1
|
Yao Y. Laminin Receptors in the CNS and Vasculature. Stroke 2025. [PMID: 40421534 DOI: 10.1161/strokeaha.125.051560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Laminin exerts a variety of important functions via binding to its receptors, including integrins and dystroglycan. With the advance in gene-targeting technology, many integrin/dystroglycan knockout/mutant mice were generated in the past 3 decades. These mutants enable loss-of-function studies and have substantially enriched our knowledge of integrin/dystroglycan functions. In this review, we summarize the functions of laminin receptors during embryonic development and in the CNS and vasculature. First, the biochemical properties of integrins and dystroglycan are briefly introduced. Next, we discuss loss-of-function studies on laminin receptors, including integrin-α3, integrin-α6, integrin-α7, integrin-β1, integrin-β4, and dystroglycan, focusing on embryonic development, the CNS, and vasculature. The phenotypes of compound knockout mice are described and compared with that of single mutants. Last, important questions and challenges in the field as well as potential future directions are discussed. Our goal is to provide a synthetic review on loss-of-function studies of laminin receptors in the CNS and vasculature, which could serve as a reference for future research, encourage the formation of new hypotheses, and stimulate new research in this field.
Collapse
Affiliation(s)
- Yao Yao
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| |
Collapse
|
2
|
Yoshida S, Tsuneoka Y, Tsukada T, Nakakura T, Kawamura A, Kai W, Yoshida K. Primary Cilia are Required for Cell-Type Determination and Angiogenesis in Pituitary Development. Endocrinology 2024; 165:bqae085. [PMID: 39001875 DOI: 10.1210/endocr/bqae085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/26/2024] [Accepted: 07/11/2024] [Indexed: 07/15/2024]
Abstract
The functional maturation of the pituitary gland requires adequate cell differentiation and vascular network formation. Although spatiotemporal signaling and transcription factors are known to govern pituitary development, the involvement of primary cilia, nonmoving hair-like organelles, remains unclear. In this study, we uncovered the contribution of primary cilia to cell-type determination and vascular network formation during pituitary development. Homozygous knockout mice lacking a ciliary kinase, Dyrk2-/-, exhibit abnormalities in ciliary structure and pituitary hypoplasia, accompanied by varying degrees of failure in differentiation among all types of hormone-producing cells in the anterior lobe. Aberrations in cell differentiation in Dyrk2-/- mice arise from a decrease in the expression of crucial transcription factors, Lhx4, Lhx3, and Prop1, resulting from the inactivity of Hedgehog (Hh) signaling during the early stages of development. Furthermore, the loss of Dyrk2 results in vascular system abnormalities during the middle to late stages of development. Mechanistically, transcriptome analyses revealed the downregulation of vitronectin-integrin αvβ3-VEGFR2 signaling, essential for orchestrating vascular development. Collectively, our findings demonstrate that primary cilia play a pivotal role as critical regulators of cell survival, cell determination, and angiogenesis during pituitary gland development through the activation of Hh signaling. These findings expand our understanding of the potential link between pituitary dysfunction in human disorders and ciliopathies.
Collapse
Affiliation(s)
- Saishu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo 105-8461, Japan
- Department of Biomolecular Science, Toho University, Chiba 274-8510, Japan
| | - Yousuke Tsuneoka
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo 143-8540, Japan
| | - Takehiro Tsukada
- Department of Biomolecular Science, Toho University, Chiba 274-8510, Japan
| | - Takashi Nakakura
- Department of Anatomy, Graduate School of Medicine, Teikyo University, Tokyo 173-8605, Japan
| | - Akira Kawamura
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Wataru Kai
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Kiyotsugu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| |
Collapse
|
3
|
Zhang D, Hugo W, Bergsneider M, Wang MB, Kim W, Han K, Vinters HV, Heaney AP. Cabergoline targets multiple pathways to inhibit PRL secretion and increases stromal fibrosis. Eur J Endocrinol 2024; 190:467-478. [PMID: 38781434 PMCID: PMC12054100 DOI: 10.1093/ejendo/lvae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/23/2024] [Accepted: 05/22/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE Unravel the potential mechanism(s) of the on- and off-target actions of dopamine agonist therapy in both human prolactinoma tumors and neighboring stromal and immune cells. DESIGN AND METHODS Five surgically resected prolactinomas (PRLomas) from 3 cabergoline (CBG)-treated patients and 2 treatment-naive patients were analyzed by using single-cell RNA sequencing (scRNA-seq) to compare the cellular composition and transcriptional landscape. RESULTS Six major cell populations, namely tumor (88.2%), immune (5.6%), stromal (4.9%), progenitor cells (0.6%), proliferating cells (0.4%), and erythrocytes (0.2%), were observed. Tumor cells from CBG-treated patients expressed lower levels of genes that regulated hormone secretion, such as SCG2, VGF, TIMP1, NNAT, and CALD1, consistent with the inhibitory effects of CBG on hormone processing and secretion. Interestingly, we also observed an increased number of CD8+ T cells in the CBG-treated tissues. These cytotoxic CD8+ T cells expressed killing granule components such as perforin and the granzymes GZMB, GNLY, and KLRD1 as well as the inflammatory cytokine CCL5. Immune cell activation of these CD8+ T cells was further analyzed in a compartment-specific manner, and increased CD25 (IL2R) expression was noted in the CD8+ T cells from the CBG-treated samples. Additionally, and confirming prior reports, we noted a higher stromal cell population in the CBG-treated samples. CONCLUSIONS Our scRNA-seq studies revealed key differences in the transcriptomic features of CBG-treated and CBG-untreated PRLomas in both tumor and microenvironment cellular constituents, and for the first time, describe the previously unknown activation of CD8+ T cells following CBG treatment, which may play a role in the tumoricidal actions of CBG.
