1
|
Alvarez S, Gupta S, Mercado-Ayon Y, Honeychurch K, Rodriguez C, Kawaguchi R, Butler SJ. Netrin1 patterns the dorsal spinal cord through modulation of Bmp signaling. Cell Rep 2024; 43:114954. [PMID: 39547237 PMCID: PMC11756817 DOI: 10.1016/j.celrep.2024.114954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 09/06/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
We have identified an unexpected role for netrin1, a canonical axonal guidance cue, as a suppressor of bone morphogenetic protein (Bmp) signaling in the developing dorsal spinal cord. Using a combination of gain- and loss-of-function approaches in chicken and mouse embryonic models, as well as mouse embryonic stem cells (mESCs), we have observed that manipulating the level of netrin1 specifically alters the patterning of the Bmp-dependent dorsal interneurons (dIs), dI1-dI3. Altered netrin1 levels also change Bmp signaling activity, as assessed using bioinformatic approaches, as well as monitoring phosophoSmad1/5/8 activation, the canonical intermediate of Bmp signaling, and Id levels, a known Bmp target. Together, these studies support the hypothesis that netrin1 acts from the intermediate spinal cord to regionally confine Bmp signaling to the dorsal spinal cord. Thus, netrin1 has reiterative activities shaping dorsal spinal circuits, first by regulating cell fate decisions and then acting as a guidance cue to direct axon extension.
Collapse
Affiliation(s)
- Sandy Alvarez
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sandeep Gupta
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yesica Mercado-Ayon
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kaitlyn Honeychurch
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Cristian Rodriguez
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; CIRM Bridges to Research Program, California State University, Northridge, CA 91330, USA
| | - Riki Kawaguchi
- Department of Psychiatry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Samantha J Butler
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual & Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
2
|
Frazer NB, Kaas GA, Firmin CG, Gamazon ER, Hatzopoulos AK. BMP Antagonist Gremlin 2 Regulates Hippocampal Neurogenesis and Is Associated with Seizure Susceptibility and Anxiety. eNeuro 2024; 11:ENEURO.0213-23.2024. [PMID: 39349059 PMCID: PMC11493175 DOI: 10.1523/eneuro.0213-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 10/02/2024] Open
Abstract
The Bone Morphogenetic Protein (BMP) signaling pathway is vital in neural progenitor cell proliferation, specification, and differentiation. The BMP signaling antagonist Gremlin 2 (Grem2) is the most potent natural inhibitor of BMP expressed in the adult brain; however its function remains unknown. To address this knowledge gap, we have analyzed mice lacking Grem2 via homologous recombination (Grem2-/- ). Histological analysis of brain sections revealed significant scattering of CA3 pyramidal cells within the dentate hilus in the hippocampus of Grem2-/- mice. Furthermore, the number of proliferating neural stem cells and neuroblasts was significantly decreased in the subgranular zone of Grem2-/- mice compared with that of wild-type (WT) controls. Due to the role of hippocampal neurogenesis in neurological disorders, we tested mice on a battery of neurobehavioral tests. Grem2-/- mice exhibited increased anxiety on the elevated zero maze in response to acute and chronic stress. Specifically, male Grem2-/- mice showed increased anxiogenesis following chronic stress, and this was correlated with higher levels of BMP signaling and decreased proliferation in the dentate gyrus. Additionally, when chemically challenged with kainic acid, Grem2-/- mice displayed a higher susceptibility to and increased severity of seizures compared with WTs. Together, our data indicate that Grem2 regulates BMP signaling and is vital in maintaining homeostasis in adult hippocampal neurogenesis and structure. Furthermore, the lack of Grem2 contributes to the development and progression of neurogenesis-related disorders such as anxiety and epilepsy.
Collapse
Affiliation(s)
- Nicolette B Frazer
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232
| | - Garrett A Kaas
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Caroline G Firmin
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Eric R Gamazon
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Antonis K Hatzopoulos
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| |
Collapse
|
3
|
Mörsdorf D, Knabl P, Genikhovich G. Highly conserved and extremely evolvable: BMP signalling in secondary axis patterning of Cnidaria and Bilateria. Dev Genes Evol 2024; 234:1-19. [PMID: 38472535 PMCID: PMC11226491 DOI: 10.1007/s00427-024-00714-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/06/2024] [Indexed: 03/14/2024]
Abstract
Bilateria encompass the vast majority of the animal phyla. As the name states, they are bilaterally symmetric, that is with a morphologically clear main body axis connecting their anterior and posterior ends, a second axis running between their dorsal and ventral surfaces, and with a left side being roughly a mirror image of their right side. Bone morphogenetic protein (BMP) signalling has widely conserved functions in the formation and patterning of the second, dorso-ventral (DV) body axis, albeit to different extents in different bilaterian species. Whilst initial findings in the fruit fly Drosophila and the frog Xenopus highlighted similarities amongst these evolutionarily very distant species, more recent analyses featuring other models revealed considerable diversity in the mechanisms underlying dorsoventral patterning. In fact, as phylogenetic sampling becomes broader, we find that this axis patterning system is so evolvable that even its core components can be deployed differently or lost in different model organisms. In this review, we will try to highlight the diversity of ways by which BMP signalling controls bilaterality in different animals, some of which do not belong to Bilateria. Future research combining functional analyses and modelling is bound to give us some understanding as to where the limits to the extent of the evolvability of BMP-dependent axial patterning may lie.
Collapse
Affiliation(s)
- David Mörsdorf
- Dept. Neurosciences and Developmental Biology, University of Vienna, UBB, Djerassiplatz 1, 1030, Vienna, Austria
| | - Paul Knabl
- Dept. Neurosciences and Developmental Biology, University of Vienna, UBB, Djerassiplatz 1, 1030, Vienna, Austria
- Vienna Doctoral School of Ecology and Evolution (VDSEE), University of Vienna, Vienna, Austria
| | - Grigory Genikhovich
- Dept. Neurosciences and Developmental Biology, University of Vienna, UBB, Djerassiplatz 1, 1030, Vienna, Austria.
| |
Collapse
|
4
|
Asashima M, Satou-Kobayashi Y. Spemann-Mangold organizer and mesoderm induction. Cells Dev 2024; 178:203903. [PMID: 38295873 DOI: 10.1016/j.cdev.2024.203903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/09/2024]
Abstract
The discovery of the Spemann-Mangold organizer strongly influenced subsequent research on embryonic induction, with research aiming to elucidate the molecular characteristics of organizer activity being currently underway. Herein, we review the history of research on embryonic induction, and describe how the mechanisms of induction phenomena and developmental processes have been investigated. Classical experiments investigating the differentiation capacity and inductive activity of various embryonic regions were conducted by many researchers, and important theories of region-specific induction and the concept for chain of induction were proposed. The transition from experimental embryology to developmental biology has enabled us to understand the mechanisms of embryonic induction at the molecular level. Consequently, many inducing substances and molecules such as transcriptional factors and peptide growth factors involved in the organizer formation were identified. One of peptide growth factors, activin, acts as a mesoderm- and endoderm-inducing substance. Activin induces several tissues and organs from the undifferentiated cell mass of amphibian embryos in a concentration-dependent manner. We review the extent to which we can control in vitro organogenesis from undifferentiated cells, and discuss the application to stem cell-based regenerative medicine based on insights gained from animal experiments, such as in amphibians.
Collapse
Affiliation(s)
- Makoto Asashima
- Advanced Comprehensive Research Organization, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-0003, Japan.
| | - Yumeko Satou-Kobayashi
- Advanced Comprehensive Research Organization, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-0003, Japan
| |
Collapse
|
5
|
Castro Colabianchi AM, González Pérez NG, Franchini LF, López SL. A maternal dorsoventral prepattern revealed by an asymmetric distribution of ventralizing molecules before fertilization in Xenopus laevis. Front Cell Dev Biol 2024; 12:1365705. [PMID: 38572484 PMCID: PMC10987785 DOI: 10.3389/fcell.2024.1365705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/07/2024] [Indexed: 04/05/2024] Open
Abstract
The establishment of the embryonic dorsoventral axis in Xenopus occurs when the radial symmetry around the egg's animal-vegetal axis is broken to give rise to the typical symmetry of Bilaterians. We have previously shown that the Notch1 protein is ventrally enriched during early embryogenesis in Xenopus laevis and zebrafish and exerts ventralizing activity through β-Catenin destabilization and the positive regulation of ventral center genes in X. laevis. These findings led us to further investigate when these asymmetries arise. In this work, we show that the asymmetrical distribution of Notch1 protein and mRNA precedes cortical rotation and even fertilization in X. laevis. Moreover, we found that in unfertilized eggs transcripts encoded by the ventralizing gene bmp4 are also asymmetrically distributed in the animal hemisphere and notch1 transcripts accumulate consistently on the same side of the eccentric maturation point. Strikingly, a Notch1 asymmetry orthogonal to the animal-vegetal axis appears during X. laevis oogenesis. Thus, we show for the first time a maternal bias in the distribution of molecules that are later involved in ventral patterning during embryonic axialization, strongly supporting the hypothesis of a dorsoventral prepattern or intrinsic bilaterality of Xenopus eggs before fertilization.
Collapse
Affiliation(s)
- Aitana M. Castro Colabianchi
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular e Histología / 1° U.A. Departamento de Histología, Embriología, Biología Celular y Genética, Laboratorio de Embriología Molecular “Prof. Dr. Andrés E. Carrasco”, Buenos Aires, Argentina
- CONICET–Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencias “Prof. E. De Robertis” (IBCN), Buenos Aires, Argentina
| | - Nicolás G. González Pérez
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular e Histología / 1° U.A. Departamento de Histología, Embriología, Biología Celular y Genética, Laboratorio de Embriología Molecular “Prof. Dr. Andrés E. Carrasco”, Buenos Aires, Argentina
- CONICET–Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencias “Prof. E. De Robertis” (IBCN), Buenos Aires, Argentina
| | - Lucía F. Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI) “Dr. Héctor N. Torres”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Silvia L. López
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Biología Celular e Histología / 1° U.A. Departamento de Histología, Embriología, Biología Celular y Genética, Laboratorio de Embriología Molecular “Prof. Dr. Andrés E. Carrasco”, Buenos Aires, Argentina
- CONICET–Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencias “Prof. E. De Robertis” (IBCN), Buenos Aires, Argentina
| |
Collapse
|
6
|
Abstract
In avian and mammalian embryos the "organizer" property associated with neural induction of competent ectoderm into a neural plate and its subsequent patterning into rostro-caudal domains resides at the tip of the primitive streak before neurulation begins, and before a morphological Hensen's node is discernible. The same region and its later derivatives (like the notochord) also have the ability to "dorsalize" the adjacent mesoderm, for example by converting lateral plate mesoderm into paraxial (pre-somitic) mesoderm. Both neural induction and dorsalization of the mesoderm involve inhibition of BMP, and the former also requires other signals. This review surveys the key experiments done to elucidate the functions of the organizer and the mechanisms of neural induction in amniotes. We conclude that the mechanisms of neural induction in amniotes and anamniotes are likely to be largely the same; apparent differences are likely to be due to differences in experimental approaches dictated by embryo topology and other practical constraints. We also discuss the relationships between "neural induction" assessed by grafts of the organizer and normal neural plate development, as well as how neural induction relates to the generation of neuronal cells from embryonic and other stem cells in vitro.
