stress fibers and inhibited the commitment to the adipocyte lineage. Our studies indicate that these differentially expressed cytoskeleton-associate proteins might determine the fate of mesenchymal stem cells to commit to the adipocyte lineage through cell shape regulation.
Collapse
Affiliation(s)
- Hai-Yan Huang
- The Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Watanabe Y, Itoh S, Goto T, Ohnishi E, Inamitsu M, Itoh F, Satoh K, Wiercinska E, Yang W, Shi L, Tanaka A, Nakano N, Mommaas AM, Shibuya H, Ten Dijke P, Kato M. TMEPAI, a transmembrane TGF-beta-inducible protein, sequesters Smad proteins from active participation in TGF-beta signaling. Mol Cell 2010; 37:123-34. [PMID: 20129061 DOI: 10.1016/j.molcel.2009.10.028] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Revised: 06/18/2009] [Accepted: 10/30/2009] [Indexed: 01/30/2023]
Abstract
Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine of key importance for controlling embryogenesis and tissue homeostasis. How TGF-beta signals are attenuated and terminated is not well understood. Here, we show that TMEPAI, a direct target gene of TGF-beta signaling, antagonizes TGF-beta signaling by interfering with TGF-beta type I receptor (TbetaRI)-induced R-Smad phosphorylation. TMEPAI can directly interact with R-Smads via a Smad interaction motif. TMEPAI competes with Smad anchor for receptor activation for R-Smad binding, thereby sequestering R-Smads from TbetaRI kinase activation. In mammalian cells, ectopic expression of TMEPAI inhibited TGF-beta-dependent regulation of plasminogen activator inhibitor-1, JunB, cyclin-dependent kinase inhibitors, and c-myc expression, whereas specific knockdown of TMEPAI expression prolonged duration of TGF-beta-induced Smad2 and Smad3 phosphorylation and concomitantly potentiated cellular responsiveness to TGF-beta. Consistently, TMEPAI inhibits activin-mediated mesoderm formation in Xenopus embryos. Therefore, TMEPAI participates in a negative feedback loop to control the duration and intensity of TGF-beta/Smad signaling.
Collapse
Affiliation(s)
- Yukihide Watanabe
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Hashemi-Tabar M, Orazizadeh M, Ghanbari A, Dehbashi FN. Kinetics of gene expression during exposure of mouse stem cells to activin A. Pak J Biol Sci 2009; 12:324-31. [PMID: 19579965 DOI: 10.3923/pjbs.2009.324.331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This study aimed to evaluate the pattern of gene expression induced by activin A in mouse Embryonic Stem Cells (ESCs). Mouse ES cells cultured in undifferentiated state by leukemia inhibitory factor and feeder layer cells. Following removing these two anti differentiation factors for 5 days and forming Embryoid Bodies (EBs), the cells divided to 8 equal cells per groups. Differentiation procedure was performed in a two staged protocol; Formed EBs for 4 days (Stage one); expanded differentiated ESCs on gelatin coated dishes for one week (stage two). In the stage one, the media of groups 2-7 contained 10, 30 and 100 ng mL(-1) Activin A. The media in stage two was the same for all groups and contained only Fetal Bovine Serum (FBS). The expression of undifferentiated, ectoderm, mesoderm and endoderm markers were compared with relative RT-PCR method and statistically analyzed. The expression of an undifferentiating marker; Nanog was increased in the Activin A treated groups of stage one. The expression of OCT4 reduced in Activin A treated groups in stage two. In the stage one, the expression of Nodal increased by Activin A. expression of sonic hedgehog (Shh) was suppressed in Activin A treated groups of both stages. In stage two, there were significant decrease for the expression of mesoderm (Brachyury) and Nodal and visceral endoderm (GATA4) markers (p < 0.01). The expression of definitive endoderm markers (PDX1, TAT) showed significantly increased in Activin A treated groups (p < 0.01). Activin A induced differentiation in high concentration by imbalance in undifferentiating markers. Nodal has a dual role, undifferentiating effect and regulation of visceral endoderm towards definitive endoderm. Overexpression of Nanog, alteration in the expression of Nodal and Shh inhibition are three mechanisms for explanation of differentiation induced by activin A in ES cells. These mechanisms induces cascade of gene expression that commits ESCs towards definitive endodermal cells.
Collapse
Affiliation(s)
- Mahmoud Hashemi-Tabar
- Cell and Molecular Research Centre, School of Medicine, Jondishapour University of Medical Sciences, Ahwaz, Iran
| | | | | | | |
Collapse
|
16
|
Keren A, Keren-Politansky A, Bengal E. A p38 MAPK-CREB pathway functions to pattern mesoderm in Xenopus. Dev Biol 2008; 322:86-94. [PMID: 18675264 DOI: 10.1016/j.ydbio.2008.07.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2008] [Revised: 06/29/2008] [Accepted: 07/07/2008] [Indexed: 12/30/2022]
Abstract
Dorsal-ventral patterning is specified by signaling centers secreting antagonizing morphogens that form a signaling gradient. Yet, how morphogen gradient is translated intracellularly into fate decisions remains largely unknown. Here, we report that p38 MAPK and CREB function along the dorsal-ventral axis in mesoderm patterning. We find that the phosphorylated form of CREB (S133) is distributed in a gradient along the dorsal-ventral mesoderm axis and that the p38 MAPK pathway mediates the phosphorylation of CREB. Knockdown of CREB prevents chordin expression and mesoderm dorsalization by the Spemann organizer, whereas ectopic expression of activated CREB-VP16 chimera induces chordin expression and dorsalizes mesoderm. Expression of high levels of p38 activator, MKK6E or CREB-VP16 in embryos converts ventral mesoderm into a dorsal organizing center. p38 MAPK and CREB function downstream of maternal Wnt/beta-catenin and the organizer-specific genes siamois and goosecoid. At low expression levels, MKK6E induces expression of lateral genes without inducing the expression of dorsal genes. Loss of CREB or p38 MAPK activity enables the expansion of the ventral homeobox gene vent1 into the dorsal marginal region, preventing the lateral expression of Xmyf5. Overall, these data indicate that dorsal-ventral mesoderm patterning is regulated by differential p38/CREB activities along the axis.
Collapse
Affiliation(s)
- Aviad Keren
- Department of Biochemistry, Rappaport Institute for Research in the Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology, P.O. Box 9649, Haifa 31096, Israel
| | | | | |
Collapse
|
17
|
Sakai M. Cell-autonomous and inductive processes among three embryonic domains control dorsal-ventral and anterior-posterior development of Xenopus laevis. Dev Growth Differ 2007; 50:49-62. [DOI: 10.1111/j.1440-169x.2007.00975.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
18
|
Adler R, Canto-Soler MV. Molecular mechanisms of optic vesicle development: complexities, ambiguities and controversies. Dev Biol 2007; 305:1-13. [PMID: 17335797 PMCID: PMC1927083 DOI: 10.1016/j.ydbio.2007.01.045] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Revised: 01/26/2007] [Accepted: 01/30/2007] [Indexed: 12/31/2022]
Abstract
Optic vesicle formation, transformation into an optic cup and integration with neighboring tissues are essential for normal eye formation, and involve the coordinated occurrence of complex cellular and molecular events. Perhaps not surprisingly, these complex phenomena have provided fertile ground for controversial and even contradictory results and conclusions. After presenting an overview of current knowledge of optic vesicle development, we will address conceptual and methodological issues that complicate research in this field. This will be done through a review of the pertinent literature, as well as by drawing on our own experience, gathered through recent studies of both intra- and extra-cellular regulation of optic vesicle development and patterning. Finally, and without attempting to be exhaustive, we will point out some important aspects of optic vesicle development that have not yet received enough attention.