Collapse
Affiliation(s)
- Dongyun Zhang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Willy Hugo
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Marvin Bergsneider
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles
| | - Marilene B. Wang
- Department of Head and Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles
| | - Won Kim
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles
| | - Karam Han
- Department of Pathology and Lab Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Harry V. Vinters
- Department of Pathology and Lab Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Anthony P. Heaney
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles
| |
Collapse
|
4
|
Tixi W, Maldonado M, Chang YT, Chiu A, Yeung W, Parveen N, Nelson MS, Hart R, Wang S, Hsu WJ, Fueger P, Kopp JL, Huising MO, Dhawan S, Shih HP. Coordination between ECM and cell-cell adhesion regulates the development of islet aggregation, architecture, and functional maturation. eLife 2023; 12:e90006. [PMID: 37610090 PMCID: PMC10482429 DOI: 10.7554/elife.90006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/12/2023] [Indexed: 08/24/2023] Open
Abstract
Pancreatic islets are three-dimensional cell aggregates consisting of unique cellular composition, cell-to-cell contacts, and interactions with blood vessels. Cell aggregation is essential for islet endocrine function; however, it remains unclear how developing islets establish aggregation. By combining genetic animal models, imaging tools, and gene expression profiling, we demonstrate that islet aggregation is regulated by extracellular matrix signaling and cell-cell adhesion. Islet endocrine cell-specific inactivation of extracellular matrix receptor integrin β1 disrupted blood vessel interactions but promoted cell-cell adhesion and the formation of larger islets. In contrast, ablation of cell-cell adhesion molecule α-catenin promoted blood vessel interactions yet compromised islet clustering. Simultaneous removal of integrin β1 and α-catenin disrupts islet aggregation and the endocrine cell maturation process, demonstrating that establishment of islet aggregates is essential for functional maturation. Our study provides new insights into understanding the fundamental self-organizing mechanism for islet aggregation, architecture, and functional maturation.
Collapse
Affiliation(s)
- Wilma Tixi
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Maricela Maldonado
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
- Department of Biomedical Engineering, College of Engineering, California State University, Long BeachLong BeachUnited States
| | - Ya-Ting Chang
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Amy Chiu
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Wilson Yeung
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Nazia Parveen
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Michael S Nelson
- Light Microscopy Core, Beckman Research Institute, City of HopeDuarteUnited States
| | - Ryan Hart
- Department of Neurobiology, Physiology and Behavior, University of California, DavisDavisUnited States
| | - Shihao Wang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British ColumbiaVancouverCanada
| | - Wu Jih Hsu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British ColumbiaVancouverCanada
| | - Patrick Fueger
- Department of Molecular & Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Janel L Kopp
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British ColumbiaVancouverCanada
| | - Mark O Huising
- Department of Neurobiology, Physiology and Behavior, University of California, DavisDavisUnited States
- Department of Physiology and Membrane Biology, School of Medicine, University of California, DavisDavisUnited States
| | - Sangeeta Dhawan
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| | - Hung Ping Shih
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of HopeDuarteUnited States
| |
Collapse
|
5
|
Gonzalez-Meljem JM, Ivins S, Andoniadou CL, Le Tissier P, Scambler P, Martinez-Barbera JP. An expression and function analysis of the CXCR4/SDF-1 signalling axis during pituitary gland development. PLoS One 2023; 18:e0280001. [PMID: 36800350 PMCID: PMC9937476 DOI: 10.1371/journal.pone.0280001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 12/19/2022] [Indexed: 02/18/2023] Open
Abstract
The chemokine SDF-1 (CXCL12) and its receptor CXCR4 control several processes during embryonic development such as the regulation of stem cell proliferation, differentiation, and migration. However, the role of this pathway in the formation of the pituitary gland is not understood. We sought to characterise the expression patterns of CXCR4, SDF-1 and CXCR7 at different stages of pituitary gland development. Our expression profiling revealed that SDF-1 is expressed in progenitor-rich regions of the pituitary anterior lobe, that CXCR4 and CXCR7 have opposite expression domains and that CXCR4 expression is conserved between mice and human embryos. We then assessed the importance of this signalling pathway in the development and function of the murine pituitary gland through conditional deletion of CXCR4 in embryonic pituitary progenitors. Successful and specific ablation of CXCR4 expression in embryonic pituitary progenitors did not lead to observable embryonic nor postnatal defects but allowed the identification of stromal CXCR4+ cells not derived from HESX1+ progenitors. Further analysis of constitutive SDF-1, CXCR7 and CXCR4 mutants of the pathway indicates that CXCR4 expression in HESX1+ cells and their descendants is not essential for normal pituitary development in mice.