Collapse
Affiliation(s)
- Claudio D Stern
- Department of Cell and Developmental Biology, University College London, London, United Kingdom.
| |
Collapse
|
7
|
Wang C, Liu Z, Zeng Y, Zhou L, Long Q, Hassan IU, Zhang Y, Qi X, Cai D, Mao B, Lu G, Sun J, Yao Y, Deng Y, Zhao Q, Feng B, Zhou Q, Chan WY, Zhao H. ZSWIM4 regulates embryonic patterning and BMP signaling by promoting nuclear Smad1 degradation. EMBO Rep 2024; 25:646-671. [PMID: 38177922 PMCID: PMC10897318 DOI: 10.1038/s44319-023-00046-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/06/2024] Open
Abstract
The dorsoventral gradient of BMP signaling plays an essential role in embryonic patterning. Zinc Finger SWIM-Type Containing 4 (zswim4) is expressed in the Spemann-Mangold organizer at the onset of Xenopus gastrulation and is then enriched in the developing neuroectoderm at the mid-gastrula stages. Knockdown or knockout of zswim4 causes ventralization. Overexpression of zswim4 decreases, whereas knockdown of zswim4 increases the expression levels of ventrolateral mesoderm marker genes. Mechanistically, ZSWIM4 attenuates the BMP signal by reducing the protein stability of SMAD1 in the nucleus. Stable isotope labeling by amino acids in cell culture (SILAC) identifies Elongin B (ELOB) and Elongin C (ELOC) as the interaction partners of ZSWIM4. Accordingly, ZSWIM4 forms a complex with the Cul2-RING ubiquitin ligase and ELOB and ELOC, promoting the ubiquitination and degradation of SMAD1 in the nucleus. Our study identifies a novel mechanism that restricts BMP signaling in the nucleus.
Collapse
Affiliation(s)
- Chengdong Wang
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ziran Liu
- Qingdao Municipal Center for Disease Control and Prevention, 266033, Qingdao, Shandong, China
| | - Yelin Zeng
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Liangji Zhou
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qi Long
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Imtiaz Ul Hassan
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuanliang Zhang
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xufeng Qi
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Dongqing Cai
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology, Jinan University, 510632, Guangzhou, Guangdong, China
| | - Bingyu Mao
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, Yunnan, China
- Kunming Institute of Zoology - The Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Chinese Academy of Sciences, Kunming, China
| | - Gang Lu
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jianmin Sun
- Department of Pathogen Biology and Immunology, School of Basic Medical Sciences, Ningxia Medical University, No. 1160 Shengli Street, 750004, Yinchuan, China
| | - Yonggang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, 650223, Kunming, Yunnan, China
- Kunming Institute of Zoology - The Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Chinese Academy of Sciences, Kunming, China
| | - Yi Deng
- Department of Biology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, and Shenzhen Key Laboratory of Cell Microenvironment, Southern University of Science and Technology, 518055, Shenzhen, China
| | - Qian Zhao
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Bo Feng
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qin Zhou
- School of Basic Medical Sciences, Harbin Medical University, 150081, Harbin, China
| | - Wai Yee Chan
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Kunming Institute of Zoology - The Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Branch of CAS Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hui Zhao
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Kunming Institute of Zoology - The Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, The Chinese University of Hong Kong, Hong Kong SAR, China.
- Hong Kong Branch of CAS Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
8
|
Mounika N, Yadav A, Kamboj P, Banerjee SK, Deka UJ, Kaur S, Adela R. Circulatory bone morphogenetic protein (BMP) 8B is a non-invasive predictive biomarker for the diagnosis of non-alcoholic steatohepatitis (NASH). PLoS One 2023; 18:e0295839. [PMID: 38127951 PMCID: PMC10734958 DOI: 10.1371/journal.pone.0295839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a complex disease which is characterized by the deposition of fats in the hepatocytes. Further, it progresses to nonalcoholic steatohepatitis (NASH), fibrosis, and hepatocellular carcinoma. The increasing prevalence of NAFLD urges to find the non-invasive predictive biomarkers. In this study, we sought to determine increased BMP8B levels as predictors for the progression of NAFLD. METHODS In the present cross-sectional study, circulatory BMP8B levels were measured in healthy controls (n = 56), NAFL patients (n = 72) and NASH patients (n = 77) by using an ELISA kit. Human hepatic BMP8B mRNA expression was measured in the liver tissue of control and NASH patients. In addition, BMP8B expression was confirmed by immunohistochemistry analysis. Furthermore, hepatic BMP8B mRNA expression was measured in wild type (WT) mice, WT mice fed with choline deficient high fat diet (WT+CDHF), iNOS (inducible nitric oxide synthase) knockout (iNOS-/-) mice, iNOS-/- fed with CDHF diet (iNOS-/-+CDHF). RESULTS Increased circulatory BMP8B levels and BMP8B mRNA expression in hepatic tissue were significantly higher in NASH patients as compared with the control subjects. BMP8B expression was increased parallel to the fibrosis score in the hepatic tissues of NASH patients. It was observed that increased BMP8B levels have shown a significant positive correlation between aspartate aminotransferase (r = 0.31, p = 0.005), alanine aminotransferase (r = 0.23, p = 0.045), APRI (r = 0.30, p = 0.009), and Fib-4 score (r = 0.25, p = 0.036) in NASH patients. BMP8B has maintained a significant association with NASH and shown high sensitivity (92.91%) and specificity (92.73%) in NASH patients. Furthermore, increased BMP8B mRNA expression levels were observed in iNOS-/-+CDHF mice. CONCLUSION Our study findings confirmed that BMP8B increases with the severity of the disease and BMP8B shows potential as a non-invasive predictive biomarker to identify NAFLD progression. However, future studies should investigate circulatory BMP8B levels in a large number of patients and also its impact on liver during NAFLD progression.
Collapse
Affiliation(s)
- Nadella Mounika
- Department of Pharmacy Practice, NIPER-Guwahati, Sila Katamur, Halugurisuk, Changsari, Dist.Kamrup, Guwahati, Assam, India
| | - Angeera Yadav
- Department of Pharmacy Practice, NIPER-Guwahati, Sila Katamur, Halugurisuk, Changsari, Dist.Kamrup, Guwahati, Assam, India
| | - Parul Kamboj
- Senior Researcher, Translational Health Science and Technology Institute (THSTI), Faridabad, India
| | - Sanjay K. Banerjee
- Senior Researcher, Translational Health Science and Technology Institute (THSTI), Faridabad, India
- Department of Biotechnology, NIPER-Guwahati, Sila Katamur, Halugurisuk, Changsari, Dist.Kamrup, Guwahati, Assam, India
| | - Utpal Jyoti Deka
- Department of Gastroenterology, Downtown Hospital, GS Rd, Bormotoria, Guwahati, Assam, India
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver & Biliary Science (ILBS), Vasant Kunj, New Delhi, India
| | - Ramu Adela
- Department of Pharmacy Practice, NIPER-Guwahati, Sila Katamur, Halugurisuk, Changsari, Dist.Kamrup, Guwahati, Assam, India
| |
Collapse
|
9
|
Alvarez S, Gupta S, Honeychurch K, Mercado-Ayon Y, Kawaguchi R, Butler SJ. Netrin1 patterns the dorsal spinal cord through modulation of Bmp signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.02.565384. [PMID: 37961605 PMCID: PMC10635094 DOI: 10.1101/2023.11.02.565384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
We have identified an unexpected role for netrin1 as a suppressor of bone morphogenetic protein (Bmp) signaling in the developing dorsal spinal cord. Using a combination of gain- and loss-of-function approaches in chicken, embryonic stem cell (ESC), and mouse models, we have observed that manipulating the level of netrin1 specifically alters the patterning of the Bmp-dependent dorsal interneurons (dIs), dI1-dI3. Altered netrin1 levels also change Bmp signaling activity, as measured by bioinformatics, and monitoring phosophoSmad1/5/8 activation, the canonical intermediate of Bmp signaling, and Id levels, a known Bmp target. Together, these studies support the hypothesis that netrin1 acts from the intermediate spinal cord to regionally confine Bmp signaling to the dorsal spinal cord. Thus, netrin1 has reiterative activities shaping dorsal spinal circuits, first by regulating cell fate decisions and then acting as a guidance cue to direct axon extension.
Collapse
|
10
|
Sun L, Ping L, Gao R, Zhang B, Chen X. lmo4a Contributes to Zebrafish Inner Ear and Vestibular Development via Regulation of the Bmp Pathway. Genes (Basel) 2023; 14:1371. [PMID: 37510276 PMCID: PMC10378989 DOI: 10.3390/genes14071371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/13/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND In vertebrates, the development of the inner ear is a delicate process, whereas its relating molecular pathways are still poorly understood. LMO4, an LIM domain-only transcriptional regulator, is drawing an increasing amount of interest for its multiple roles regarding human embryonic development and the modulation of ototoxic side effects of cisplatin including cochlear apoptosis and hearing loss. The aim of the present study is to further explore the role of lmo4a in zebrafish inner ear development and thus explore its functional role. METHODS The Spatial Transcript Omics DataBase was referred to in order to evaluate the expression of lmo4a during the first 24 h of zebrafish development. In situ hybridization was applied to validate and extend the expression profile of lmo4a to 3 days post-fertilization. The morpholino (MO) knockdown and CRISPR/Cas9 knockout (KO) of lmo4a was applied. Morphological analyses of otic vesical, hair cells, statoacoustic ganglion and semicircular canals were conducted. The swimming pattern of lmo4a KO and MO zebrafish was tracked. In situ hybridization was further applied to verify the expression of genes of the related pathways. Rescue of the phenotype was attempted by blockage of the bmp pathway via heat shock and injection of Dorsomorphin. RESULTS lmo4a is constitutively expressed in the otic placode and otic vesicle during the early stages of zebrafish development. Knockdown and knockout of lmo4a both induced smaller otocysts, less hair cells, immature statoacoustic ganglion and malformed semicircular canals. Abnormal swimming patterns could be observed in both lmo4a MO and KO zebrafish. eya1 in preplacodal ectoderm patterning was downregulated. bmp2 and bmp4 expressions were found to be upregulated and extended in lmo4a morphants, and blockage of the Bmp pathway partially rescued the vestibular defects. CONCLUSIONS We concluded that lmo4a holds a regulative effect on the Bmp pathway and is required for the normal development of zebrafish inner ear. Our study pointed out the conservatism of LMO4 in inner ear development between mammals and zebrafish as well as shed more light on the molecular mechanisms behind it. Further research is needed to distinguish the relationships between lmo4 and the Bmp pathway, which may lead to diagnostic and therapeutic approaches towards human inner ear malformation.
Collapse
Affiliation(s)
- Le Sun
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing 100730, China;
| | - Lu Ping
- Chinese Academy of Medical Sciences and Peking Union Medical College, #9 Dongdan Santiao, Dongcheng District, Beijing 100050, China;
| | - Ruzhen Gao
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Bo Zhang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing 100871, China;
| | - Xiaowei Chen
- Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, #1 Shuaifuyuan, Dongcheng District, Beijing 100730, China;
| |
Collapse
|
11
|
Ahmed T, Ramonett A, Kwak EA, Kumar S, Flores PC, Ortiz HR, Langlais PR, Hund TJ, Mythreye K, Lee NY. Endothelial tip/stalk cell selection requires BMP9-induced β IV-spectrin expression during sprouting angiogenesis. Mol Biol Cell 2023; 34:ar72. [PMID: 37126382 PMCID: PMC10295478 DOI: 10.1091/mbc.e23-02-0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 05/02/2023] Open
Abstract
βIV-Spectrin is a membrane cytoskeletal protein with specialized roles in the nervous system and heart. Recent evidence also indicates a fundamental role for βIV-spectrin in angiogenesis as its endothelial-specific gene deletion in mice enhances embryonic lethality due to hypervascularization and hemorrhagic defects. During early vascular sprouting, βIV-spectrin is believed to inhibit tip cell sprouting in favor of the stalk cell phenotype by mediating VEGFR2 internalization and degradation. Despite these essential roles, mechanisms governing βIV-spectrin expression remain unknown. Here we identify bone morphogenetic protein 9 (BMP9) as a major inducer of βIV-spectrin gene expression in the vascular system. We show that BMP9 signals through the ALK1/Smad1 pathway to induce βIV-spectrin expression, which then recruits CaMKII to the cell membrane to induce phosphorylation-dependent VEGFR2 turnover. Although BMP9 signaling promotes stalk cell behavior through activation of hallmark stalk cell genes ID-1/3 and Hes-1 and Notch signaling cross-talk, we find that βIV-spectrin acts upstream of these pathways as loss of βIV-spectrin in neonate mice leads to retinal hypervascularization due to excessive VEGFR2 levels, increased tip cell populations, and strong Notch inhibition irrespective of BMP9 treatment. These findings demonstrate βIV-spectrin as a BMP9 gene target critical for tip/stalk cell selection during nascent vessel sprouting.