Collapse
Affiliation(s)
- Ruben Adler
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21287-9257, USA.
| | | |
Collapse
|
19
|
Depew MJ, Simpson CA. 21st century neontology and the comparative development of the vertebrate skull. Dev Dyn 2006; 235:1256-91. [PMID: 16598716 DOI: 10.1002/dvdy.20796] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Classic neontology (comparative embryology and anatomy), through the application of the concept of homology, has demonstrated that the development of the gnathostome (jawed vertebrate) skull is characterized both by a fidelity to the gnathostome bauplan and the exquisite elaboration of final structural design. Just as homology is an old concept amended for modern purposes, so are many of the questions regarding the development of the skull. With due deference to Geoffroy-St. Hilaire, Cuvier, Owen, Lankester et al., we are still asking: How are bauplan fidelity and elaboration of design maintained, coordinated, and modified to generate the amazing diversity seen in cranial morphologies? What establishes and maintains pattern in the skull? Are there universal developmental mechanisms underlying gnathostome autapomorphic structural traits? Can we detect and identify the etiologies of heterotopic (change in the topology of a developmental event), heterochronic (change in the timing of a developmental event), and heterofacient (change in the active capacetence, or the elaboration of capacity, of a developmental event) changes in craniofacial development within and between taxa? To address whether jaws are all made in a like manner (and if not, then how not), one needs a starting point for the sake of comparison. To this end, we present here a "hinge and caps" model that places the articulation, and subsequently the polarity and modularity, of the upper and lower jaws in the context of cranial neural crest competence to respond to positionally located epithelial signals. This model expands on an evolving model of polarity within the mandibular arch and seeks to explain a developmental patterning system that apparently keeps gnathostome jaws in functional registration yet tractable to potential changes in functional demands over time. It relies upon a system for the establishment of positional information where pattern and placement of the "hinge" is driven by factors common to the junction of the maxillary and mandibular branches of the first arch and of the "caps" by the signals emanating from the distal-most first arch midline and the lamboidal junction (where the maxillary branch meets the frontonasal processes). In this particular model, the functional registration of jaws is achieved by the integration of "hinge" and "caps" signaling, with the "caps" sharing at some critical level a developmental history that potentiates their own coordination. We examine the evidential foundation for this model in mice, examine the robustness with which it can be applied to other taxa, and examine potential proximate sources of the signaling centers. Lastly, as developmental biologists have long held that the anterior-most mesendoderm (anterior archenteron roof or prechordal plate) is in some way integral to the normal formation of the head, including the cranial skeletal midlines, we review evidence that the seminal patterning influences on the early anterior ectoderm extend well beyond the neural plate and are just as important to establishing pattern within the cephalic ectoderm, in particular for the "caps" that will yield medial signaling centers known to coordinate jaw development.
Collapse
Affiliation(s)
- Michael J Depew
- Department of Craniofacial Development, King's College London, Guy's Hospital, London Bridge, London, United Kingdom.
| | | |
Collapse
|
20
|
Kumano G, Ezal C, Smith WC. ADMP2 is essential for primitive blood and heart development in Xenopus. Dev Biol 2006; 299:411-23. [PMID: 16959239 DOI: 10.1016/j.ydbio.2006.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2005] [Revised: 06/29/2006] [Accepted: 08/02/2006] [Indexed: 10/24/2022]
Abstract
We describe here the cloning of a new member of the TGF-beta family with similarity to the anti-dorsalizing morphogenetic proteins (ADMPs). This new gene, ADMP2, is expressed in a broad band of mesendoderm cells that appear to include the progenitors of the endoderm and the ventral mesoderm. Antisense morpholino oligonucleotide knockdown of ADMP2 results in near-complete disruption of primitive blood and heart development, while the development of other mesoderm derivatives, including pronephros, muscle and lateral plate is not disrupted. Moreover, the development of the primitive blood in ADMP2 knockdown embryos cannot be rescued by BMP. These results suggests that ADMP2 plays an early role in specifying presumptive ventral mesoderm in the leading edge mesoderm, and that ADMP2 activity may be necessary to respond to BMP signaling in the context of ventral mesoderm induction.
Collapse
Affiliation(s)
- Gaku Kumano
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | | | | |
Collapse
|
21
|
Cartry J, Nichane M, Ribes V, Colas A, Riou JF, Pieler T, Dollé P, Bellefroid EJ, Umbhauer M. Retinoic acid signalling is required for specification of pronephric cell fate. Dev Biol 2006; 299:35-51. [PMID: 16979153 DOI: 10.1016/j.ydbio.2006.06.047] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2006] [Revised: 06/20/2006] [Accepted: 06/27/2006] [Indexed: 10/24/2022]
Abstract
The mechanisms by which a subset of mesodermal cells are committed to a nephrogenic fate are largely unknown. In this study, we have investigated the role of retinoic acid (RA) signalling in this process using Xenopus laevis as a model system and Raldh2 knockout mice. Pronephros formation in Xenopus embryo is severely impaired when RA signalling is inhibited either through expression of a dominant-negative RA receptor, or by expressing the RA-catabolizing enzyme XCyp26 or through treatment with chemical inhibitors. Conversely, ectopic RA signalling expands the size of the pronephros. Using a transplantation assay that inhibits RA signalling specifically in pronephric precursors, we demonstrate that this signalling is required within this cell population. Timed antagonist treatments show that RA signalling is required during gastrulation for expression of Xlim-1 and XPax-8 in pronephric precursors. Moreover, experiments conducted with a protein synthesis inhibitor indicate that RA may directly regulate Xlim-1. Raldh2 knockout mouse embryos fail to initiate the expression of early kidney-specific genes, suggesting that implication of RA signalling in the early steps of kidney formation is evolutionary conserved in vertebrates.
Collapse
Affiliation(s)
- Jérôme Cartry
- Laboratoire de Biologie du Développement, équipe Signalisation et Morphogenèse, UMR CNRS 7622, Université Paris VI, 9 quai Saint-Bernard, 75005 Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Peng Y, Jiang BH, Yang PH, Cao Z, Shi X, Lin MCM, He ML, Kung HF. Phosphatidylinositol 3-kinase signaling is involved in neurogenesis during Xenopus embryonic development. J Biol Chem 2004; 279:28509-14. [PMID: 15123704 DOI: 10.1074/jbc.m402294200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Phosphatidylinositol 3-kinase (PI3K) has numerous cellular functions, including cell survival and proliferation. In this study, we demonstrated that the expression of the active form of PI3K induced dorsal differentiation and axis duplication and strongly induced the expression of neural markers. In contrast, the inhibition of PI3K activity by its dominant negative mutant induced the phenotype of losing posterior structures and the expression of ventral markers. Akt is an essential target of PI3K for neurogenesis. The expression of the active form of Akt induced axis duplication and increased the expression of neural markers. Inhibition of the Akt activity abolished the PI3K-induced double heads and axes. This signal transmits through its target, glycogen synthase kinase 3beta, which is known to mediate Wnt signaling for Xenopus development. These results identify a new function of PI3K/Akt signaling in axis formation and neurogenesis during Xenopus embryonic development and provide a direct link between growth factor-mediated PI3K/Akt signaling and Wnt signaling during embryonic development.