Collapse
Affiliation(s)
- Jose Mario Gonzalez-Meljem
- Tecnologico de Monterrey, School of Engineering and Sciences, Mexico City, Mexico
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL-Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Sarah Ivins
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL-Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Cynthia Lilian Andoniadou
- Division of Craniofacial Development and Stem Cell Biology, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Paul Le Tissier
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter Scambler
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL-Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL-Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
6
|
Banerjee A, Biswas D, Barpanda A, Halder A, Sibal S, Kattimani R, Shah A, Mahadevan A, Goel A, Srivastava S. The First Pituitary Proteome Landscape From Matched Anterior and Posterior Lobes for a Better Understanding of the Pituitary Gland. Mol Cell Proteomics 2022; 22:100478. [PMID: 36470533 PMCID: PMC9877467 DOI: 10.1016/j.mcpro.2022.100478] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
To date, very few mass spectrometry (MS)-based proteomics studies are available on the anterior and posterior lobes of the pituitary. In the past, MS-based investigations have focused exclusively on the whole pituitary gland or anterior pituitary lobe. In this study, for the first time, we performed a deep MS-based analysis of five anterior and five posterior matched lobes to build the first lobe-specific pituitary proteome map, which documented 4090 proteins with isoforms, mostly mapped into chromosomes 1, 2, and 11. About 1446 differentially expressed significant proteins were identified, which were studied for lobe specificity, biological pathway enrichment, protein-protein interaction, regions specific to comparison of human brain and other neuroendocrine glands from Human Protein Atlas to identify pituitary-enriched proteins. Hormones specific to each lobe were also identified and validated with parallel reaction monitoring-based target verification. The study identified and validated hormones, growth hormone and thyroid-stimulating hormone subunit beta, exclusively to the anterior lobe whereas oxytocin-neurophysin 1 and arginine vasopressin to the posterior lobe. The study also identified proteins POU1F1 (pituitary-specific positive transcription factor 1), POMC (pro-opiomelanocortin), PCOLCE2 (procollagen C-endopeptidase enhancer 2), and NPTX2 (neuronal pentraxin-2) as pituitary-enriched proteins and was validated for their lobe specificity using parallel reaction monitoring. In addition, three uPE1 proteins, namely THEM6 (mesenchymal stem cell protein DSCD75), FSD1L (coiled-coil domain-containing protein 10), and METTL26 (methyltransferase-like 26), were identified using the NeXtProt database, and depicted tumor markers S100 proteins having high expression in the posterior lobe. In summary, the study documents the first matched anterior and posterior pituitary proteome map acting as a reference control for a better understanding of functional and nonfunctional pituitary adenomas and extrapolating the aim of the Human Proteome Project towards the investigation of the proteome of life.
Collapse
Affiliation(s)
- Arghya Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Deepatarup Biswas
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Abhilash Barpanda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Ankit Halder
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Shamira Sibal
- Lokmanya Tilak Municipal Medical College, Mumbai, India
| | | | - Abhidha Shah
- Department of Neurosurgery at King Edward Memorial Hospital and Seth G. S. Medical College, Mumbai, India
| | - Anita Mahadevan
- Human Brain Bank, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bangalore, India
| | - Atul Goel
- Department of Neurosurgery at King Edward Memorial Hospital and Seth G. S. Medical College, Mumbai, India
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India.
| |
Collapse
|
7
|
Pituitary Apoplexy: Risk Factors and Underlying Molecular Mechanisms. Int J Mol Sci 2022; 23:ijms23158721. [PMID: 35955859 PMCID: PMC9369054 DOI: 10.3390/ijms23158721] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/31/2022] [Accepted: 08/04/2022] [Indexed: 12/02/2022] Open
Abstract
Pituitary apoplexy is a rare syndrome, graded from asymptomatic subclinical apoplexy to a life-threatening condition due to pituitary ischemia or haemorrhage of an enlarged pituitary gland. The risk factors and the molecular underlying mechanisms are yet to be elucidated. We provide an overview of the general concepts, the potential factors associated with pituitary adenoma susceptibility for apoplectic events and the molecular mechanisms that could be involved such as HIF-1α/VEGF pathways and metalloproteinases activation, among others. The knowledge of the molecular mechanisms that could participate in the pathogenesis of pituitary apoplexy is crucial to advancement in the identification of future diagnostic tools and therapeutic targets in this rare but sometimes fatal condition.