Collapse
Affiliation(s)
- Tasmia Ahmed
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, AZ 85724
| | - Aaron Ramonett
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724
| | - Eun-A Kwak
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724
| | - Sanjay Kumar
- Division of Biology, Indian Institute of Science Education and Research, Tirupati 517507, India
| | - Paola Cruz Flores
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, AZ 85724
| | - Hannah R. Ortiz
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724
| | | | - Thomas J. Hund
- Department of Biomedical Engineering, Ohio State University, Columbus, OH 43210
| | - Karthikeyan Mythreye
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Nam Y. Lee
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, AZ 85724
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724
- Comprehensive Cancer Center, University of Arizona, Tucson, AZ 85724
| |
Collapse
|
12
|
Johnson K, Freedman S, Braun R, LaBonne C. Quantitative analysis of transcriptome dynamics provides novel insights into developmental state transitions. BMC Genomics 2022; 23:723. [PMID: 36273135 PMCID: PMC9588240 DOI: 10.1186/s12864-022-08953-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/19/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND During embryogenesis, the developmental potential of initially pluripotent cells becomes progressively restricted as they transit to lineage restricted states. The pluripotent cells of Xenopus blastula-stage embryos are an ideal system in which to study cell state transitions during developmental decision-making, as gene expression dynamics can be followed at high temporal resolution. RESULTS Here we use transcriptomics to interrogate the process by which pluripotent cells transit to four different lineage-restricted states: neural progenitors, epidermis, endoderm and ventral mesoderm, providing quantitative insights into the dynamics of Waddington's landscape. Our findings provide novel insights into why the neural progenitor state is the default lineage state for pluripotent cells and uncover novel components of lineage-specific gene regulation. These data reveal an unexpected overlap in the transcriptional responses to BMP4/7 and Activin signaling and provide mechanistic insight into how the timing of signaling inputs such as BMP are temporally controlled to ensure correct lineage decisions. CONCLUSIONS Together these analyses provide quantitative insights into the logic and dynamics of developmental decision making in early embryos. They also provide valuable lineage-specific time series data following the acquisition of specific lineage states during development.
Collapse
Affiliation(s)
- Kristin Johnson
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL, 60208, USA
| | - Simon Freedman
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL, 60208, USA
- Department of Engineering Sciences and Applied Math, Northwestern University, Evanston, IL, USA
| | - Rosemary Braun
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL, 60208, USA
- Department of Engineering Sciences and Applied Math, Northwestern University, Evanston, IL, USA
- Northwestern Institute On Complex Systems, Northwestern University, Evanston, IL, USA
| | - Carole LaBonne
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA.
- NSF-Simons Center for Quantitative Biology, Northwestern University, Evanston, IL, 60208, USA.
| |
Collapse
|
13
|
Michiue T, Tsukano K. Feedback Regulation of Signaling Pathways for Precise Pre-Placodal Ectoderm Formation in Vertebrate Embryos. J Dev Biol 2022; 10:35. [PMID: 36135368 PMCID: PMC9504399 DOI: 10.3390/jdb10030035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/19/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Intracellular signaling pathways are essential to establish embryonic patterning, including embryonic axis formation. Ectodermal patterning is also governed by a series of morphogens. Four ectodermal regions are thought to be controlled by morphogen gradients, but some perturbations are expected to occur during dynamic morphogenetic movement. Therefore, a mechanism to define areas precisely and reproducibly in embryos, including feedback regulation of signaling pathways, is necessary. In this review, we outline ectoderm pattern formation and signaling pathways involved in the establishment of the pre-placodal ectoderm (PPE). We also provide an example of feedback regulation of signaling pathways for robust formation of the PPE, showing the importance of this regulation.
Collapse
Affiliation(s)
- Tatsuo Michiue
- Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | | |
Collapse
|
14
|
Tsukano K, Yamamoto T, Watanabe T, Michiue T. Xenopus Dusp6 modulates FGF signaling precisely to pattern pre-placodal ectoderm. Dev Biol 2022; 488:81-90. [DOI: 10.1016/j.ydbio.2022.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 05/07/2022] [Accepted: 05/16/2022] [Indexed: 12/23/2022]
|
15
|
Ventx Family and Its Functional Similarities with Nanog: Involvement in Embryonic Development and Cancer Progression. Int J Mol Sci 2022; 23:ijms23052741. [PMID: 35269883 PMCID: PMC8911082 DOI: 10.3390/ijms23052741] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/21/2022] [Accepted: 02/27/2022] [Indexed: 12/27/2022] Open
Abstract
The Ventx family is one of the subfamilies of the ANTP (antennapedia) superfamily and belongs to the NK-like (NKL) subclass. Ventx is a homeobox transcription factor and has a DNA-interacting domain that is evolutionarily conserved throughout vertebrates. It has been extensively studied in Xenopus, zebrafish, and humans. The Ventx family contains transcriptional repressors widely involved in embryonic development and tumorigenesis in vertebrates. Several studies have documented that the Ventx family inhibited dorsal mesodermal formation, neural induction, and head formation in Xenopus and zebrafish. Moreover, Ventx2.2 showed functional similarities to Nanog and Barx1, leading to pluripotency and neural-crest migration in vertebrates. Among them, Ventx protein is an orthologue of the Ventx family in humans. Studies have demonstrated that human Ventx was strongly associated with myeloid-cell differentiation and acute myeloid leukemia. The therapeutic potential of Ventx family inhibition in combating cancer progression in humans is discussed. Additionally, we briefly discuss genome evolution, gene duplication, pseudo-allotetraploidy, and the homeobox family in Xenopus.
Collapse
|
16
|
Yoon J, Kumar V, Goutam RS, Kim SC, Park S, Lee U, Kim J. Bmp Signal Gradient Modulates Convergent Cell Movement via Xarhgef3.2 during Gastrulation of Xenopus Embryos. Cells 2021; 11:44. [PMID: 35011606 PMCID: PMC8750265 DOI: 10.3390/cells11010044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 01/31/2023] Open
Abstract
Gastrulation is a critical step in the establishment of a basic body plan during development. Convergence and extension (CE) cell movements organize germ layers during gastrulation. Noncanonical Wnt signaling has been known as major signaling that regulates CE cell movement by activating Rho and Rac. In addition, Bmp molecules are expressed in the ventral side of a developing embryo, and the ventral mesoderm region undergoes minimal CE cell movement while the dorsal mesoderm undergoes dynamic cell movements. This suggests that Bmp signal gradient may affect CE cell movement. To investigate whether Bmp signaling negatively regulates CE cell movements, we performed microarray-based screening and found that the transcription of Xenopus Arhgef3.2 (Rho guanine nucleotide exchange factor) was negatively regulated by Bmp signaling. We also showed that overexpression or knockdown of Xarhgef3.2 caused gastrulation defects. Interestingly, Xarhgef3.2 controlled gastrulation cell movements through interacting with Disheveled (Dsh2) and Dsh2-associated activator of morphogenesis 1 (Daam1). Our results suggest that Bmp gradient affects gastrulation cell movement (CE) via negative regulation of Xarhgef3.2 expression.
Collapse
Affiliation(s)
- Jaeho Yoon
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea; (J.Y.); (V.K.); (R.S.G.); (S.-C.K.)
- National Cancer Institute, Frederick, MD 21702, USA
| | - Vijay Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea; (J.Y.); (V.K.); (R.S.G.); (S.-C.K.)
| | - Ravi Shankar Goutam
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea; (J.Y.); (V.K.); (R.S.G.); (S.-C.K.)
| | - Sung-Chan Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea; (J.Y.); (V.K.); (R.S.G.); (S.-C.K.)
| | - Soochul Park
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea;
| | - Unjoo Lee
- Department of Electrical Engineering, Hallym University, Chuncheon 24252, Korea;
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea; (J.Y.); (V.K.); (R.S.G.); (S.-C.K.)
| |
Collapse
|
17
|
Kumar V, Goutam RS, Umair Z, Park S, Lee U, Kim J. Foxd4l1.1 Negatively Regulates Chordin Transcription in Neuroectoderm of Xenopus Gastrula. Cells 2021; 10:cells10102779. [PMID: 34685759 PMCID: PMC8534798 DOI: 10.3390/cells10102779] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 02/06/2023] Open
Abstract
Inhibition of the bone morphogenetic proteins (BMPs) is the primary step toward neuroectoderm formation in vertebrates. In this process, the Spemann organizer of the dorsal mesoderm plays a decisive role by secreting several extracellular BMP inhibitors such as Chordin (Chrd). Chrd physically interacts with BMP proteins and inhibits BMP signaling, which triggers the expression of neural-specific transcription factors (TFs), including Foxd4l1.1. Thus, Chrd induces in a BMP-inhibited manner and promotes neuroectoderm formation. However, the regulatory feedback mechanism of Foxd4l1.1 on mesodermal genes expression during germ-layer specification has not been fully elucidated. In this study, we investigated the regulatory mechanism of Foxd4l1.1 on chrd (a mesodermal gene). We demonstrate that Foxd4l1.1 inhibits chrd expression during neuroectoderm formation in two ways: First, Foxd4l1.1 directly binds to FRE (Foxd4l1.1 response elements) within the chrd promoter region to inhibit transcription. Second, Foxd4l1.1 physically interacts with Smad2 and Smad3, and this interaction blocks Smad2 and Smad3 binding to activin response elements (AREs) within the chrd promoter. Site-directed mutagenesis of FRE within the chrd(-2250) promoter completely abolished repressor activity of the Foxd4l1.1. RT-PCR and reporter gene assay results indicate that Foxd4l1.1 strongly inhibits mesoderm- and ectoderm-specific marker genes to maintain neural fate. Altogether, these results suggest that Foxd4l1.1 negatively regulates chrd transcription by dual mechanism. Thus, our study demonstrates the existence of precise reciprocal regulation of chrd transcription during neuroectoderm and mesoderm germ-layer specification in Xenopus embryos.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea; (V.K.); (R.S.G.); (Z.U.)
| | - Ravi Shankar Goutam
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea; (V.K.); (R.S.G.); (Z.U.)
| | - Zobia Umair
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea; (V.K.); (R.S.G.); (Z.U.)
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 21999, Korea
| | - Soochul Park
- Department of Biological Sciences, Sookmyung Women’s University, Seoul 04310, Korea;
| | - Unjoo Lee
- Department of Electrical Engineering, Hallym University, Chuncheon 24252, Korea
- Correspondence: (U.L.); (J.K.)
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Chuncheon 24252, Korea; (V.K.); (R.S.G.); (Z.U.)