Collapse
Affiliation(s)
- Ying Peng
- Department of Neurology, Nanfang Hospital, The First Military Medical University, Guangzhou 510515, China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Cha SW, Hwang YS, Chae JP, Lee SY, Lee HS, Daar I, Park MJ, Kim J. Inhibition of FGF signaling causes expansion of the endoderm in Xenopus. Biochem Biophys Res Commun 2004; 315:100-6. [PMID: 15013431 DOI: 10.1016/j.bbrc.2004.01.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2003] [Indexed: 02/08/2023]
Abstract
Fibroblast growth factor (FGF) is established as an initiator of signaling events critical for neurogenesis and mesoderm formation during early Xenopus embryogenesis. However, less is known about the role FGF signaling plays in endoderm specification. Here, we show for the first time that endoderm-specific genes are induced when FGF signaling is blocked in animal cap explants. This block of FGF signaling is also responsible for a significant enhancement of endodermal gene expression in animal cap explants that are injected with a dominant-negative BMP-4 receptor (DNBR) RNA or treated with activin, however, neural and mesoderm gene expression is diminished. Consistent with these results, the injection of dominant-negative FGF receptor (DNFR) RNA expands endodermal cell fate boundaries while FGF treatment dramatically reduces endoderm in whole embryos. Taken together, these results indicate that inhibition of FGF signaling promotes endoderm formation, whereas the presence of active FGF signaling is necessary for neurogenesis/mesoderm formation.
Collapse
Affiliation(s)
- Sang-Wook Cha
- Department of Anatomy, School of Medicine, Kyungpook National University, Daegu 700-422, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Harrison CA, Gray PC, Koerber SC, Fischer W, Vale W. Identification of a functional binding site for activin on the type I receptor ALK4. J Biol Chem 2003; 278:21129-35. [PMID: 12665502 DOI: 10.1074/jbc.m302015200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activins, like other members of the transforming growth factor-beta (TGF-beta) superfamily, initiate signaling by assembling a complex of two types of transmembrane serine/threonine receptor kinases classified as type II (ActRII or ActRIIB) and type I (ALK4). A kinase-deleted version of ALK4 can form an inactive complex with activin and ActRII/IIB and thereby acts in a dominant negative manner to block activin signaling. Using the complex structure of bone morphogenetic protein-2 bound to its type I receptor (ALK3) as a guide, we introduced extracellular domain mutations in the context of the truncated ALK4 (ALK4-trunc) construct and assessed the ability of the mutants to inhibit activin function. We have identified five hydrophobic amino acid residues on the ALK4 extracellular domain (Leu40, Ile70, Val73, Leu75, and Pro77) that, when mutated to alanine, have substantial effects on ALK4-trunc dominant negative activity. In addition, eleven mutants partially affected activin binding to ALK4. Together, these residues likely constitute the binding surface for activin on ALK4. Cross-linking studies measuring binding of 125I-activin-A to the ALK4-trunc mutants in the presence of ActRII implicated the same residues. Our results indicate that there is only a partial overlap of the binding sites on ALK4 and ALK3 for activin-A and bone morphogenetic protein-2, respectively. In addition three of the residues required for activin binding to ALK4 are conserved on the type I TGF-beta receptor ALK5, suggesting the corresponding region on ALK5 may be important for TGF-beta binding.
Collapse
MESH Headings
- Activin Receptors, Type I/chemistry
- Activin Receptors, Type I/genetics
- Activin Receptors, Type I/metabolism
- Activins/metabolism
- Amino Acids/genetics
- Animals
- Binding Sites
- Bone Morphogenetic Protein Receptors, Type I
- Cells, Cultured
- Epithelial Cells/cytology
- Epithelial Cells/physiology
- Gene Expression
- Humans
- Kidney/cytology
- Lung/cytology
- Mink
- Molecular Sequence Data
- Mutagenesis
- Protein Serine-Threonine Kinases
- Protein Structure, Tertiary
- Proteins
- Receptor, Transforming Growth Factor-beta Type I
- Receptors, Growth Factor
- Receptors, Transforming Growth Factor beta/chemistry
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Sequence Homology, Amino Acid
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Craig A Harrison
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
25
|
Pownall ME, Welm BE, Freeman KW, Spencer DM, Rosen JM, Isaacs HV. An inducible system for the study of FGF signalling in early amphibian development. Dev Biol 2003; 256:89-99. [PMID: 12654294 DOI: 10.1016/s0012-1606(02)00120-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The use of a novel inducible FGF signalling system in the frog Xenopus laevis is reported. We show that the lipophilic, synthetic, dimerizing agent AP20187 is able to rapidly activate signalling through an ectopically expressed mutant form of FGFR1 (iFGFR1) in Xenopus embryos. iFGFR1 lacks an extracellular ligand binding domain and contains an AP20187 binding domain fused to the intracellular domain of mouse FGFR1. Induction of signalling by AP20187 is possible until at least early neurula stages, and we demonstrate that ectopically expressed iFGFR1 protein persists until late neurula stages. We show that activation of signalling through iFGFR1 can mimic a number of previously reported FGF activities, including mesoderm induction, repression of anterior development, and neural posteriorization. We show that competence to morphological posteriorization of the anteroposterior axis by FGF signalling only extends until about stage 10.5. We demonstrate that the competence of neural tissue to express the posterior markers Hoxa7 and Xcad3, in response to FGF signalling, is lost by the end of gastrula stages. We also show that activation of FGF signalling stimulates morphogenetic movements in neural tissue until at least the end of the gastrula stage.
Collapse
Affiliation(s)
- M E Pownall
- Department of Biology, PO Box 373, University of York, YO10 5YW, York, United Kingdom
| | | | | | | | | | | |
Collapse
|
26
|
Kumano G, Smith WC. Revisions to the Xenopus gastrula fate map: implications for mesoderm induction and patterning. Dev Dyn 2002; 225:409-21. [PMID: 12454919 DOI: 10.1002/dvdy.10177] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
A revised fate map of the gastrula Xenopus embryo predicts the existence of patterning mechanisms that operate within the animal/vegetal axis of the mesoderm-forming marginal zone. We review here molecular and embryologic data that demonstrate that such mechanisms are present and that they operate independently of the Spemann organizer. Evidence suggests that polarized fibroblast growth factor activity in the animal/vegetal axis patterns this axis. We present a model of mesoderm induction and patterning that integrates the new data on Spemann organizer-independent animal/vegetal patterning with data on other inductive pathways known to act on the gastrula marginal zone.
Collapse
Affiliation(s)
- Gaku Kumano
- Neuroscience Research Institute, and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California 93106, USA
| | | |
Collapse
|
27
|
Ogawa K, Ishihara S, Saito Y, Mineta K, Nakazawa M, Ikeo K, Gojobori T, Watanabe K, Agata K. Induction of a noggin-like gene by ectopic DV interaction during planarian regeneration. Dev Biol 2002; 250:59-70. [PMID: 12297096 DOI: 10.1006/dbio.2002.0790] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In previous studies, we have shown that dorsoventral (DV) interaction evokes not only blastema formation, but also morphogenetic events similar to those that occur in regeneration. However, it is still unclear what kinds of signal molecules are involved in the DV interaction. To investigate the signal systems involved in the DV interaction, we focused on a noggin-like gene (Djnlg) identified by the planarian EST project. Djnlg is the first noggin homologue isolated from an invertebrate. In DjNLG, the positions of nine cysteine residues which may be essential for dimer formation were well conserved, but overall, the amino acid sequence of DjNLG did not show high similarity to the sequences of vertebrate Noggins. Expression of Djnlg was observed only in the proximal region of the branch structures in the brain of intact planarians, suggesting that Djnlg may have a role in pattern formation in the brain. Interestingly, transient strong expression of Djnlg was observed in the amputated region of regenerating planarians. Djnlg-expressing cells were detected beneath the muscle 9 h after amputation and were then detected in the ventral subepidermal region of the blastema. The induction of Djnlg expression by amputation was not affected by X-ray irradiation, even though the stem cells were completely eliminated, implying the existence of signal-producing cells which may provide a positional cue to the stem cells. In DV reversed grafting, expression of Djnlg was strongly induced in the DV boundary between the host and donor. These results suggest that ectopic DV interaction may induce expression of Djnlg in the positional cue-producing cells, and that it might be involved in stimulation of blastema formation as well as DV patterning of the body.