Collapse
|
8
|
Ouyang Q, Hu S, Li L, Ran M, Zhu J, Zhao Y, Hu B, Hu J, He H, Li L, Wang J. Integrated mRNA and miRNA transcriptome analysis provides novel insights into the molecular mechanisms underlying goose pituitary development during the embryo-to-hatchling transition. Poult Sci 2021; 100:101380. [PMID: 34358958 PMCID: PMC8350522 DOI: 10.1016/j.psj.2021.101380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/01/2021] [Accepted: 07/04/2021] [Indexed: 11/25/2022] Open
Abstract
It is well established that the endocrine system plays a pivotal role in preparing the avian embryos for the abrupt switch from chorioallantoic to pulmonary respiration during the critical embryo-to-hatchling transition. However, as the master gland of the endocrine system, there has been little research focusing on the molecular mechanisms controlling the development and function of the pituitary gland during the peri-hatch period in birds. In the present study, we aimed to determine the genome-wide mRNA and miRNA transcriptome profiles of the pituitary during the embryo-to-hatchling transition period from embryonic day 22 (E22) to post-hatching day 6 (P6) in the goose (Anser cygnoides). Of note, expression of Anser_cygnoides_newGene_32456 and LOC106031011 were significantly different among these 4 stages (i.e., E22, E26, P2, and P6). Meanwhile, the neuroactive ligand-receptor interaction pathway was significantly enriched by the DEGs commonly identified among three pairwise comparisons. At the miRNA transcriptome level, there were not commonly identified DE miRNAs among these 4 stages, while the 418 of their predicted target genes were mutually shared. Both the target genes of DE miRNAs in each comparison and these 418 shared target genes were significantly enriched in the ECM-receptor interaction and focal adhesion pathways. In the predicted miRNA-mRNA interaction networks of these 2 pathways, novel_miRNA_467, novel_miRNA_154, and novel_miRNA_340 were the hub miRNAs. In addition, multiple DE miRNAs also showed predicted target relationships with the DEGs associated with extracellular matrix (ECM) components. Among them, expression of novel_miR_120, tgu-miR-92-3p, and novel_miR_398 was significantly negatively correlated with that of LAMC3 (laminin subunit gamma3), suggesting that these miRNAs may regulate pituitary tissue remodeling and functional changes through targeting LAMC3 during development. These identified DE mRNAs and miRNAs as well as their predicted interaction networks involved in regulation of tissue remodeling and cellular functions were most likely to play critical roles in facilitating the embryo-to-hatchling transition. These results provide novel insights into the early developmental process of avian pituitary gland and will help better understand the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Qingyuan Ouyang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Li Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Mingxia Ran
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Jiaran Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Yiting Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Bo Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| |
Collapse
|
9
|
Song G, Luo BH. Atypical structure and function of integrin α V β 8. J Cell Physiol 2020; 236:4874-4887. [PMID: 33368230 DOI: 10.1002/jcp.30242] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/10/2020] [Accepted: 12/12/2020] [Indexed: 12/12/2022]
Abstract
Integrins are heterodimeric transmembrane proteins that play important roles in various biological processes. Most integrins serve as adhesion molecules and transmit bidirectional signaling across the cell membrane through global conformational changes from the bent closed to the extended open conformation. However, integrin β8 is distinctive in structure and function. Its cytoplasmic domain lacks the conserved protein-binding sequence, which is important in transmitting inside-out signals, suggesting that integrin β8 may have a different activation mechanism or lack such signaling. In addition, the ligand-binding or activating metal ion Mn2+ does not induce a global conformational change in integrin β8 . It may have only one conformation, that is, an extended, closed conformation, but with high affinity for ligands under physiological conditions, and is, therefore, considered an atypical integrin member. The extended structure and high ligand-binding affinity of integrin αv β8 make it ideal for encountering and binding ligands expressed on an opposing cell or in the extracellular matrix. In this review, we summarize the progress in integrin β8 research with a focus on its distinctive function and structure among integrin members.