- Correspondence: (U.L.); (J.K.)
| |
Collapse
|
18
|
Shu DY, Lovicu FJ. Insights into Bone Morphogenetic Protein-(BMP-) Signaling in Ocular Lens Biology and Pathology. Cells 2021; 10:cells10102604. [PMID: 34685584 PMCID: PMC8533954 DOI: 10.3390/cells10102604] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 01/23/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) are a diverse class of growth factors that belong to the transforming growth factor-beta (TGFβ) superfamily. Although originally discovered to possess osteogenic properties, BMPs have since been identified as critical regulators of many biological processes, including cell-fate determination, cell proliferation, differentiation and morphogenesis, throughout the body. In the ocular lens, BMPs are important in orchestrating fundamental developmental processes such as induction of lens morphogenesis, and specialized differentiation of its fiber cells. Moreover, BMPs have been reported to facilitate regeneration of the lens, as well as abrogate pathological processes such as TGFβ-induced epithelial-mesenchymal transition (EMT) and apoptosis. In this review, we summarize recent insights in this topic and discuss the complexities of BMP-signaling including the role of individual BMP ligands, receptors, extracellular antagonists and cross-talk between canonical and non-canonical BMP-signaling cascades in the lens. By understanding the molecular mechanisms underlying BMP activity, we can advance their potential therapeutic role in cataract prevention and lens regeneration.
Collapse
Affiliation(s)
- Daisy Y. Shu
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, USA;
| | - Frank J. Lovicu
- School of Medical Sciences, The University of Sydney, Sydney, NSW 2006, Australia
- Save Sight Institute, The University of Sydney, Sydney, NSW 2000, Australia
- Correspondence: ; Tel.: +61-2-9351-5170
| |
Collapse
|
19
|
Porras-Gómez TJ, Villagrán-SantaCruz M, Moreno-Mendoza N. Biology of primordial germ cells in vertebrates with emphasis in urodeles amphibians. Mol Reprod Dev 2021; 88:773-792. [PMID: 34532913 DOI: 10.1002/mrd.23533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 11/08/2022]
Abstract
Primordial germ cells (PGCs) are highly specialized cells that play a relevant role in the maintenance and evolution of the species, since they create new combinations of genetic information between the organisms. Amphibians are a class of amniote vertebrates that are divided into three subclasses, the anurans (frogs and toads), the urodeles (salamanders and newts), and the gymnophiones (caecilians). The study of PGCs in amphibians has been addressed in more detail in anurans while little is known about the biology of this cell lineage in urodeles. Studies in some urodeles species have suggested that PGCs are of mesodermal origin, specifying in the lateral plate mesoderm at the late gastrula stage. With classical experiments it shown that, there is an induction of mesoderm, therefore most likely urodeles PGCs develop from unspecialized mesodermal tissue that responds to extracellular signals. However, some fundamental biological processes of PGCs such as the analysis of their specification, arrival, and colonization to the gonads, and their maintenance and differentiation into mature and fertile gametes remain to be elucidated. Therefore, knowledge about the biology of PGCs is of great importance to ensure the perpetuation of urodeles amphibians, as some species are in danger of becoming extinct.
Collapse
Affiliation(s)
- Tania J Porras-Gómez
- Laboratorio de Biología Tisular y Reproductora, Departamento de Biología Comparada, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Maricela Villagrán-SantaCruz
- Laboratorio de Biología Tisular y Reproductora, Departamento de Biología Comparada, Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Norma Moreno-Mendoza
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
20
|
Ponzetti M, Rucci N. Osteoblast Differentiation and Signaling: Established Concepts and Emerging Topics. Int J Mol Sci 2021; 22:ijms22136651. [PMID: 34206294 PMCID: PMC8268587 DOI: 10.3390/ijms22136651] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoblasts, the cells that build up our skeleton, are remarkably versatile and important cells that need tight regulation in all the phases of their differentiation to guarantee proper skeletal development and homeostasis. Although we know many of the key pathways involved in osteoblast differentiation and signaling, it is becoming clearer and clearer that this is just the tip of the iceberg, and we are constantly discovering novel concepts in osteoblast physiology. In this review, we discuss well-established pathways of osteoblastic differentiation, i.e., the classical ones committing mesenchymal stromal cells to osteoblast, and then osteocytes as well as recently emerged players. In particular, we discuss micro (mi)RNAs, long non-coding (lnc)RNAs, circular (circ)RNAs, and extracellular vesicles, focusing on the mechanisms through which osteoblasts are regulated by these factors, and conversely, how they use extracellular vesicles to communicate with the surrounding microenvironment.
Collapse
|
21
|
Characterization of the different oligomeric states of the DAN family antagonists SOSTDC1 and SOST. Biochem J 2021; 477:3167-3182. [PMID: 32779697 PMCID: PMC7473711 DOI: 10.1042/bcj20200552] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022]
Abstract
The DAN (differential screening-selected gene aberrative in neuroblastoma) family are a group of secreted extracellular proteins which typically bind to and antagonize BMP (bone morphogenetic protein) ligands. Previous studies have revealed discrepancies between the oligomerization state of certain DAN family members, with SOST (a poor antagonist of BMP signaling) forming a monomer while Grem1, Grem2, and NBL1 (more potent BMP antagonists) form non-disulfide linked dimers. The protein SOSTDC1 (Sclerostin domain containing protein 1) is sequentially similar to SOST, but has been shown to be a better BMP inhibitor. In order to determine the oligomerization state of SOSTDC1 and determine what effect dimerization might have on the mechanism of DAN family antagonism of BMP signaling, we isolated the SOSTDC1 protein and, using a battery of biophysical, biochemical, and structural techniques, showed that SOSTDC1 forms a highly stable non-covalent dimer. Additionally, this SOSTDC1 dimer was shown, using an in vitro cell based assay system, to be an inhibitor of multiple BMP signaling growth factors, including GDF5, while monomeric SOST was a very poor antagonist. These results demonstrate that SOSTDC1 is distinct from paralogue SOST in terms of both oligomerization and strength of BMP inhibition.
Collapse
|
22
|
Kumar S, Umair Z, Kumar V, Kumar S, Lee U, Kim J. Foxd4l1.1 negatively regulates transcription of neural repressor ventx1.1 during neuroectoderm formation in Xenopus embryos. Sci Rep 2020; 10:16780. [PMID: 33033315 PMCID: PMC7545198 DOI: 10.1038/s41598-020-73662-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 09/16/2020] [Indexed: 11/09/2022] Open
Abstract
Neuroectoderm formation is the first step in development of a proper nervous system for vertebrates. The developmental decision to form a non-neural ectoderm versus a neural one involves the regulation of BMP signaling, first reported many decades ago. However, the precise regulatory mechanism by which this is accomplished has not been fully elucidated, particularly for transcriptional regulation of certain key transcription factors. BMP4 inhibition is a required step in eliciting neuroectoderm from ectoderm and Foxd4l1.1 is one of the earliest neural genes highly expressed in the neuroectoderm and conserved across vertebrates, including humans. In this work, we focused on how Foxd4l1.1 downregulates the neural repressive pathway. Foxd4l1.1 inhibited BMP4/Smad1 signaling and triggered neuroectoderm formation in animal cap explants of Xenopus embryos. Foxd4l1.1 directly bound within the promoter of endogenous neural repressor ventx1.1 and inhibited ventx1.1 transcription. Foxd4l1.1 also physically interacted with Xbra in the nucleus and inhibited Xbra-induced ventx1.1 transcription. In addition, Foxd4l1.1 also reduced nuclear localization of Smad1 to inhibit Smad1-mediated ventx1.1 transcription. Foxd4l1.1 reduced the direct binding of Xbra and Smad1 on ventx1.1 promoter regions to block Xbra/Smad1-induced synergistic activation of ventx1.1 transcription. Collectively, Foxd4l1.1 negatively regulates transcription of a neural repressor ventx1.1 by multiple mechanisms in its exclusively occupied territory of neuroectoderm, and thus leading to primary neurogenesis. In conjunction with the results of our previous findings that ventx1.1 directly represses foxd4l1.1, the reciprocal repression of ventx1.1 and foxd4l1.1 is significant in at least in part specifying the mechanism for the non-neural versus neural ectoderm fate determination in Xenopus embryos.
Collapse
Affiliation(s)
- Shiv Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea
| | - Zobia Umair
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea
| | - Vijay Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea
| | - Santosh Kumar
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea
| | - Unjoo Lee
- Department of Electrical Engineering, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea.
| | - Jaebong Kim
- Department of Biochemistry, Institute of Cell Differentiation and Aging, College of Medicine, Hallym University, Gangwon-Do, Chuncheon, 24252, Republic of Korea.
| |
Collapse
|
23
|
Fractalkine Regulates HEC-1A/JEG-3 Interaction by Influencing the Expression of Implantation-Related Genes in an In Vitro Co-Culture Model. Int J Mol Sci 2020; 21:ijms21093175. [PMID: 32365902 PMCID: PMC7246682 DOI: 10.3390/ijms21093175] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/24/2020] [Accepted: 04/29/2020] [Indexed: 12/15/2022] Open
Abstract
Embryo implantation is a complex process regulated by a network of biological molecules. Recently, it has been described that fractalkine (CX3CL1, FKN) might have an important role in the feto-maternal interaction during gestation since the trophoblast cells express fractalkine receptor (CX3CR1) and the endometrium cells secrete fractalkine. CX3CR1 controls three major signalling pathways, PLC-PKC pathway, PI3K/AKT/NFκB pathway and Ras-mitogen-activated protein kinases (MAPK) pathways regulating proliferation, growth, migration and apoptosis. In this study, we focused on the molecular mechanisms of FKN treatment influencing the expression of implantation-related genes in trophoblast cells (JEG-3) both in mono-and in co-culture models. Our results reveal that FKN acted in a concentration and time dependent manner on JEG-3 cells. FKN seemed to operate as a positive regulator of implantation via changing the action of progesterone receptor (PR), activin receptor and bone morphogenetic protein receptor (BMPR). FKN modified also the expression of matrix metalloproteinase 2 and 9 controlling invasion. The presence of HEC-1A endometrial cells in the co-culture contributed to the effect of fractalkine on JEG-3 cells regulating implantation. The results suggest that FKN may contribute to the successful attachment and implantation of embryo.
Collapse
|
24
|
May RD, Frauchiger DA, Albers CE, Tekari A, Benneker LM, Klenke FM, Hofstetter W, Gantenbein B. Application of Cytokines of the Bone Morphogenetic Protein (BMP) Family in Spinal Fusion - Effects on the Bone, Intervertebral Disc and Mesenchymal Stromal Cells. Curr Stem Cell Res Ther 2020; 14:618-643. [PMID: 31455201 PMCID: PMC7040507 DOI: 10.2174/1574888x14666190628103528] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Abstract
Low back pain is a prevalent socio-economic burden and is often associated with damaged or degenerated intervertebral discs (IVDs). When conservative therapy fails, removal of the IVD (discectomy), followed by intersomatic spinal fusion, is currently the standard practice in clinics. The remaining space is filled with an intersomatic device (cage) and with bone substitutes to achieve disc height compensation and bone fusion. As a complication, in up to 30% of cases, spinal non-fusions result in a painful pseudoarthrosis. Bone morphogenetic proteins (BMPs) have been clinically applied with varied outcomes. Several members of the BMP family, such as BMP2, BMP4, BMP6, BMP7, and BMP9, are known to induce osteogenesis. Questions remain on why hyper-physiological doses of BMPs do not show beneficial effects in certain patients. In this respect, BMP antagonists secreted by mesenchymal cells, which might interfere with or block the action of BMPs, have drawn research attention as possible targets for the enhancement of spinal fusion or the prevention of non-unions. Examples of these antagonists are noggin, gremlin1 and 2, chordin, follistatin, BMP3, and twisted gastrulation. In this review, we discuss current evidence of the osteogenic effects of several members of the BMP family on osteoblasts, IVD cells, and mesenchymal stromal cells. We consider in vitro and in vivo studies performed in human, mouse, rat, and rabbit related to BMP and BMP antagonists in the last two decades. We give insights into the effects that BMP have on the ossification of the spine. Furthermore, the benefits, pitfalls, and possible safety concerns using these cytokines for the improvement of spinal fusion are discussed.