Collapse
Affiliation(s)
- Kazuya Ogawa
- Laboratory of Regeneration Biology, Department of Life Science, Faculty of Science, Himeji Institute of Technology, Hyogo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Tanaka-Kunishima M, Takahashi K. Cleavage-arrested cell triplets from ascidian embryo differentiate into three cell types depending on cell combination and contact timing. J Physiol 2002; 540:153-76. [PMID: 11927677 PMCID: PMC2290224 DOI: 10.1113/jphysiol.2001.013293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2001] [Accepted: 01/04/2002] [Indexed: 11/08/2022] Open
Abstract
During early ascidian development, which is a prototype of early vertebrate development, anterior neuroectoderm cells (a4.2) from the eight-cell embryo are destined to become anterior neural structures including the brain vesicle, while presumptive notochordal neural cells (A4.1) become larval posterior neural structures including motoneurons. Whereas, an anterior quadrant cell (A3) of the four-cell embryo, from which both anterior neuroectoderm (a4.2) and notochordal neural cells (A4.1) are derived, has both fates. Cleavage-arrested cell triplets were prepared from the anterior quadrant cell and a pair of anterior neuroectoderm cells (A3-aa triplet) or a pair of presumptive notochordal neural cells (A3-AA triplet), and cultured in contact. Differentiation of cells in the triplet was determined electrophysiologically by observing cell type-specific currents. In the A3-aa triplet, when two neuroectoderm cells and an anterior quadrant cell were prepared from the same batch of embryos, all three cells in the triplet developed into neuronal cells in 60 % of cases, but in 40 % of cases all of them differentiated into epidermal cells. However, when the batch of embryos from which neuroectoderm cells were prepared was fertilized 3 h later than that from which the anterior quadrant cell was prepared all three cells in the triplet consistently became neuronal cells. In contrast, when the batch of embryos from which neuroectoderm cells were prepared was fertilized 3 h earlier, all three cells became epidermal. In the A3-AA triplet no switching of differentiation occurred and all three cells in the triplet differentiated into neuronal cells, although the amplitude of inward current was often small. In neuralized A3-aa triplets the spikes in the anterior quadrant cell were characteristically small in amplitude and brief in duration, suggesting the presence of A-currents, which is a characteristic feature of posterior neuronal differentiation. In contrast, the spikes in the anterior neuroectoderm cells were large in amplitude and long in duration, chracteristic to the anterior neuronal type. The majority of single isolated anterior quadrant cells became non-excitable. However, the minority was apparently autonomously neuralized to become the posterior neuronal type. In neuralized A3-AA triplets, the majority of anterior quadrant cells was induced to become the anterior neuronal type. When isolated anterior quadrant cells were neuralized with subtilisin, a protease, they also predominantly became the anterior neuronal type. While, in medium containing a fibroblast growth factor posterior neuralization of isolated anterior quadrant cells was facilitated, but the anterior neuronal type, although minor, appeared anew. These observations indicate that the multiple fates of the anterior quadrant cell expressed in vivo were effectively reproduced in this experimental condition at the single cell level. Interactive differentiation in this triplet system recapitulates not only fundamental neural induction of ascidian neuroectoderm cells, but also functional and positional specificity within the neuronal group.
Collapse
Affiliation(s)
- Motoko Tanaka-Kunishima
- Department of Medical Physiology, Meiji Pharmaceutical University, Noshio 2-522-1, Kiyose, Tokyo MZC204-8588, Japan.
| | | |
Collapse
|
29
|
Falchuk KH, Contin JM, Dziedzic TS, Feng Z, French TC, Heffron GJ, Montorzi M. A role for biliverdin IXalpha in dorsal axis development of Xenopus laevis embryos. Proc Natl Acad Sci U S A 2002; 99:251-6. [PMID: 11782548 PMCID: PMC117547 DOI: 10.1073/pnas.012616099] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2001] [Accepted: 11/19/2001] [Indexed: 11/18/2022] Open
Abstract
The determinants of Xenopus laevis embryos that act before their first cell division are mandatory for the formation of mRNas required to establish the dorsal axis. Although their chemical identities are unknown, a number of their properties have long been recognized. One of the determinants is present in the cytoplasm and is sensitive to UV light. Thus, exposing stage 1 embryos to either standard 254-nm or, as shown here, to 366-nm UV light during the 0.3-0.4 time fraction of their first cycle inactivates the cytoplasmic determinant. As a consequence, both types of irradiated embryos fail to express dorsal markers, e.g., goosecoid and chordin, without affecting formation of ventral markers, e.g., Vent-1. The developmental outcome is dorsal axis-deficient morphology. We report here that biliverdin IXalpha, a normal constituent of cytoplasmic yolk platelets, is photo-transformed by irradiation with either 254- or 366-nm UV light and that the transformation triggers the dorsal axis deficiency. When the 254- or 366-nm UV-irradiated embryos, fated to dorsal axis deficiency, are incubated solely with microM amounts of biliverdin, they recover and form the axis. In contrast, incubation with either in vitro photo-transformed biliverdin or biliverdin IXalpha dimethyl ester does not induce recovery. The results define an approach to produce dorsal axis-deficient embryos by photo-transforming its biliverdin by irradiation with 366-nm UV light and identify an unsuspected role for biliverdin IXalpha in X. laevis embryogenesis.
Collapse
Affiliation(s)
- Kenneth H Falchuk
- Center for Biochemical and Biophysical Sciences and Medicine, Harvard Medical School, Cambridge, MA 02139, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Muñoz-Sanjuán I, H-Brivanlou A. Early posterior/ventral fate specification in the vertebrate embryo. Dev Biol 2001; 237:1-17. [PMID: 11518501 DOI: 10.1006/dbio.2001.0350] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
One of the central questions in developmental biology is that of how one cell can give rise to all specialized cell types and organs in the organism. Within the embryo, all tissues are composed of cells derived from one or more of the three germ layers, the ectoderm, the mesoderm, and the endoderm. Understanding the molecular events that underlie both the specification and patterning of the germ layers has been a long-standing interest for developmental biologists. Recent years have seen a rapid advancement in the elucidation of the molecular players implicated in patterning the vertebrate embryo. In this review, we will focus solely on the ventral and posterior fate acquisition in the ventral-lateral domains of the pregastrula embryo. We will address the embryonic origins of various tissues and will present embryological and experimental evidence to illustrate how "classically defined" ventral and posterior structures develop in all three germ layers. We will discuss the status of our current knowledge by focusing on the African frog Xenopus laevis, although we will also gather evidence from other vertebrates, where available. In particular, genetic studies in the zebrafish and mouse have been very informative in addressing the requirement for individual genes in these processes. The amphibian system has enjoyed great interest since the early days of experimental embryology, and constitutes the best understood system in terms of early patterning signals and axis specification. We want to draw interest to the embryological origins of cells that will develop into what we have collectively termed "posterior" and "ventral" cells/tissues, and we will address the involvement of the major signaling pathways implicated in posterior/ventral fate specification. Particular emphasis is given as to how these signaling pathways are integrated during early development for the specification of posterior and ventral fates.