Collapse
Affiliation(s)
- Guannan Song
- Department of Life Science, University of Louisiana State University, Baton Rouge, Louisiana, USA
| | - Bing-Hao Luo
- Department of Life Science, University of Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
10
|
Lodge EJ, Xekouki P, Silva TS, Kochi C, Longui CA, Faucz FR, Santambrogio A, Mills JL, Pankratz N, Lane J, Sosnowska D, Hodgson T, Patist AL, Francis-West P, Helmbacher F, Stratakis CA, Andoniadou CL. Requirement of FAT and DCHS protocadherins during hypothalamic-pituitary development. JCI Insight 2020; 5. [PMID: 33108146 PMCID: PMC7714405 DOI: 10.1172/jci.insight.134310] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Pituitary developmental defects lead to partial or complete hormone deficiency and significant health problems. The majority of cases are sporadic and of unknown cause. We screened 28 patients with pituitary stalk interruption syndrome (PSIS) for mutations in the FAT/DCHS family of protocadherins that have high functional redundancy. We identified seven variants, four of which putatively damaging, in FAT2 and DCHS2 in six patients with pituitary developmental defects recruited through a cohort of patients with mostly ectopic posterior pituitary gland and/or pituitary stalk interruption. All patients had growth hormone deficiency and two presented with multiple hormone deficiencies and small glands. FAT2 and DCHS2 were strongly expressed in the mesenchyme surrounding the normal developing human pituitary. We analyzed Dchs2-/- mouse mutants and identified anterior pituitary hypoplasia and partially penetrant infundibular defects. Overlapping infundibular abnormalities and distinct anterior pituitary morphogenesis defects were observed in Fat4-/- and Dchs1-/- mouse mutants but all animal models displayed normal commitment to the anterior pituitary cell type. Together our data implicate FAT/DCHS protocadherins in normal hypothalamic-pituitary development and identify FAT2 and DCHS2 as candidates underlying pituitary gland developmental defects such as ectopic pituitary gland and/or pituitary stalk interruption.
Collapse
Affiliation(s)
- Emily J. Lodge
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
| | - Paraskevi Xekouki
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
| | - Tatiane S. Silva
- Pediatric Endocrinology Unit, Irmandade da Santa Casa de Misericórdia de São Paulo, São Paulo, Brazil
| | - Cristiane Kochi
- Pediatric Endocrinology Unit, Irmandade da Santa Casa de Misericórdia de São Paulo, São Paulo, Brazil
| | - Carlos A. Longui
- Pediatric Endocrinology Unit, Irmandade da Santa Casa de Misericórdia de São Paulo, São Paulo, Brazil
| | - Fabio R. Faucz
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Alice Santambrogio
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - James L. Mills
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - John Lane
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Dominika Sosnowska
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
| | - Tina Hodgson
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
| | - Amanda L. Patist
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
| | - Philippa Francis-West
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
| | | | - Constantine A. Stratakis
- Section on Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Cynthia L. Andoniadou
- Centre for Craniofacial & Regenerative Biology, King’s College London, Guy’s Campus, London, United Kingdom
- Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
11
|
Xiang X, Wang L, Zhou L, Chen Y, Xia H. Metformin upregulates the expression of Gli1 in vascular endothelial cells in hyperoxia-exposed neonatal mice. Am J Transl Res 2020; 12:6092-6106. [PMID: 33194016 PMCID: PMC7653632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 08/27/2020] [Indexed: 06/11/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is characterized by arrested alveolar and vascular development in premature infants. Metformin protects against the cardiovascular impairment induced by diabetes. The aim of this study was to investigate whether metformin could also enhance pulmonary vascular development in hyperoxic neonatal mice and investigate possible mechanisms involved. C57BL/6J newborn mice were randomly assigned to either of two groups - the room air group or the hyperoxia group - within 12 h postnatally. The mice were subcutaneously injected with metformin (100 mg/kg) or saline for 14 days. Lung morphology and PECAM-1 (CD31) expression in the lung were evaluated at postnatal days 7 and 14. Ki-67 and Gli1 expression in vascular endothelial cells was evaluated at postnatal day 14 by immunofluorescence staining. Flow cytometry (FCM) was also used to analyze Gli1 expression. Human umbilical vein endothelial cell (HUVECs) were used to investigate the role of metformin in vascular proliferation and tubular formation under 90% oxygen in vitro by cell counting Kti-8 (CCK8) assays and tube formation assays. Exposure to hyperoxia resulted in impaired lung development in newborn mice. Metformin enhanced the terminal airspace and radial alveolar count in newborn mice thus exposed. Immunohistochemistry staining and western blot assays revealed that metformin enhanced the expression of CD31 in hyperoxia-exposed newborn mice. Immunofluorescence staining showed that metformin enhanced the expression of Ki-67 in vascular endothelial cells. Furthermore, both immunofluorescence staining and FCM demonstrated that metformin increased Gli1 expression in vascular endothelial cells. Additionally, cell counting Kit-8 (CCK8) and viability assays of HUVECs in vitro both indicated that metformin improved the vascular proliferation and tube formation of HUVECs under 90% oxygen. These results indicated that metformin enhanced lung vascular development and upregulated the expression of Gli1 in the pulmonary vascular endothelial cells in hyperoxic neonatal mice.