Collapse
Affiliation(s)
- Rahel Deborah May
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | | | - Christoph Emmanuel Albers
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| | - Adel Tekari
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Lorin Michael Benneker
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| | - Frank Michael Klenke
- Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| | - Willy Hofstetter
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Benjamin Gantenbein
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, University of Bern, Bern, Switzerland
| |
Collapse
|
25
|
Reich S, Weinstein DC. Repression of Inappropriate Gene Expression in the Vertebrate Embryonic Ectoderm. Genes (Basel) 2019; 10:E895. [PMID: 31698780 PMCID: PMC6895975 DOI: 10.3390/genes10110895] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/28/2019] [Accepted: 11/04/2019] [Indexed: 01/01/2023] Open
Abstract
During vertebrate embryogenesis, precise regulation of gene expression is crucial for proper cell fate determination. Much of what we know about vertebrate development has been gleaned from experiments performed on embryos of the amphibian Xenopus laevis; this review will focus primarily on studies of this model organism. An early critical step during vertebrate development is the formation of the three primary germ layers-ectoderm, mesoderm, and endoderm-which emerge during the process of gastrulation. While much attention has been focused on the induction of mesoderm and endoderm, it has become clear that differentiation of the ectoderm involves more than the simple absence of inductive cues; rather, it additionally requires the inhibition of mesendoderm-promoting genes. This review aims to summarize our current understanding of the various inhibitors of inappropriate gene expression in the presumptive ectoderm.
Collapse
Affiliation(s)
- Shoshana Reich
- PhD Program in Biology, The Graduate Center, The City University of New York, New York, NY 10016, USA
| | - Daniel C. Weinstein
- PhD Program in Biology, The Graduate Center, The City University of New York, New York, NY 10016, USA
- Department of Biology, Queens College, The City University of New York, Queens, NY 11367, USA
| |
Collapse
|
26
|
Asymmetric paralog evolution between the "cryptic" gene Bmp16 and its well-studied sister genes Bmp2 and Bmp4. Sci Rep 2019; 9:3136. [PMID: 30816280 PMCID: PMC6395752 DOI: 10.1038/s41598-019-40055-1] [Citation(s) in RCA: 229] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 02/07/2019] [Indexed: 12/05/2022] Open
Abstract
The vertebrate gene repertoire is characterized by “cryptic” genes whose identification has been hampered by their absence from the genomes of well-studied species. One example is the Bmp16 gene, a paralog of the developmental key genes Bmp2 and -4. We focus on the Bmp2/4/16 group of genes to study the evolutionary dynamics following gen(om)e duplications with special emphasis on the poorly studied Bmp16 gene. We reveal the presence of Bmp16 in chondrichthyans in addition to previously reported teleost fishes and reptiles. Using comprehensive, vertebrate-wide gene sampling, our phylogenetic analysis complemented with synteny analyses suggests that Bmp2, -4 and -16 are remnants of a gene quartet that originated during the two rounds of whole-genome duplication (2R-WGD) early in vertebrate evolution. We confirm that Bmp16 genes were lost independently in at least three lineages (mammals, archelosaurs and amphibians) and report that they have elevated rates of sequence evolution. This finding agrees with their more “flexible” deployment during development; while Bmp16 has limited embryonic expression domains in the cloudy catshark, it is broadly expressed in the green anole lizard. Our study illustrates the dynamics of gene family evolution by integrating insights from sequence diversification, gene repertoire changes, and shuffling of expression domains.
Collapse
|
27
|
Gazdagh G, Blyth M, Scurr I, Turnpenny PD, Mehta SG, Armstrong R, McEntagart M, Newbury-Ecob R, Tobias ES, Joss S. Extending the clinical and genetic spectrum of ARID2 related intellectual disability. A case series of 7 patients. Eur J Med Genet 2019; 62:27-34. [DOI: 10.1016/j.ejmg.2018.04.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/10/2018] [Accepted: 04/22/2018] [Indexed: 02/01/2023]
|
28
|
Ma JY, You D, Li WY, Lu XL, Sun S, Li HW. Bone morphogenetic proteins and inner ear development. J Zhejiang Univ Sci B 2018; 20:131-145. [PMID: 30112880 DOI: 10.1631/jzus.b1800084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bone morphogenetic proteins (BMPs) are the largest subfamily of the transforming growth factor-β superfamily, and they play important roles in the development of numerous organs, including the inner ear. The inner ear is a relatively small organ but has a highly complex structure and is involved in both hearing and balance. Here, we discuss BMPs and BMP signaling pathways and then focus on the role of BMP signal pathway regulation in the development of the inner ear and the implications this has for the treatment of human hearing loss and balance dysfunction.
Collapse
Affiliation(s)
- Jiao-Yao Ma
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Dan You
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Wen-Yan Li
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Xiao-Ling Lu
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Shan Sun
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Hua-Wei Li
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China.,Institutes of Biomedical Sciences and the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
29
|
Castro Colabianchi AM, Revinski DR, Encinas PI, Baez MV, Monti RJ, Rodríguez Abinal M, Kodjabachian L, Franchini LF, López SL. Notch1 is asymmetrically distributed from the beginning of embryogenesis and controls the ventral center. Development 2018; 145:dev.159368. [PMID: 29866901 DOI: 10.1242/dev.159368] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 05/25/2018] [Indexed: 12/14/2022]
Abstract
Based on functional evidence, we have previously demonstrated that early ventral Notch1 activity restricts dorsoanterior development in Xenopus We found that Notch1 has ventralizing properties and abolishes the dorsalizing activity of β-catenin by reducing its steady state levels, in a process that does not require β-catenin phosphorylation by glycogen synthase kinase 3β. In the present work, we demonstrate that Notch1 mRNA and protein are enriched in the ventral region from the beginning of embryogenesis in Xenopus This is the earliest sign of ventral development, preceding the localized expression of wnt8a, bmp4 and Ventx genes in the ventral center and the dorsal accumulation of nuclear β-catenin. Knockdown experiments indicate that Notch1 is necessary for the normal expression of genes essential for ventral-posterior development. These results indicate that during early embryogenesis ventrally located Notch1 promotes the development of the ventral center. Together with our previous evidence, these results suggest that ventral enrichment of Notch1 underlies the process by which Notch1 participates in restricting nuclear accumulation of β-catenin to the dorsal side.
Collapse
Affiliation(s)
- Aitana M Castro Colabianchi
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias 'Prof. E. De Robertis' (IBCN), Facultad de Medicina. Laboratorio de Embriología Molecular 'Prof. Dr. Andrés E. Carrasco', C1121ABG Buenos Aires, Argentina
| | - Diego R Revinski
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias 'Prof. E. De Robertis' (IBCN), Facultad de Medicina. Laboratorio de Embriología Molecular 'Prof. Dr. Andrés E. Carrasco', C1121ABG Buenos Aires, Argentina.,Aix Marseille Université, CNRS, IBDM, 13288 Marseille, France
| | - Paula I Encinas
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias 'Prof. E. De Robertis' (IBCN), Facultad de Medicina. Laboratorio de Embriología Molecular 'Prof. Dr. Andrés E. Carrasco', C1121ABG Buenos Aires, Argentina
| | - María Verónica Baez
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias 'Prof. E. De Robertis' (IBCN), Facultad de Medicina. Laboratorio de Embriología Molecular 'Prof. Dr. Andrés E. Carrasco', C1121ABG Buenos Aires, Argentina
| | - Renato J Monti
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias 'Prof. E. De Robertis' (IBCN), Facultad de Medicina. Laboratorio de Embriología Molecular 'Prof. Dr. Andrés E. Carrasco', C1121ABG Buenos Aires, Argentina
| | - Mateo Rodríguez Abinal
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias 'Prof. E. De Robertis' (IBCN), Facultad de Medicina. Laboratorio de Embriología Molecular 'Prof. Dr. Andrés E. Carrasco', C1121ABG Buenos Aires, Argentina
| | | | - Lucía F Franchini
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), C1428ADN Buenos Aires, Argentina
| | - Silvia L López
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias 'Prof. E. De Robertis' (IBCN), Facultad de Medicina. Laboratorio de Embriología Molecular 'Prof. Dr. Andrés E. Carrasco', C1121ABG Buenos Aires, Argentina
| |
Collapse
|
30
|
Evaluation of the toxic effects of celecoxib on Xenopus embryo development. Biochem Biophys Res Commun 2018; 501:329-335. [DOI: 10.1016/j.bbrc.2018.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 03/01/2018] [Indexed: 12/21/2022]
|
31
|
Abstract
TGF-β family ligands function in inducing and patterning many tissues of the early vertebrate embryonic body plan. Nodal signaling is essential for the specification of mesendodermal tissues and the concurrent cellular movements of gastrulation. Bone morphogenetic protein (BMP) signaling patterns tissues along the dorsal-ventral axis and simultaneously directs the cell movements of convergence and extension. After gastrulation, a second wave of Nodal signaling breaks the symmetry between the left and right sides of the embryo. During these processes, elaborate regulatory feedback between TGF-β ligands and their antagonists direct the proper specification and patterning of embryonic tissues. In this review, we summarize the current knowledge of the function and regulation of TGF-β family signaling in these processes. Although we cover principles that are involved in the development of all vertebrate embryos, we focus specifically on three popular model organisms: the mouse Mus musculus, the African clawed frog of the genus Xenopus, and the zebrafish Danio rerio, highlighting the similarities and differences between these species.