Collapse
Affiliation(s)
- I Muñoz-Sanjuán
- Laboratory of Vertebrate Embryology, The Rockefeller University, New York, New York 10021, USA
| | | |
Collapse
|
31
|
Abstract
The filamentous cyanobacterium Anabaena sp. strain PCC 7120 forms a developmental pattern of single heterocysts separated by approximately 10 vegetative cells. Heterocysts differentiate from vegetative cells and are specialized for nitrogen fixation. The patS gene, which encodes a small peptide that inhibits heterocyst differentiation, is expressed in proheterocysts and plays a critical role in establishing the heterocyst pattern. Here we present further analysis of patS expression and heterocyst pattern formation. A patS-gfp reporter strain revealed clusters of patS-expressing cells during the early stage of heterocyst differentiation. PatS signaling is likely to be involved in the resolution of these clusters. Differentiating cells were inhibited by PatS during the time period 6 to 12 h after heterocyst induction, when groups of differentiating cells were being resolved to a single proheterocyst. Increased transcription of patS during development coincided with expression from a new transcription start site. In vegetative cells grown on nitrate, the 5' end of a transcript for patS was localized 314 bases upstream from the first translation initiation codon. After heterocyst induction, a new transcript with a 5' end at -39 bases replaced the vegetative cell transcript. A patS mutant grown for several days under nitrogen-fixing conditions showed partial restoration of the normal heterocyst pattern, presumably because of a gradient of nitrogen compounds supplied by the heterocysts. The patS mutant formed heterocysts when grown in the presence of nitrate but showed no nitrogenase activity and no obvious heterocyst pattern. We conclude that PatS and products of nitrogen fixation are the main signals determining the heterocyst pattern.
Collapse
Affiliation(s)
- H S Yoon
- Department of Biology, Texas A&M University, College Station, Texas 77843-3258, USA
| | | |
Collapse
|
32
|
Kalcheim C. Mechanisms of early neural crest development: from cell specification to migration. INTERNATIONAL REVIEW OF CYTOLOGY 2001; 200:143-96. [PMID: 10965468 DOI: 10.1016/s0074-7696(00)00004-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The neural crest is a group of embryonic progenitors that forms during the process of neurulation by interactions that take place between the prospective epidermis and the specified neuroectoderm. Although initially an integral part of the neuroepithelium, neural crest cells separate from the central nervous system primordium by a process of epitheliomesenchymal transition and become a motile cell population. These mesenchymal cells then migrate through stereotypic pathways, some of which are common and others unique to various vertebrate species. Furthermore, the availability of distinct migratory pathways also differs according to embryonic stage and axial level. Studies have begun to address the molecular basis of neural crest specification, delamination, and migration. The present review summarizes some major advances in our understanding of the nature of the intercellular interactions and the molecules that mediate them during early phases of neural crest ontogeny.
Collapse
Affiliation(s)
- C Kalcheim
- Department of Anatomy and Cell Biology, Hebrew University of Jerusalem, Hadassah Medical School, Israel
| |
Collapse
|
33
|
Dhanasekaran SM, Vempati UD, Kondaiah P. Isolation and characterization of a transforming growth factor-beta Type II receptor cDNA from Xenopus laevis. Gene 2001; 263:171-8. [PMID: 11223255 DOI: 10.1016/s0378-1119(00)00575-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Transforming Growth Factor-beta (TGF-beta) and their receptors have been characterized from many organisms. Two TGF-beta signaling receptors called Type I and II have been described for various ligands of the superfamily from organisms ranging from Drosophila to humans. In Xenopus laevis, TGF-beta2 and 5 have been reported and presumably, play important roles during early development. Several Type I and type II receptors for many ligands of the TGF-beta superfamily except TGF-beta type II receptor (TbetaIIR), have been characterized in Xenopus laevis. A chemical cross linking experiment using iodinated TGF-beta1 and -beta5, revealed four specific binding proteins on XTC cells. In order to understand the TGF-beta involvement during Xenopus development, a TGF-beta type II receptor (XTbetaIIR) has been isolated from a XTC cDNA library. XTbetaIIR was a partial cDNA lacking a portion of the signal peptide. The sequence analysis and homology comparison with the human TbetaIIR revealed 67% amino acid similarity in the extra cellular domain, 60% similarity in the transmembrane domain and 87% similarity in the cytoplasmic kinase domain, suggesting that XTbetaIIR is a putative TGF-beta type II receptor. In addition, the consensus amino acid motif for serine threonine receptor kinases was also present. Further, a dominant negative expression construct lacking the cytoplasmic kinase domain (engineered with the signal peptide from human TGF-beta type II receptor), was able to abolish TGF-beta mediated induction of a luciferase reporter plasmid, in a transient cell transfection assay. This substantiates the notion that XTbetaIIR cDNA can act as a receptor for TGF-beta. RT-PCR analysis using RNA isolated from various developmental stages of Xenopus laevis revealed expression of this gene in all the early stages of development and in the adult organs, except in stages 46/48.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Cross-Linking Reagents
- DNA, Complementary/chemistry
- DNA, Complementary/genetics
- DNA, Complementary/isolation & purification
- Gene Expression Regulation, Developmental
- Humans
- Luciferases/genetics
- Luciferases/metabolism
- Molecular Sequence Data
- Protein Kinases/genetics
- Protein Serine-Threonine Kinases
- Protein Structure, Tertiary
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Transforming Growth Factor beta/chemistry
- Receptors, Transforming Growth Factor beta/genetics
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Sequence Alignment
- Sequence Analysis, DNA
- Sequence Homology, Amino Acid
- Transforming Growth Factor beta/chemistry
- Tumor Cells, Cultured
- Xenopus laevis/embryology
- Xenopus laevis/genetics
- Xenopus laevis/growth & development
Collapse
Affiliation(s)
- S M Dhanasekaran
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560 012, India
| | | | | |
Collapse
|
34
|
Abstract
The embryonic vasculature forms by the processes of vasculogenesis and angiogenesis. Angioblasts (endothelial cell precursors) appear to be induced by fibroblast growth factor 2 (FGF-2). The angioblasts contributing to the dorsal aortae arise by an epithelial to mesenchymal transformation of cells originating from the splanchnic mesoderm. QH-l and vascular endothelial growth factor receptor 2 (VEGFR-2) both appear to label these cells as they adopt a mesenchymal morphology. Since VEGFR-2 is the earliest known VEGF receptor this suggests that VEGF is not involved in angioblast induction. VEGF does appear to be critical, however, for growth and morphogenesis of angioblasts into the initial vascular pattern. Controlled delivery of FGF-2 from beads and aggregates of cells transfected with quail VEGF have been used in our laboratory to study the role of these growth factors in angioblast induction and migration. We have induced cells from the epithelial quail somite to differentiate into angioblasts with FGF-2 both in the embryo and in culture. This is a useful model system to study the origins of endothelial cells that are normally more diffusely induced during gastrulation by an obscure process probably involving signals from the embryonic endoderm. The origins of arterial versus venous endothelial cells is also poorly understood but recent findings on the distribution of ephrins and Eph receptors suggest that molecular differences exist prior to the onset of circulation. Finally, studies on the role of growth factors in such diverse phenomena as stem cell biology, angiogenesis, and molecular medicine in addition to vascular development suggest multiple roles for FGF-2 and VEGF in vascular development.