Collapse
Affiliation(s)
- Xiaowen Xiang
- Department of Neonatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092, China
| | - Li Wang
- Department of Neonatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092, China
| | - Lin Zhou
- Department of Neonatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092, China
| | - Yanru Chen
- Department of Neonatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092, China
| | - Hongping Xia
- Department of Neonatology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200092, China
| |
Collapse
|
12
|
Basigin Associates with Integrin in Order to Regulate Perineurial Glia and Drosophila Nervous System Morphology. J Neurosci 2020; 40:3360-3373. [PMID: 32265259 DOI: 10.1523/jneurosci.1397-19.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 03/03/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022] Open
Abstract
The Drosophila nervous system is ensheathed by a layer of outer glial cells, the perineurial glia, and a specialized extracellular matrix, the neural lamella. The function of perineurial glial cells and how they interact with the extracellular matrix are just beginning to be elucidated. Integrin-based focal adhesion complexes link the glial membrane to the extracellular matrix, but little is known about integrin's regulators in the glia. The transmembrane Ig domain protein Basigin/CD147/EMMPRIN is highly expressed in the perineurial glia surrounding the Drosophila larval nervous system. Here we show that Basigin associates with integrin at the focal adhesions to uphold the structure of the glia-extracellular matrix sheath. Knockdown of Basigin in perineurial glia using RNAi results in significant shortening of the ventral nerve cord, compression of the glia and extracellular matrix in the peripheral nerves, and reduction in larval locomotion. We determined that Basigin is expressed in close proximity to integrin at the glial membrane, and that expression of the extracellular integrin-binding domain of Basigin is sufficient to rescue peripheral glial compression. We also found that a reduction in expression of integrin at the membrane rescues the ventral nerve cord shortening, peripheral glial compression, and locomotor phenotypes, and that reduction in the integrin-binding protein Talin can partially rescue glial compression. These results identify Basigin as a potential negative regulator of integrin in the glia, supporting proper glial and extracellular matrix ensheathment of the nervous system.SIGNIFICANCE STATEMENT The glial cells and extracellular matrix play important roles in supporting and protecting the nervous system, but the interactions between these components have not been well characterized. Our study identified expression of a conserved Ig superfamily protein, Basigin, at the glial membrane of Drosophila where it associates with the integrin-based focal adhesion complexes to ensure proper ensheathment of the CNS and PNS. Loss of Basigin in the glia results in an overall compression of the nervous system due to integrin dysregulation, which causes locomotor defects in the animals. This underlies the importance of glia-matrix communication for structural and functional support of the nervous system.
Collapse
|
13
|
Peng L, Fu C, Liang Z, Zhang Q, Xiong F, Chen L, He C, Wei Q. Pulsed Electromagnetic Fields Increase Angiogenesis and Improve Cardiac Function After Myocardial Ischemia in Mice. Circ J 2020; 84:186-193. [PMID: 31915323 DOI: 10.1253/circj.cj-19-0758] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Previous studies have shown that pulsed electromagnetic fields (PEMF) stimulate angiogenesis and may be a potential treatment strategy to improve cardiac function after myocardial infarction (MI). This study explored the effects and its related mechanisms of PEMF in MI mice. METHODS AND RESULTS MI mice were used in PEMF treatment (15 Hz 1.5 mT PEMF or 30 Hz 3.0 mT PEMF) for 45 min per day for 2 weeks. Furthermore, an in vivo Matrigel plug assay was used to observe the effect of PEMF in promoting angiogenesis. Compared with the sham PEMF group, PEMF treatment with 30 Hz 3.0 mT significantly improved heart function. PEMF treatment with 15 Hz 1.5 mT and 30 Hz 3.0 mT both increased capillary density, decreased infarction area size, increased the protein expression of vascular endothelial growth factor (VEGF), vascular endothelial growth factor receptor 2 (VEGFR2), Ser473-phosphorylated Akt (pSer473-Akt) and S1177-phosphorylated endothelial nitric oxide synthase (pS1177-eNOS), and increased the mRNA level of VEGF and hypoxia inducible factor 1-alpha (HIF-1α) in the infarct border zone. Additionally, treatment with 30 Hz 3.0 mT also increased protein and mRNA level of fibroblast growth factor 2 (FGF2), and protein level of β1 integrin, and shows a stronger therapeutic effect. CONCLUSIONS PEMF treatment could promote angiogenesis of the infarct border zone and improve cardiac function in MI mice. A treatment parameter of 30 Hz 3.0 mT is remarkably effective in MI mice. The effect is associated with the proangiogenic signaling pathways of HIF-1α/VEGF/Akt/eNOS or HIF-1α/FGF2/Akt/eNOS.