Collapse
Affiliation(s)
- Joseph Zinski
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| | - Benjamin Tajer
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| | - Mary C Mullins
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| |
Collapse
|
32
|
Bisgrove BW, Su YC, Yost HJ. Maternal Gdf3 is an obligatory cofactor in Nodal signaling for embryonic axis formation in zebrafish. eLife 2017; 6:28534. [PMID: 29140249 PMCID: PMC5745076 DOI: 10.7554/elife.28534] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 11/10/2017] [Indexed: 11/18/2022] Open
Abstract
Zebrafish Gdf3 (Dvr1) is a member of the TGFβ superfamily of cell signaling ligands that includes Xenopus Vg1 and mammalian Gdf1/3. Surprisingly, engineered homozygous mutants in zebrafish have no apparent phenotype. Elimination of Gdf3 in oocytes of maternal-zygotic mutants results in embryonic lethality that can be fully rescued with gdf3 RNA, demonstrating that Gdf3 is required only early in development, beyond which mutants are viable and fertile. Gdf3 mutants are refractory to Nodal ligands and Nodal repressor Lefty1. Signaling driven by TGFβ ligand Activin and constitutively active receptors Alk4 and Alk2 remain intact in gdf3 mutants, indicating that Gdf3 functions at the same pathway step as Nodal. Targeting gdf3 and ndr2 RNA to specific lineages indicates that exogenous gdf3 is able to fully rescue mutants only when co-expressed with endogenous Nodal. Together, these findings demonstrate that Gdf3 is an essential cofactor of Nodal signaling during establishment of the embryonic axis. All vertebrates – animals with backbones like fish and humans – have body plans with three clear axes: head-to-tail, back-to-front and left-to-right. Animals lay down these plans as embryos, when signaling molecules bind to receptors on the surface of their cells. These signaling molecules include related proteins called “Nodal” and “Growth and Differentiation Factors”. However, there has been much debate in the field of developmental biology about whether these proteins work together or independently during the early development of vertebrates. Zebrafish are often used to study animal development, and Bisgrove et al. decided to test whether these fish need a Growth and Differentiation Factor known as Gdf3 by deleting it using genome editing. It turns out that zebrafish can survive and develop as normal without the gene for Gdf3, just as long as their mothers still had a working copy of the gene. Yet, when the offspring of mutant females did not inherit the instructions to make Gdf3 from their mothers, they died within a couple of days. This was true even if the offspring inherited a working copy of the gene from their fathers. Bisgrove et al. then went on to show that embryos from a mutant mother could be saved with an injection of short-lived RNA molecules that include the instructions to make some Gdf3 proteins. The injected mutant embryos could live to adulthood. This shows that Gdf3 is only needed during the embryo’s early development. Further experiments suggested that Gdf3 does cannot activate its receptors on its own. Instead, it is likely that Gdf3 interacts with Nodal to form a two-protein complex that activates the receptors. Two other groups of researchers have independently reported similar findings. Mutations affecting proteins very similar to Gdf3 have been found in people with congenital heart defects. By revealing the interaction between Gdf3 and Nodal, these new findings could help scientists to understand the genetic causes of this condition in more detail. Further studies using the mutant zebrafish could also be used to explore the causes of other developmental diseases.
Collapse
Affiliation(s)
- Brent W Bisgrove
- Molecular Medicine Program, Eccles Institute of Human Genetics, University of Utah, Salt Lake City, United States
| | - Yi-Chu Su
- Molecular Medicine Program, Eccles Institute of Human Genetics, University of Utah, Salt Lake City, United States
| | - H Joseph Yost
- Molecular Medicine Program, Eccles Institute of Human Genetics, University of Utah, Salt Lake City, United States
| |
Collapse
|
33
|
Kennedy AE, Kandalam S, Olivares-Navarrete R, Dickinson AJG. E-cigarette aerosol exposure can cause craniofacial defects in Xenopus laevis embryos and mammalian neural crest cells. PLoS One 2017; 12:e0185729. [PMID: 28957438 PMCID: PMC5619826 DOI: 10.1371/journal.pone.0185729] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 09/18/2017] [Indexed: 12/28/2022] Open
Abstract
Since electronic cigarette (ECIG) introduction to American markets in 2007, vaping has surged in popularity. Many, including women of reproductive age, also believe that ECIG use is safer than traditional tobacco cigarettes and is not hazardous when pregnant. However, there are few studies investigating the effects of ECIG exposure on the developing embryo and nothing is known about potential effects on craniofacial development. Therefore, we have tested the effects of several aerosolized e-cigarette liquids (e-cigAM) in an in vivo craniofacial model, Xenopus laevis, as well as a mammalian neural crest cell line. Results demonstrate that e-cigAM exposure during embryonic development induces a variety of defects, including median facial clefts and midface hypoplasia in two of e-cigAMs tested e-cigAMs. Detailed quantitative analyses of the facial morphology revealed that nicotine is not the main factor in inducing craniofacial defects, but can exacerbate the effects of the other e-liquid components. Additionally, while two different e-cigAMs can have very similar consequences on facial appearances, there are subtle differences that could be due to the differences in e-cigAM components. Further assessment of embryos exposed to these particular e-cigAMs revealed cranial cartilage and muscle defects and a reduction in the blood supply to the face. Finally, the expression of markers for vascular and cartilage differentiation was reduced in a mammalian neural crest cell line corroborating the in vivo effects. Our work is the first to show that ECIG use could pose a potential hazard to the developing embryo and cause craniofacial birth defects. This emphasizes the need for more testing and regulation of this new popular product.
Collapse
Affiliation(s)
- Allyson E. Kennedy
- Virginia Commonwealth University, Department of Biology, Richmond, VA, United States of America
| | - Suraj Kandalam
- Virginia Commonwealth University, Department of Biomedical Engineering, Richmond, VA, United States of America
| | - Rene Olivares-Navarrete
- Virginia Commonwealth University, Department of Biomedical Engineering, Richmond, VA, United States of America
| | - Amanda J. G. Dickinson
- Virginia Commonwealth University, Department of Biology, Richmond, VA, United States of America
| |
Collapse
|
34
|
Suzuki A, Yoshida H, van Heeringen SJ, Takebayashi-Suzuki K, Veenstra GJC, Taira M. Genomic organization and modulation of gene expression of the TGF-β and FGF pathways in the allotetraploid frog Xenopus laevis. Dev Biol 2017; 426:336-359. [DOI: 10.1016/j.ydbio.2016.09.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 06/10/2016] [Accepted: 09/19/2016] [Indexed: 12/13/2022]
|
35
|
Tan S, Huan P, Liu B. Expression patterns indicate that BMP2/4 and Chordin, not BMP5-8 and Gremlin, mediate dorsal-ventral patterning in the mollusk Crassostrea gigas. Dev Genes Evol 2017; 227:75-84. [PMID: 27987051 DOI: 10.1007/s00427-016-0570-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 12/07/2016] [Indexed: 02/06/2023]
Abstract
Though several bilaterian animals use a conserved BMP2/4-Chordin antagonism to pattern the dorsal-ventral (DV) axis, the only lophotrochozoan species in which early DV patterning has been studied to date, the leech Helobdella robusta, appears to employ BMP5-8 and Gremlin. These findings call into question the conservation of a common DV patterning mechanism among bilaterian animals. To explore whether the unusual DV patterning mechanism in H. robusta is also used in other lophotrochozoan species, we investigated the expression of orthologous genes in the early embryo of a bivalve mollusk, Crassostrea gigas. Searching of the genome and phylogenetic analysis revealed that C. gigas possesses single orthologs of BMP2/4, Chordin, and BMP5-8 and no Gremlin homolog. Whole mount in situ hybridization revealed mRNA localization of BMP2/4 and Chordin on the opposite sides of embryos, suggesting the potential involvement of a BMP2/4-Chordin antagonism in DV patterning in this species. Furthermore, universal BMP5-8 expression and the absence of a Gremlin homolog in the C. gigas genome called into question any major contribution by BMP5-8 and Gremlin to early DV patterning in this species. Additionally, we identified seven genes showing asymmetric expression along the DV axis, providing further insight into DV patterning in C. gigas. We present the first report of a Chordin gene in a lophotrochozoan species and of the opposite expression of BMP2/4 (dorsal) and Chordin (ventral) along the D/V axis of a lophotrochozoan embryo. The findings of this study further the knowledge of axis formation in lophotrochozoan species and provide insight into the evolution of the animal DV patterning mechanism.
Collapse
Affiliation(s)
- Sujian Tan
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, China
- University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Pin Huan
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, China
| | - Baozhong Liu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266000, China.
| |
Collapse
|
36
|
Yoshida H, Okada M, Takebayashi-Suzuki K, Ueno N, Suzuki A. Involvement of JunB Proto-Oncogene in Tail Formation During Early Xenopus Embryogenesis. Zoolog Sci 2016; 33:282-9. [PMID: 27268982 DOI: 10.2108/zs150136] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Integration of signaling pathways is important for the establishment of the body plan during embryogenesis. However, little is known about how the multiple signals interact to regulate morphogenesis. Here, we show that junb is expressed in the posterior neural plate and the caudal fin during Xenopus embryogenesis and that overexpression of wild-type JunB induces small head phenotypes and ectopic tail-like structures. A mutant form of JunB that lacked GSK3 and MAPK phosphorylation sites showed stronger tail-like structure-inducing activity than wild-type JunB. Moreover, the mutant JunB induced expression of tailbud and neural marker genes, but not somite and chordoneural hinge (CNH) marker genes in ectopic tail-like structures. In ectodermal explants of Xenopus embryos, overexpression of JunB increased the expression of tailbud and posterior marker genes including fgf3, xbra (t) and wnt8. These results indicate that JunB is capable of inducing the ectopic formation of tissues similar to the tailbud, and that the tailbud-inducing activity of JunB is likely to be regulated by FGF and Wnt pathways. Overall, our results suggest that JunB is a regulator of tail organization possibly through integration of several morphogen signaling pathways.
Collapse
Affiliation(s)
- Hitoshi Yoshida
- 1 Institute for Amphibian Biology, Graduate School of Science, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima 739-8526, Japan
| | - Maya Okada
- 1 Institute for Amphibian Biology, Graduate School of Science, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima 739-8526, Japan
| | - Kimiko Takebayashi-Suzuki
- 1 Institute for Amphibian Biology, Graduate School of Science, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima 739-8526, Japan
| | - Naoto Ueno
- 2 Division of Morphogenesis, National Institute for Basic Biology, 38 Nishigonaka, Myodaiji, Okazaki, Aichi 444-8585, Japan.,3 Department of Basic Biology, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Shonan Village, Hayama, Kanagawa 240-0193, Japan
| | - Atsushi Suzuki
- 1 Institute for Amphibian Biology, Graduate School of Science, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima 739-8526, Japan
| |
Collapse
|
37
|
Bami M, Mavrogenis AF, Angelini A, Milonaki M, Mitsiokapa E, Stamoulis D, Soucacos PN. Bone morphogenetic protein signaling in musculoskeletal cancer. J Cancer Res Clin Oncol 2016; 142:2061-72. [PMID: 27043154 DOI: 10.1007/s00432-016-2149-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 03/17/2016] [Indexed: 02/08/2023]
Abstract
PURPOSE Bone morphogenetic proteins (BMPs) belong to the transforming growth factor-β (TGF-β) superfamily of proteins; they were initially named after their ability to induce ectopic bone formation. Published studies have proved BMPs' role in a variety of biological processes such as embryogenesis and patterning of body axes, and maintaining adult tissue homeostasis. Other studies have focused on BMPs properties, functions and possible involvement in skeletal diseases, including cancer. METHODS A literature search mainly paying attention to the role of BMPs in musculoskeletal tumors was performed in electronic databases. RESULTS This article discusses BMPs synthesis and signaling, and summarizes their prominent roles in the skeletal system for the differentiation of osteoblasts, osteocytes and chondrocytes. CONCLUSIONS The review emphasizes on the role of BMP signaling in the initiation and progression of musculoskeletal cancer.