Collapse
Affiliation(s)
- T J Poole
- State University of New York Upstate Medical University, Department of Cell and Developmental Biology, Syracuse, New York 13210, USA
| | | | | |
Collapse
|
35
|
Metzler DE, Metzler CM, Sauke DJ. Growth and Development. Biochemistry 2001. [DOI: 10.1016/b978-012492543-4/50035-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
36
|
Abstract
According to the three-signal model of mesoderm patterning in Xenopus, all mesoderm, with the exception of the Spemann organizer, is originally specified as ventral type, such as lateral plate and primary blood islands. It is proposed that the blood islands become restricted to the ventralmost mesoderm because they are not exposed to the BMP-inhibiting activity of the Spemann organizer. We present evidence here that, contrary to predictions of this model, the blood islands remain ventrally restricted even in the absence of Spemann organizer signaling. We further observed that inhibition of FGF signaling with a dominant negative receptor resulted in the expansion of the blood island-forming territory with a concomitant loss of somite. The requirement for FGF signaling in specifying somite versus blood island territories was observed as early as midgastrulation. The nonoverlapping expression domains of Xnr-2 and Xbra in the gastrula marginal zone appear to mark presumptive blood island and somite, respectively. Inhibition of FGF signaling with dominant negative receptor leads to an expansion of Xnr-2 expression and to a corresponding reduction in Xbra expression. On the other hand, we found no evidence that manipulation of BMP signaling, either positively or negatively, altered the expression domains of Xnr-2 and Xbra. These results suggest that FGF signaling, rather than BMP-inhibiting activity, is essential for restriction of the ventral blood islands to ventral mesoderm.
Collapse
Affiliation(s)
- G Kumano
- Department of Molecular, Cellular and Developmental Biology, University of California at Santa Barbara, Santa Barbara, California 93106, USA
| | | |
Collapse
|
37
|
Adler CE, Miyoshi-Akiyama T, Aleman LM, Tanaka M, Smith JM, Mayer BJ. Abl family kinases and Cbl cooperate with the Nck adaptor to modulate Xenopus development. J Biol Chem 2000; 275:36472-8. [PMID: 10967110 DOI: 10.1074/jbc.m005424200] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We previously showed that overexpression of the Nck Src homology (SH) 2/SH3 adaptor in Xenopus embryos induced developmental defects including anterior truncation and mesoderm ventralization. Mutagenic analysis indicated that this was due to relocalization of endogenous proteins that bind the first two SH3 domains of Nck. We therefore screened a Xenopus expression library with Nck SH3 domains to identify Nck-interacting proteins, and evaluated candidate binding proteins for a potential role in Nck-induced anterior truncation/ventralization. Of 39 binding proteins analyzed, only the Abl-related kinase Arg and the Cbl proto-oncogene product bound preferentially to the first two SH3 domains in tandem compared with the individual domains, consistent with a role in the developmental phenotype. High level overexpression of c-Abl or Arg alone induced anterior truncation, as did lower levels of an activated form of Abl; Cbl alone had no effect. In a sensitized system where subthreshold amounts of a ventralizing Nck mutant were expressed, co-expression of the combination of Abl or Arg and Cbl at modest levels strongly potentiated anterior truncation, while Arg, Abl, or Cbl alone were without effect. These results suggest a role for both Cbl and Abl family kinases in patterning the Xenopus embryo.
Collapse
Affiliation(s)
- C E Adler
- Laboratory of Molecular Medicine, Children's Hospital and Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
38
|
Wall NA, Craig EJ, Labosky PA, Kessler DS. Mesendoderm induction and reversal of left-right pattern by mouse Gdf1, a Vg1-related gene. Dev Biol 2000; 227:495-509. [PMID: 11071769 DOI: 10.1006/dbio.2000.9926] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
TGFbeta signals play important roles in establishing the body axes and germ layers in the vertebrate embryo. Vg1 is a TGFbeta-related gene that, due to its maternal expression and vegetal localization in Xenopus, has received close examination as a potential regulator of development in Xenopus, zebrafish, and chick. However, a mammalian Vg1 ortholog has not been identified. To isolate mammalian Vg1 we screened a mouse expression library with a Vg1-specific monoclonal antibody and identified a single cross-reactive clone encoding mouse Gdf1. Gdf1 is expressed uniformly throughout the embryonic region at 5.5-6.5 days postcoitum and later in the node, midbrain, spinal cord, paraxial mesoderm, lateral plate mesoderm, and limb bud. When expressed in Xenopus embryos, native GDF1 is not processed, similar to Vg1. In contrast, a chimeric protein containing the prodomain of Xenopus BMP2 fused to the GDF1 mature domain is efficiently processed and signals via Smad2 to induce mesendoderm and axial duplication. Finally, right-sided expression of chimeric GDF1, but not native GDF1, reverses laterality and results in right-sided Xnr1 expression and reversal of intestinal and heart looping. Therefore, GDF1 can regulate left-right patterning, consistent with the Gdf1 loss-of-function analysis in the mouse (C. T. Rankin, T. Bunton, A. M. Lawler, and S. J. Lee, 2000, Nature Genet. 24, 262-265) and a proposed role for Vg1 in Xenopus. Our results establish that Gdf1 is posttranslationally regulated, that mature GDF1 activates a Smad2-dependent signaling pathway, and that mature GDF1 is sufficient to reverse the left-right axis. Moreover, these findings demonstrate that GDF1 and Vg1 are equivalent in biochemical and functional assays, suggesting that Gdf1 provides a Vg1-like function in the mammalian embryo.
Collapse
Affiliation(s)
- N A Wall
- Biology Department, Lawrence University, Appleton, Wisconsin 54912, USA
| | | | | | | |
Collapse
|
39
|
Niehrs C, Dosch R, Onichtchouk D. Embryonic patterning of Xenopus mesoderm by Bmp-4. ERNST SCHERING RESEARCH FOUNDATION WORKSHOP 2000:165-90. [PMID: 10943310 DOI: 10.1007/978-3-662-04264-9_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- C Niehrs
- Division of Molecular Embryology, Deutsches Krebsforschungszentrum, Heidelberg, Germany
| | | | | |
Collapse
|
40
|
Wacker S, Grimm K, Joos T, Winklbauer R. Development and control of tissue separation at gastrulation in Xenopus. Dev Biol 2000; 224:428-39. [PMID: 10926778 DOI: 10.1006/dbio.2000.9794] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
During Xenopus gastrulation, the internalizing mesendodermal cell mass is brought into contact with the multilayered blastocoel roof. The two tissues do not fuse, but remain separated by the cleft of Brachet. This maintenance of a stable interface is a precondition for the movement of the two tissues past each other. We show that separation behavior, i.e., the property of internalized cells to remain on the surface of the blastocoel roof substratum, spreads before and during gastrulation from the vegetal endoderm into the anterior and eventually the posterior mesoderm, roughly in parallel to internalization movement. Correspondingly, the blastocoel roof develops differential repulsion behavior, i.e., the ability to specifically repell cells showing separation behavior. From the effects of overexpressing wild-type or dominant negative XB/U or EP/C cadherins we conclude that separation behavior may require modulation of cadherin function. Further, we show that the paired-class homeodomain transcription factors Mix.1 and gsc are involved in the control of separation behavior in the anterior mesoderm. We present evidence that in this function, Mix.1 and gsc may cooperate to repress transcription.