Collapse
Affiliation(s)
- Lihong Peng
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University
- Key Laboratory of Rehabilitation Medicine in Sichuan Province
| | - Chenying Fu
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University
| | - Zejun Liang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University
- Key Laboratory of Rehabilitation Medicine in Sichuan Province
| | - Qing Zhang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University
- Key Laboratory of Rehabilitation Medicine in Sichuan Province
| | - Feng Xiong
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University
- Key Laboratory of Rehabilitation Medicine in Sichuan Province
| | - Li Chen
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University
- Key Laboratory of Rehabilitation Medicine in Sichuan Province
| | - Chengqi He
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University
- Key Laboratory of Rehabilitation Medicine in Sichuan Province
| | - Quan Wei
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University
- Key Laboratory of Rehabilitation Medicine in Sichuan Province
| |
Collapse
|
14
|
Fletcher PA, Smiljanic K, Maso Prévide R, Iben JR, Li T, Rokic MB, Sherman A, Coon SL, Stojilkovic SS. Cell Type- and Sex-Dependent Transcriptome Profiles of Rat Anterior Pituitary Cells. Front Endocrinol (Lausanne) 2019; 10:623. [PMID: 31620083 PMCID: PMC6760010 DOI: 10.3389/fendo.2019.00623] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 08/28/2019] [Indexed: 01/14/2023] Open
Abstract
Understanding the physiology and pathology of an organ composed of a variety of cell populations depends critically on genome-wide information on each cell type. Here, we report single-cell transcriptome profiling of over 6,800 freshly dispersed anterior pituitary cells from postpubertal male and female rats. Six pituitary-specific cell types were identified based on known marker genes and characterized: folliculostellate cells and hormone-producing corticotrophs, gonadotrophs, thyrotrophs, somatotrophs, and lactotrophs. Also identified were endothelial and blood cells from the pituitary capillary network. The expression of numerous developmental and neuroendocrine marker genes in both folliculostellate and hormone-producing cells supports that they have a common origin. For several genes, the validity of transcriptome analysis was confirmed by qRT-PCR and single cell immunocytochemistry. Folliculostellate cells exhibit impressive transcriptome diversity, indicating their major roles in production of endogenous ligands and detoxification enzymes, and organization of extracellular matrix. Transcriptome profiles of hormone-producing cells also indicate contributions toward those functions, while also clearly demonstrating their endocrine function. This survey highlights many novel genetic markers contributing to pituitary cell type identity, sexual dimorphism, and function, and points to relationships between hormone-producing and folliculostellate cells.
Collapse
Affiliation(s)
- Patrick A. Fletcher
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Kosara Smiljanic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Rafael Maso Prévide
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - James R. Iben
- Molecular Genomics Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Tianwei Li
- Molecular Genomics Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Milos B. Rokic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Arthur Sherman
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Steven L. Coon
- Molecular Genomics Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Stanko S. Stojilkovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
15
|
Alfano D, Altomonte A, Cortes C, Bilio M, Kelly RG, Baldini A. Tbx1 regulates extracellular matrix-cell interactions in the second heart field. Hum Mol Genet 2019; 28:2295-2308. [DOI: 10.1093/hmg/ddz058] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/14/2019] [Accepted: 03/14/2019] [Indexed: 12/31/2022] Open
Abstract
Abstract
Tbx1, the major candidate gene for DiGeorge or 22q11.2 deletion syndrome, is required for efficient incorporation of cardiac progenitors of the second heart field (SHF) into the heart. However, the mechanisms by which TBX1 regulates this process are still unclear. Here, we have used two independent models, mouse embryos and cultured cells, to define the role of TBX1 in establishing morphological and dynamic characteristics of SHF in the mouse. We found that loss of TBX1 impairs extracellular matrix (ECM)-integrin-focal adhesion (FA) signaling in both models. Mosaic analysis in embryos suggested that this function is non-cell autonomous, and, in cultured cells, loss of TBX1 impairs cell migration and FAs. Additionally, we found that ECM-mediated integrin signaling is disrupted upon loss of TBX1. Finally, we show that interfering with the ECM-integrin-FA axis between E8.5 and E9.5 in mouse embryos, corresponding to the time window within which TBX1 is required in the SHF, causes outflow tract dysmorphogenesis. Our results demonstrate that TBX1 is required to maintain the integrity of ECM-cell interactions in the SHF and that this interaction is critical for cardiac outflow tract development. More broadly, our data identifies a novel TBX1 downstream pathway as an important player in SHF tissue architecture and cardiac morphogenesis.