Collapse
Affiliation(s)
- Myrto Bami
- Orthopaedic Research and Education Center, "Panayotis N. Soucacos", Athens University Medical School, Attikon University Hospital, Athens, Greece
| | - Andreas F Mavrogenis
- First Department of Orthopaedics, Athens University Medical School, Attikon University Hospital, Athens, Greece.
| | - Andrea Angelini
- Department of Orthopaedics, Istituto Ortopedico Rizzoli, University of Bologna, Via Pupilli, 1, 40136, Bologna, Italy
| | - Mandy Milonaki
- Orthopaedic Research and Education Center, "Panayotis N. Soucacos", Athens University Medical School, Attikon University Hospital, Athens, Greece
| | - Evanthia Mitsiokapa
- Orthopaedic Research and Education Center, "Panayotis N. Soucacos", Athens University Medical School, Attikon University Hospital, Athens, Greece
| | - Dimitrios Stamoulis
- Orthopaedic Research and Education Center, "Panayotis N. Soucacos", Athens University Medical School, Attikon University Hospital, Athens, Greece
| | - Panayotis N Soucacos
- First Department of Orthopaedics, Athens University Medical School, Attikon University Hospital, Athens, Greece
| |
Collapse
|
38
|
Tang XB, Zhang J, Wang WL, Yuan ZW, Bai YZ. The expression analysis of Bmpr1a and Bmp2 during hindgut development in rat embryos with anorectal malformations. Exp Mol Pathol 2016; 101:143-9. [PMID: 27477499 DOI: 10.1016/j.yexmp.2016.07.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/07/2016] [Accepted: 07/27/2016] [Indexed: 11/20/2022]
Abstract
The aim of this study was to determine Bmpr1a and Bmp2 expression patterns during anorectal development in normal and anorectal malformation (ARM) embryos with a view to establishing the possible role of Bmpr1a and Bmp2 in ARM pathogenesis. ARM was induced with ethylenethiourea on the 10th gestational day (GD10) in rat embryos. The embryos were harvested by Cesarean deliveries. The expression of Bmpr1a and Bmp2 was evaluated in normal rat embryos (n=213) and ARM embryos (n=236) from GD14 to GD16. Immunohistochemical staining revealed, in normal embryos, that Bmpr1a and Bmp2 was mainly expressed on the epithelium of the urorectal septum (URS) and the cloacal membrane (CM) on GD14 and GD15. When the rectum separated from the urogenital sinus (UGS) on GD16, Bmpr1a- and Bmp2-immunolabeled cells were observed on the anorectal epithelium. In ARM embryos, the epithelium of the hindgut and URS demonstrated faint immunostaining for Bmpr1a and Bmp2. Analyses by Western blot and Real-time PCR revealed that Bmpr1a and Bmp2 protein and mRNA expression were significantly decreased in the ARM hindgut compared with normal hindgut on GD14 and GD15 (P<0.05). In ARM embryos, an imbalance in the spatiotemporal expression of Bmpr1a and Bmp2 was noted during anorectal morphogenesis from GD14 to GD16. Therefore, downregulation of Bmpr1a and Bmp2 at the time of cloacal separation into the primitive rectum and UGS might be related to the development of ARM.
Collapse
Affiliation(s)
- Xiao Bing Tang
- Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang 110004, PR China
| | - Jin Zhang
- Department of Pediatrics, Women and Children's Hospital of Qingdao City, Qingdao 266034, PR China
| | - Wei Lin Wang
- Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang 110004, PR China
| | - Zheng Wei Yuan
- The Key Laboratory of Health Ministry for Congenital Malformation, Shenyang 110004, PR China
| | - Yu Zuo Bai
- Department of Pediatric Surgery, Shengjing Hospital, China Medical University, Shenyang 110004, PR China.
| |
Collapse
|
39
|
BMP and retinoic acid regulate anterior-posterior patterning of the non-axial mesoderm across the dorsal-ventral axis. Nat Commun 2016; 7:12197. [PMID: 27406002 PMCID: PMC4947171 DOI: 10.1038/ncomms12197] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 06/09/2016] [Indexed: 11/23/2022] Open
Abstract
Despite the fundamental importance of patterning along the dorsal–ventral (DV) and anterior–posterior (AP) axes during embryogenesis, uncertainty exists in the orientation of these axes for the mesoderm. Here we examine the origin and formation of the zebrafish kidney, a ventrolateral mesoderm derivative, and show that AP patterning of the non-axial mesoderm occurs across the classic gastrula stage DV axis while DV patterning aligns along the animal–vegetal pole. We find that BMP signalling acts early to establish broad anterior and posterior territories in the non-axial mesoderm while retinoic acid (RA) functions later, but also across the classic DV axis. Our data support a model in which RA on the dorsal side of the embryo induces anterior kidney fates while posterior kidney progenitors are protected ventrally by the RA-catabolizing enzyme Cyp26a1. This work clarifies our understanding of vertebrate axis orientation and establishes a new paradigm for how the kidney and other mesodermal derivatives arise during embryogenesis. It is unclear how the dorsal-ventral (DV) and anterior-posterior (AP) axes established in the gastrula affect tissues. Here, the authors show that in zebrafish kidney development, with regard to non-axial mesoderm, the classic DV axis corresponds to the AP axis, and is regulated by BMP and retinoic acid.
Collapse
|
40
|
Li FF, Deng X, Zhou J, Yan P, Zhao EY, Liu SL. Characterization of human bone morphogenetic protein gene variants for possible roles in congenital heart disease. Mol Med Rep 2016; 14:1459-64. [PMID: 27357418 PMCID: PMC4940093 DOI: 10.3892/mmr.2016.5428] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 04/25/2016] [Indexed: 12/25/2022] Open
Abstract
Congenital heart disease (CHD) is a complex illness with high rates of morbidity and mortality. In embryonic development, the heart is the first formed organ, which is strictly controlled by gene regulatory networks, including transcription factors, signaling pathways, epigenetic factors and microRNAs. Bone morphogenetic protein (BMP)-2 and -4 are essential in cardiogenesis as they can induce the expression of transcription factors, NKX2-5 and GATA binding protein 4, which are important in the development of the heart. The inhibition of BMP-2 and 4- inhibits the late expression of NKX2-5 and affects cardiac differentiation. The aim of the present study was to investigate whether BMP-2 and -4 variations may be associated with CHD in Chinese Han populations. The rs1049007, rs235768 and rs17563 single nucleotide polymorphisms (SNPs), which are genetic variations located within the translated region of the BMP-2 and -4, were evaluated in 230 patients with CHD from the Chinese Han population and 160 non CHD control individuals. Statistical analyses were performed using the χ2 test, implemented using SPSS software (version 13.0). The Hardy Weinberg equilibrium test was performed on the population using online Online Encyclopedia for Genetic Epidemiology studies software, and multiple-sequence alignments of the BMP proteins were performed using Vector NTI software. No statistically significant associations were identified between these genetic variations and the risk of CHD (rs1049007, P value=0.560; rs235768, P value=0.972; rs17563, P value=0.787). In addition, no correlation was found between the patients with CHD and the non-CHD control individuals. Therefore, the rs1049007, rs235768 and rs17563 genetic variations of BMP-2 were not associated with CHD in the Chinese Han population.
Collapse
Affiliation(s)
- Fei-Feng Li
- Genomics Research Center (one of the State‑Key Laboratory of Biopharmaceutical Engineering), Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xia Deng
- Genomics Research Center (one of the State‑Key Laboratory of Biopharmaceutical Engineering), Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jing Zhou
- Intensive Care Unit, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Peng Yan
- Department of Colorectal Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Er-Ying Zhao
- Genomics Research Center (one of the State‑Key Laboratory of Biopharmaceutical Engineering), Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Shu-Lin Liu
- Genomics Research Center (one of the State‑Key Laboratory of Biopharmaceutical Engineering), Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
41
|
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
|
42
|
Lei X, Cui K, Li Z, Su J, Jiang J, Zhang H, Liu Q, Shi D. BMP-1 participates in the selection and dominance of buffalo follicles by regulating the proliferation and apoptosis of granulosa cells. Theriogenology 2015; 85:999-1012. [PMID: 26778140 DOI: 10.1016/j.theriogenology.2015.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 11/10/2015] [Accepted: 11/14/2015] [Indexed: 11/30/2022]
Abstract
BMP1/TLD-related metalloproteinases play a key role in morphogenesis via the proteolytic maturation of a number of extracellular matrix proteins and the activation of a subset of growth factors of the transforming growth factor beta superfamily. Recent data indicated that BMP1 is expressed in sheep ovarian follicles and showed a protease activity. The aim of the present study was to characterize the function of the buffalo BMP1 gene in folliculogenesis. A 3195-bp buffalo BMP1 mRNA fragment was firstly cloned and sequenced, which contained a whole 2967-bp codon sequence. The multialigned results suggested that BMP1 is highly conserved among different species both at the nucleic acid and the amino acid level. BMP1 is located in the oogonium of the fetal buffalo ovary and in the granulosa cells (GCs) and the oocytes of adult ovary from the primordial to the large antral follicles. Further study showed that BMP1 promoted cell cycle and proliferation and inhibited apoptosis in IVC GCs. Adding BMP1 recombinant protein to the culture medium of the GCs increased the expression of the key cell cycle regulators such as cyclin D1 and cyclin D2 and downregulated the expression of cell apoptosis pathway genes such as Cytochrome C, Fas, FasL, and Chop, both at the mRNA and at the protein levels. It also upregulated the expression of PAPP-A, IGF system, and VEGF, and so forth, which play important roles in the selection and dominance of growth follicles. The opposite results were observed by adding BMP1 antibody to the investigation groups. This study suggests that BMP1 regulates the proliferation and apoptosis of IVC GCs by changing the expression pattern of related genes and may potentially promote the selection and dominance of the buffalo follicles.
Collapse
Affiliation(s)
- Xiaocan Lei
- Animal Science Department, Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, China
| | - Kuiqing Cui
- Animal Science Department, Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, China
| | - Zhipeng Li
- Animal Science Department, Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, China
| | - Jie Su
- Animal Science Department, Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, China
| | - Jianrong Jiang
- Animal Science Department, Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, China
| | - Haihang Zhang
- Animal Science Department, Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, China
| | - Qingyou Liu
- Animal Science Department, Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, China.
| | - Deshun Shi
- Animal Science Department, Animal Reproduction Institute, State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, Guangxi University, Nanning, China
| |
Collapse
|
43
|
Tuazon FB, Mullins MC. Temporally coordinated signals progressively pattern the anteroposterior and dorsoventral body axes. Semin Cell Dev Biol 2015; 42:118-33. [PMID: 26123688 PMCID: PMC4562868 DOI: 10.1016/j.semcdb.2015.06.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 06/16/2015] [Indexed: 10/23/2022]
Abstract
The vertebrate body plan is established through the precise spatiotemporal coordination of morphogen signaling pathways that pattern the anteroposterior (AP) and dorsoventral (DV) axes. Patterning along the AP axis is directed by posteriorizing signals Wnt, fibroblast growth factor (FGF), Nodal, and retinoic acid (RA), while patterning along the DV axis is directed by bone morphogenetic proteins (BMP) ventralizing signals. This review addresses the current understanding of how Wnt, FGF, RA and BMP pattern distinct AP and DV cell fates during early development and how their signaling mechanisms are coordinated to concomitantly pattern AP and DV tissues.
Collapse
Affiliation(s)
- Francesca B Tuazon
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, 1152 BRBII/III, 421 Curie Boulevard, Philadelphia, PA 19104-6058, United States
| | - Mary C Mullins
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, 1152 BRBII/III, 421 Curie Boulevard, Philadelphia, PA 19104-6058, United States.
| |
Collapse
|
44
|
Sox5 Is a DNA-binding cofactor for BMP R-Smads that directs target specificity during patterning of the early ectoderm. Dev Cell 2014; 31:374-382. [PMID: 25453832 DOI: 10.1016/j.devcel.2014.10.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 07/16/2014] [Accepted: 10/02/2014] [Indexed: 11/21/2022]
Abstract
The SoxD factor, Sox5, is expressed in ectodermal cells at times and places where BMP signaling is active, including the cells of the animal hemisphere at blastula stages and the neural plate border and neural crest at neurula stages. Sox5 is required for proper ectoderm development, and deficient embryos display patterning defects characteristic of perturbations of BMP signaling, including loss of neural crest and epidermis and expansion of the neural plate. We show that Sox5 is essential for activation of BMP target genes in embryos and explants, that it physically interacts with BMP R-Smads, and that it is essential for recruitment of Smad1/4 to BMP regulatory elements. Our findings identify Sox5 as the long-sought DNA-binding partner for BMP R-Smads essential to plasticity and pattern in the early ectoderm.