Collapse
Affiliation(s)
- S Wacker
- Universität zu Köln, Zoologisches Institut, Köln, Germany
| | | | | | | |
Collapse
|
41
|
Xu RH, Peng Y, Fan J, Yan D, Yamagoe S, Princler G, Sredni D, Ozato K, Kung HF. Histone acetylation is a checkpoint in FGF-stimulated mesoderm induction. Dev Dyn 2000; 218:628-35. [PMID: 10906781 DOI: 10.1002/1097-0177(2000)9999:9999<::aid-dvdy1024>3.0.co;2-h] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
We have previously demonstrated that the transcription factor, AP-1 (c-jun/c-fos heterodimer), mediates fibroblast growth factor (FGF) signaling during mesoderm induction in Xenopus embryo. In the present studies, we show that histone acetylation is involved in FGF-mediated signaling leading to mesoderm induction. Histone acetylation is a dynamic process regulated by the activities of two histone-modifying enzymes, the histone acetyltransferase(s) and histone deacetylase(s) (HDACs). We found that basal and FGF-regulated activator protein 1 (AP-1) activity in Xenopus embryo is markedly reduced by treatment of trichostatin A (TSA), a specific inhibitor of HDAC. However, activity of another transcription factor, NFkappaB, is enhanced by TSA treatment. AP-1-mediated mesoderm induction in the animal caps is dramatically suppressed by TSA at a dose-dependent manner. This suppression can be rescued by ectopic expression of HDAC3 at early stage. Finally, we found that histone acetylation in animal caps is inhibited by FGF whereas enhanced by TSA (as a control). Therefore, we propose that histone acetylation is a checkpoint for transduction of the FGF/AP-1 signals to induce mesoderm. Published 2000 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- R H Xu
- WiCell Research Institute, Wisconsin Alumni Research Foundation, University of Wisconsin, Madison, Wisconsin 53705-7365, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kaufman CD, Martínez-Rodriguez G, Hackett PB. Ectopic expression of c-ski disrupts gastrulation and neural patterning in zebrafish. Mech Dev 2000; 95:147-62. [PMID: 10906458 DOI: 10.1016/s0925-4773(00)00351-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The c-ski proto-oncogene encodes a transcriptional regulator that has been implicated in the development of different tissues at different times during vertebrate development. We identified two novel paralogues of the c-ski gene family, skiA and skiB in zebrafish (Danio rerio). The skiA protein is maternal and ubiquitous while skiB is zygotic. Overexpression of SkiA or SkiB disrupted gastrulation and resulted in a dorsalized phenotype. In situ analyses suggested that overexpression of Ski leads to a slight expansion of dorsal-axial mesoderm, diminishment or loss of ventral mesoderm and radialization of dorsal neuroectoderm. The dorsalized phenotype could be rescued by the ventral specifying factor, BMP4. These results provide evidence that Ski proteins participate in dorsal-ventral specification of both neuroectoderm and mesoderm.
Collapse
Affiliation(s)
- C D Kaufman
- Department of Genetics, Cell Biology and Development, University of Minnesota, 1445 Gortner Avenue, St. Paul, MN 55108-1095, USA
| | | | | |
Collapse
|
43
|
Lanford PJ, Platt C, Popper AN. Structure and function in the saccule of the goldfish (Carassius auratus): a model of diversity in the non-amniote ear. Hear Res 2000; 143:1-13. [PMID: 10771179 DOI: 10.1016/s0378-5955(00)00015-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The vertebrate inner ear is comprised of a remarkable diversity of cell types, including several types of sensory hair cells. In amniotes (reptiles, birds, and mammals), the morphological and physiological characteristics that distinguish these cell types have been well documented, while cellular variation in the ears of non-amniotes (all other vertebrate groups) has remained underrecognized. Since non-amniotes have become increasingly popular models for developmental and genetic research, a more comprehensive understanding of structure and function in the inner ears of these species is warranted. This paper first reviews the large body of data describing the morphology and physiology of hair cells and afferent neurons in the inner ear of the goldfish (Carassius auratus). In particular, we examine the structure of the goldfish saccule, an endorgan that has been the subject of numerous investigations on audition. New data on the structural variation of synaptic bodies in saccular hair cells are also presented, and the functional implications of these data are discussed. Finally, we conclude that hair cell structure varies along the length of the goldfish saccule in a manner consistent with known physiological characteristics of the endorgan. The saccule provides an excellent model for investigating structure-function relationships in the vertebrate inner ear, as well as the development of auditory and vestibular sensory epithelia.
Collapse
Affiliation(s)
- P J Lanford
- Department of Biology, University of Maryland, College Park, MD, USA.
| | | | | |
Collapse
|
44
|
Nagano T, Ito Y, Tashiro K, Kobayakawa Y, Sakai M. Dorsal induction from dorsal vegetal cells in Xenopus occurs after mid-blastula transition. Mech Dev 2000; 93:3-14. [PMID: 10781935 DOI: 10.1016/s0925-4773(00)00251-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We performed some experiments to investigate the temporal and spatial details of the dorsal induction exerted by dorsal vegetal cells in Xenopus embryo. Two dorsal vegetal cells (D1 cells) were transplanted into the ventral vegetal region of a recipient at the 32-cell stage. At various times after transplantation, the ventral animal-equatorial part was explanted and cultured. The explants isolated 5.5 h after transplantation (time 5.5) elongated and formed somites. In RT-PCR analysis, the expression of dorsal gene, chordin was activated in the explants isolated after time 4.0 (about the 4000-cell stage which corresponds to the mid blastula transition (MBT)) at control stage 10. In another series of experiments, ventral animal-equatorial and dorsal vegetal parts were isolated from the 4000-cell stage embryos and they were combined for 2.0-2.5 h. These ventral animal-equatorial explants elongated and formed somites. The chordin expression was also observed in the explants. But the 32- and 256-cell stage dorsal vegetal cells failed to exert the dorsalizing activity within the 2.0-2.5 h of the conjugation. These results suggest that 2 h contact after MBT is necessary and sufficient for the dorsal induction from the dorsal vegetal cells and it occurs as a result of the zygotic gene expression. Consistent with this idea, the zygotic dorsal genes, siamois and chordin were expressed on the upper regions of the transplanted D1 descendants at stage 10. Furthermore, this region began to gastrulate when the D1 cell was transplanted with upside-down orientation. Our data indicate that the upper region of the D1 descendants by itself act as the Spemann organizer rather than the Nieuwkoop center.
Collapse
Affiliation(s)
- T Nagano
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan.
| | | | | | | | | |
Collapse
|
45
|
Agius E, Oelgeschläger M, Wessely O, Kemp C, De Robertis EM. Endodermal Nodal-related signals and mesoderm induction in Xenopus. Development 2000; 127:1173-83. [PMID: 10683171 PMCID: PMC2292107 DOI: 10.1242/dev.127.6.1173] [Citation(s) in RCA: 277] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In Xenopus, mesoderm induction by endoderm at the blastula stage is well documented, but the molecular nature of the endogenous inductive signals remains unknown. The carboxy-terminal fragment of Cerberus, designated Cer-S, provides a specific secreted antagonist of mesoderm-inducing Xenopus Nodal-Related (Xnr) factors. Cer-S does not inhibit signalling by other mesoderm inducers such as Activin, Derriere, Vg1 and BMP4, nor by the neural inducer Xnr3. In the present study we show that Cer-S blocks the induction of both dorsal and ventral mesoderm in animal-vegetal Nieuwkoop-type recombinants. During blastula stages Xnr1, Xnr2 and Xnr4 are expressed in a dorsal to ventral gradient in endodermal cells. Dose-response experiments using cer-S mRNA injections support the existence of an endogenous activity gradient of Xnrs. Xnr expression at blastula can be activated by the vegetal determinants VegT and Vg1 acting in synergy with dorsal (beta)-catenin. The data support a modified model for mesoderm induction in Xenopus, in which mesoderm induction is mediated by a gradient of multiple Nodal-related signals released by endoderm at the blastula stage.
Collapse
Affiliation(s)
- E Agius
- Howard Hughes Medical Institute, Department of Biological Chemistry, University of California, Los Angeles, CA 90095-1662, USA
| | | | | | | | | |
Collapse
|
46
|
Abstract
Development of an organ is directed by cell and tissue interactions and these also occur during the formation of functional kidney. During vertebrate development inductive signalling between mesenchyme and epithelium controls the organogenesis of all three kinds of kidneys: pronephros, mesonephros and metanephros. In higher animals the metanephros differentiates into the permanent kidney and in this review we will mainly concentrate on its development. Molecular interactions currently known to function during nephrogenesis have primarily been based on the use of knockout techniques. These studies have highlighted the role for transcription factors, signalling molecules, growth factors and their receptors and also for extracellular matrix components in kidney development. Finally in this review we will represent our own model for kidney development according to the knowledge of the genes involved in the development of the functional excretory organ, kidney.
Collapse
Affiliation(s)
- S Kuure
- Department of Biochemistry and Biocenter Oulu, Faculties of Science and Medicine, University of Oulu, FIN-90570, Oulu, Finland
| | | | | |
Collapse
|
47
|
Cleaver O, Seufert DW, Krieg PA. Endoderm patterning by the notochord: development of the hypochord in Xenopus. Development 2000; 127:869-79. [PMID: 10648245 DOI: 10.1242/dev.127.4.869] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The patterning and differentiation of the vertebrate endoderm requires signaling from adjacent tissues. In this report, we demonstrate that signals from the notochord are critical for the development of the hypochord, which is a transient, endodermally derived structure that lies immediately ventral to the notochord in the amphibian and fish embryo. It appears likely that the hypochord is required for the formation of the dorsal aorta in these organisms. We show that removal of the notochord during early neurulation leads to the complete failure of hypochord development and to the elimination of expression of the hypochord marker, VEGF. Removal of the notochord during late neurulation, however, does not interfere with hypochord formation. These results suggest that signals arising in the notochord instruct cells in the underlying endoderm to take on a hypochord fate during early neural stages, and that the hypochord does not depend on further notochord signals for maintenance. In reciprocal experiments, when the endoderm receives excess notochord signaling, a significantly enlarged hypochord develops. Overall, these results demonstrate that, in addition to patterning neural and mesodermal tissues, the notochord plays an important role in patterning of the endoderm.
Collapse
Affiliation(s)
- O Cleaver
- Division of Molecular Cell and Developmental Biology, School of Biological Sciences, University of Texas at Austin, Austin, TX 78712, USA
| | | | | |
Collapse
|
48
|
Gray PC, Greenwald J, Blount AL, Kunitake KS, Donaldson CJ, Choe S, Vale W. Identification of a binding site on the type II activin receptor for activin and inhibin. J Biol Chem 2000; 275:3206-12. [PMID: 10652306 DOI: 10.1074/jbc.275.5.3206] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Type II activin receptors (ActRII and ActRIIB) are single-transmembrane domain serine/threonine kinase receptors that bind activin to initiate the signaling and cellular responses triggered by this hormone. Inhibin also binds type II activin receptors and antagonizes many activin effects. Here we describe alanine scanning mutagenesis of the ActRII extracellular domain. We identify a cluster of three hydrophobic residues (Phe(42), Trp(60), and Phe(83)) that, when individually mutated to alanine in the context of the full-length receptor, cause the disruption of activin and inhibin binding to ActRII. Each of the alanine-substituted ActRII mutants retaining activin binding maintains the ability to form cross-linked complexes with activin and supports activin cross-linking to the type I activin receptor ALK4. Unlike wild-type ActRII, the three mutants unable to bind activin do not cause an increase in activin signaling when transiently expressed in a corticotroph cell line. Together, our results implicate these residues in forming a critical binding surface on ActRII required for functional interactions with both activin and inhibin. This first identification of a transforming growth factor-beta family member binding site may provide a general basis for characterizing binding sites for other members of the superfamily.
Collapse
Affiliation(s)
- P C Gray
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
The demonstration of the existence of tissue-specific adult stem cells has had a great impact on our understanding of stem cell biology and its application in clinical medicine. Their existence has revolutionized the implications for the treatment of many degenerative diseases characterized by either the loss or malfunction of discrete cell types. However, successful exploitation of this opportunity requires that we have sufficient know-how of stem cell manipulation. Because stem cells are the founders of virtually all tissues during embryonic development, we believe that understanding the cellular and molecular mechanisms of embryogenesis and organogenesis will ultimately serve as a platform to identify factors and conditions that regulate stem cell behavior. Discovery of stem cell regulatory factors will create potential pharmaceutical opportunities for treatment of degenerative diseases, as well as providing critical knowledge of the processes by which stem cells can be expanded in vitro, differentiated, and matured into desired functional cells for implantation into humans. A well-characterized example of this is the hematopoietic system where the discovery of erythropoietin (EPO) and granulocyte-colony stimulating factor (G-CSF), which regulate hematopoietic progenitor cell behavior, have provided significant clinical success in disease treatment as well as providing important insights into hematopoiesis. In contrast, little is known about the identity of pancreatic stem cells, the focus of this review. Recent reports of the potential existence of pancreatic stem cells and their utility in rescuing the diabetic state now raise the same possibilities of generating insulin-producing beta cells as well as other cell types of the pancreatic islet from a stem cell. In this review, we will focus on the potential of these new developments and how our understanding of pancreas development can help design strategies and approaches by which a cell replacement therapy can be implemented for the treatment of insulin-dependent diabetes which is manifested by the loss of beta cells in the pancreas.
Collapse
Affiliation(s)
- M Peshavaria
- Ontogeny, Inc, Cambridge, Massachusetts 02138-1118, USA.
| | | |
Collapse
|
50
|
Varga ZM, Wegner J, Westerfield M. Anterior movement of ventral diencephalic precursors separates the primordial eye field in the neural plate and requires cyclops. Development 1999; 126:5533-46. [PMID: 10572031 DOI: 10.1242/dev.126.24.5533] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A currently favored hypothesis postulates that a single field of cells in the neural plate forms bilateral retinas. To learn how retinal precursors segregate, we followed individual labeled neural plate cells in zebrafish. In the late gastrula, a single field of odd-paired-like-expressing cells contributed to both retinas, bordered posteriorly by diencephalic precursors expressing mariposa. Median mariposa-expressing cells moved anteriorly, separating the eyes, and formed ventral anterior diencephalon, the presumptive hypothalamus. In cyclops mutants, corresponding cells failed to move anteriorly, a ventral diencephalon never formed, and the eyes remained fused. Ablation of the region containing these cells induced cyclopia in wild types. Our results indicate that movement of a median subpopulation of diencephalic precursors separates retinal precursors into left and right eyes. Wild-type cyclops gene function is required for these morphogenetic movements.
Collapse
Affiliation(s)
- Z M Varga
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA.
| | | | | |
Collapse
|