Collapse
|
45
|
Wang RN, Green J, Wang Z, Deng Y, Qiao M, Peabody M, Zhang Q, Ye J, Yan Z, Denduluri S, Idowu O, Li M, Shen C, Hu A, Haydon RC, Kang R, Mok J, Lee MJ, Luu HL, Shi LL. Bone Morphogenetic Protein (BMP) signaling in development and human diseases. Genes Dis 2014; 1:87-105. [PMID: 25401122 PMCID: PMC4232216 DOI: 10.1016/j.gendis.2014.07.005] [Citation(s) in RCA: 714] [Impact Index Per Article: 64.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 07/15/2014] [Indexed: 02/06/2023] Open
Abstract
Bone Morphogenetic Proteins (BMPs) are a group of signaling molecules that belongs to the Transforming Growth Factor-β (TGF-β) superfamily of proteins. Initially discovered for their ability to induce bone formation, BMPs are now known to play crucial roles in all organ systems. BMPs are important in embryogenesis and development, and also in maintenance of adult tissue homeostasis. Mouse knockout models of various components of the BMP signaling pathway result in embryonic lethality or marked defects, highlighting the essential functions of BMPs. In this review, we first outline the basic aspects of BMP signaling and then focus on genetically manipulated mouse knockout models that have helped elucidate the role of BMPs in development. A significant portion of this review is devoted to the prominent human pathologies associated with dysregulated BMP signaling.
Collapse
Affiliation(s)
- Richard N. Wang
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jordan Green
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zhongliang Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Youlin Deng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Min Qiao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Michael Peabody
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Qian Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Jixing Ye
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- School of Bioengineering, Chongqing University, Chongqing, China
| | - Zhengjian Yan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Sahitya Denduluri
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Olumuyiwa Idowu
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Melissa Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Christine Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Alan Hu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Richard Kang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - James Mok
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue L. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Lewis L. Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
46
|
Schlosser G. Early embryonic specification of vertebrate cranial placodes. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2014; 3:349-63. [PMID: 25124756 DOI: 10.1002/wdev.142] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 05/09/2014] [Accepted: 05/16/2014] [Indexed: 12/16/2022]
Abstract
UNLABELLED Cranial placodes contribute to many sensory organs and ganglia of the vertebrate head. The olfactory, otic, and lateral line placodes form the sensory receptor cells and neurons of the nose, ear, and lateral line system; the lens placode develops into the lens of the eye; epibranchial, profundal, and trigeminal placodes contribute sensory neurons to cranial nerve ganglia; and the adenohypophyseal placode gives rise to the anterior pituitary, a major endocrine control organ. Despite these differences in fate, all placodes are now known to originate from a common precursor, the preplacodal ectoderm (PPE). The latter is a horseshoe-shaped domain of ectoderm surrounding the anterior neural plate and neural crest and is defined by expression of transcription factor Six1, its cofactor Eya1, and other members of the Six and Eya families. Studies in zebrafish, Xenopus, and chick reveal that the PPE is specified together with other ectodermal territories (epidermis, neural crest, and neural plate) during early embryogenesis. During gastrulation, domains of ventrally (e.g., Dlx3/Dlx5, GATA2/GATA3, AP2, Msx1, FoxI1, and Vent1/Vent2) and dorsally (e.g., Zic1, Sox3, and Geminin) restricted transcription factors are established in response to a gradient of BMP and help to define non-neural and neural competence territories, respectively. At neural plate stages, the PPE is then induced in the non-neural competence territory by signals from the adjacent neural plate and mesoderm including FGF, BMP inhibitors, and Wnt inhibitors. Subsequently, signals from more localized signaling centers induce restricted expression domains of various transcription factors within the PPE, which specify multiplacodal areas and ultimately individual placodes. For further resources related to this article, please visit the WIREs website. CONFLICT OF INTEREST The author has declared no conflicts of interest for this article.
Collapse
Affiliation(s)
- Gerhard Schlosser
- Department of Zoology, School of Natural Sciences & Regenerative Medicine Institute (REMEDI), National University of Ireland, Galway, Galway, Ireland
| |
Collapse
|
47
|
Chatfield J, O'Reilly MA, Bachvarova RF, Ferjentsik Z, Redwood C, Walmsley M, Patient R, Loose M, Johnson AD. Stochastic specification of primordial germ cells from mesoderm precursors in axolotl embryos. Development 2014; 141:2429-40. [PMID: 24917499 PMCID: PMC4050694 DOI: 10.1242/dev.105346] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 04/22/2014] [Indexed: 01/18/2023]
Abstract
A common feature of development in most vertebrate models is the early segregation of the germ line from the soma. For example, in Xenopus and zebrafish embryos primordial germ cells (PGCs) are specified by germ plasm that is inherited from the egg; in mice, Blimp1 expression in the epiblast mediates the commitment of cells to the germ line. How these disparate mechanisms of PGC specification evolved is unknown. Here, in order to identify the ancestral mechanism of PGC specification in vertebrates, we studied PGC specification in embryos from the axolotl (Mexican salamander), a model for the tetrapod ancestor. In the axolotl, PGCs develop within mesoderm, and classic studies have reported their induction from primitive ectoderm (animal cap). We used an axolotl animal cap system to demonstrate that signalling through FGF and BMP4 induces PGCs. The role of FGF was then confirmed in vivo. We also showed PGC induction by Brachyury, in the presence of BMP4. These conditions induced pluripotent mesodermal precursors that give rise to a variety of somatic cell types, in addition to PGCs. Irreversible restriction of the germ line did not occur until the mid-tailbud stage, days after the somatic germ layers are established. Before this, germline potential was maintained by MAP kinase signalling. We propose that this stochastic mechanism of PGC specification, from mesodermal precursors, is conserved in vertebrates.
Collapse
Affiliation(s)
- Jodie Chatfield
- School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham NG7 2UH, UK
| | - Marie-Anne O'Reilly
- School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham NG7 2UH, UK
| | - Rosemary F Bachvarova
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Zoltan Ferjentsik
- School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham NG7 2UH, UK
| | - Catherine Redwood
- School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham NG7 2UH, UK
| | - Maggie Walmsley
- Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Oxford University, Oxford OX3 9DS, UK
| | - Roger Patient
- Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Oxford University, Oxford OX3 9DS, UK
| | - Mathew Loose
- School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham NG7 2UH, UK
| | - Andrew D Johnson
- School of Life Sciences, University of Nottingham, Queens Medical Centre, Nottingham NG7 2UH, UK
| |
Collapse
|
48
|
Organizer-derived Bmp2 is required for the formation of a correct Bmp activity gradient during embryonic development. Nat Commun 2014; 5:3766. [PMID: 24777107 PMCID: PMC4071459 DOI: 10.1038/ncomms4766] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Accepted: 03/31/2014] [Indexed: 01/21/2023] Open
Abstract
Bone morphogenetic proteins (Bmps) control dorsoventral patterning of vertebrate embryos through the establishment of a ventrodorsal gradient of the activated downstream cytoplasmic effectors Smad1/5/8. Some Bmp ligands are expressed in the ventral and lateral regions and in the organizer during gastrulation of the embryo, but it remains unclear how organizer-derived Bmps contribute to total Bmp ligand levels and to the establishment of the correct phospho-Smad1/5/8 gradient along the ventrodorsal axis. Here we demonstrate that interference with organizer-specific Bmp2b signalling in zebrafish embryos alters the phospho-Smad1/5/8 gradient throughout the ventrodorsal axis, elevates the levels of the Bmp antagonist Chordin and dorsalizes the embryos. Moreover, we show that organizer-derived Bmp2b represses chordin transcription in the organizer and contributes to the control of the Chordin gradient. Combining these experimental results with simulations of Bmp’s reaction-diffusion dynamics, our data indicate that organizer-produced Bmp2b is required for the establishment and maintenance of a Bmp activity gradient and for appropriate embryonic dorsoventral patterning during gastrulation. The morphogen, Bmp, regulates differentiation of cell fates along the ventral to dorsal axis during vertebrate embryonic development. Here, Xue et al. show that Bmp2b produced by the organizer during early gastrulation in zebrafish embryos has a role in the establishment of an appropriate Bmp morphogen activity gradient and the correct dorsoventral patterning of the embryos.
Collapse
|
49
|
Amin NM, Tandon P, Osborne Nishimura E, Conlon FL. RNA-seq in the tetraploid Xenopus laevis enables genome-wide insight in a classic developmental biology model organism. Methods 2014; 66:398-409. [PMID: 23792920 PMCID: PMC3884041 DOI: 10.1016/j.ymeth.2013.06.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 05/07/2013] [Accepted: 06/13/2013] [Indexed: 01/08/2023] Open
Abstract
Advances in sequencing technology have significantly advanced the landscape of developmental biology research. The dissection of genetic networks in model and non-model organisms has been greatly enhanced with high-throughput sequencing technologies. RNA-seq has revolutionized the ability to perform developmental biology research in organisms without a published genome sequence. Here, we describe a protocol for developmental biologists to perform RNA-seq on dissected tissue or whole embryos. We start with the isolation of RNA and generation of sequencing libraries. We further show how to interpret and analyze the large amount of sequencing data that is generated in RNA-seq. We explore the abilities to examine differential expression, gene duplication, transcript assembly, alternative splicing and SNP discovery. For the purposes of this article, we use Xenopus laevis as the model organism to discuss uses of RNA-seq in an organism without a fully annotated genome sequence.
Collapse
Affiliation(s)
- Nirav M Amin
- University of North Carolina, McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599-3280, USA; Department of Genetics, UNC-Chapel Hill, Chapel Hill, NC 27599-3280, USA
| | - Panna Tandon
- University of North Carolina, McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599-3280, USA; Department of Genetics, UNC-Chapel Hill, Chapel Hill, NC 27599-3280, USA
| | | | - Frank L Conlon
- University of North Carolina, McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599-3280, USA; Department of Biology, UNC-Chapel Hill, Chapel Hill, NC 27599-3280, USA; Department of Genetics, UNC-Chapel Hill, Chapel Hill, NC 27599-3280, USA.
| |
Collapse
|
50
|
Kraushaar DC, Dalton S, Wang L. Heparan sulfate: a key regulator of embryonic stem cell fate. Biol Chem 2014; 394:741-51. [PMID: 23370908 DOI: 10.1515/hsz-2012-0353] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 01/23/2013] [Indexed: 12/11/2022]
Abstract
Heparan sulfate (HS) belongs to a class of glycosaminoglycans and is a highly sulfated, linear polysaccharide. HS biosynthesis and modification involves numerous enzymes. HS exists as part of glycoproteins named HS proteoglycans, which are expressed abundantly on the cell surface and in the extracellular matrix. HS interacts with numerous proteins, including growth factors, morphogens, and adhesion molecules, and thereby regulates important developmental processes in invertebrates and vertebrates. Embryonic stem cells (ESCs) are distinguished by their characteristics of self-renewal and pluripotency. Self-renewal allows ESCs to proliferate indefinitely in their undifferentiated state, whereas pluripotency implies their capacity to differentiate into the three germ layers and ultimately all cell types of the adult body. Both traits are tightly regulated by numerous cell signaling pathways. Recent studies have highlighted the importance of HS in the modulation of ESC functions, specifically their lineage fate. Here, we review the current advances that have been made in understanding the structural changes of HS during ESC differentiation and in deciphering the molecular mechanisms by which HS modulates cell fate. Finally, we discuss the applications of heparinoids and chemical inhibitors of HS biosynthesis for the manipulation of ESC culture and directed differentiation.
Collapse
Affiliation(s)
- Daniel C Kraushaar
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, USA
| | | | | |
Collapse
|