Collapse
Affiliation(s)
- Daniela Alfano
- CNR–Institute of Genetics and Biophysics Adriano Buzzati-Traverso, Via Pietro Castellino, Naples, Italy
| | - Alessandra Altomonte
- CNR–Institute of Genetics and Biophysics Adriano Buzzati-Traverso, Via Pietro Castellino, Naples, Italy
| | - Claudio Cortes
- Aix-Marseille Université, CNRS UMR, IBDM, Marseille, France
| | - Marchesa Bilio
- CNR–Institute of Genetics and Biophysics Adriano Buzzati-Traverso, Via Pietro Castellino, Naples, Italy
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR, IBDM, Marseille, France
| | - Antonio Baldini
- CNR–Institute of Genetics and Biophysics Adriano Buzzati-Traverso, Via Pietro Castellino, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
16
|
Azuma M, Tsukada T, Inagaki T, Casmad F, Jindatip D, Tofrizal A, Maliza R, Batchuluun K, Syaidah R, Ohno N, Fujiwara K, Kikuchi M, Yashiro T. Immunohistochemical Study of the Laminin α5 Chain and Its Specific Receptor, Basal Cell Adhesion Molecule (BCAM), in both Fetal and Adult Rat Pituitary Glands. Acta Histochem Cytochem 2018; 51:145-152. [PMID: 30510328 PMCID: PMC6261841 DOI: 10.1267/ahc.18014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 08/10/2018] [Indexed: 01/01/2023] Open
Abstract
Laminin, a major basement membrane protein, comprises three subunit chains: α, β, and γ chains. Among these chains, only the laminin α chain is capable of signaling via laminin receptors. Although laminin isoforms containing the α5 chain were reported to be the first laminin produced during rat anterior pituitary gland development, the functions of these isoforms are unknown. We used immunohistochemical techniques to localize the laminin α5 chain and its specific receptor, basal cell adhesion molecule (BCAM), in fetal and adult pituitary gland. Laminin α5 chain immunoreactivity was observed in the basement membrane of the primordial adenohypophysis at embryonic days 12.5 to 19.5. Double immunostaining showed that BCAM was present and co-localized with the laminin α5 chain in the tissue. Quantitative analysis showed that the laminin α5 chain and BCAM were expressed in the anterior pituitary gland during postnatal development and in adulthood (postnatal day 60). In the adult gland, co-localization of the laminin α5 chain and BCAM was observed, and BCAM was detected in both the folliculo-stellate cells and endothelial cells. These results suggest that laminin α5 chain signaling via BCAM occurs in both the fetal adenohypophysis and adult anterior pituitary gland.
Collapse
Affiliation(s)
- Morio Azuma
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine
| | - Takehiro Tsukada
- Department of Biomolecular Science, Faculty of Science, Toho University
| | - Takeshi Inagaki
- Division of Forensic Medicine, Department of Anatomy, Jichi Medical University School of Medicine
| | - Fujianti Casmad
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine
| | - Depicha Jindatip
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine
- Department of Anatomy, Faculty of Medicine, Chulalongkorn University
| | - Alimuddin Tofrizal
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine
| | - Rita Maliza
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine
| | - Khongorzul Batchuluun
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine
| | - Rahimi Syaidah
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine
| | - Nobuhiko Ohno
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine
| | - Ken Fujiwara
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine
| | - Motoshi Kikuchi
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine
- Laboratory of Natural History, Jichi Medical University School of Medicine
| | - Takashi Yashiro
- Division of Histology and Cell Biology, Department of Anatomy, Jichi Medical University School of Medicine
| |
Collapse
|
17
|
Oh JY, Suh HN, Choi GE, Lee HJ, Jung YH, Ko SH, Kim JS, Chae CW, Lee CK, Han HJ. Modulation of sonic hedgehog-induced mouse embryonic stem cell behaviours through E-cadherin expression and integrin β1-dependent F-actin formation. Br J Pharmacol 2018; 175:3548-3562. [PMID: 29933500 DOI: 10.1111/bph.14423] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 05/23/2018] [Accepted: 06/12/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE The sonic hedgehog pathway (Shh) plays a central role in maintaining stem cell function and behaviour in various processes related to self-renewal and tissue regeneration. However, the therapeutic effect of Shh on mouse embryonic stem cells (mESCs) has not yet been clearly elucidated. Thus, we investigated the effect of Shh on the regulation of mESC behaviour as well as the effect of Shh-pretreated mESCs in skin wound healing. EXPERIMENTAL APPROACH The underlying mechanisms of Shh signalling pathway in growth and motility of mESCs were investigated using Western blot analysis, a cell proliferation assay and cell migration assay. In addition, the effect of Shh-pretreated mESCs in skin wound healing was determined using a mouse excisional wound splinting model. KEY RESULTS Shh disrupted the adherens junction through proteolysis by activating MMPs. In addition, the release of β-catenin from adherens junctions mediated by Shh led to cell cycle-dependent mESC proliferation. Shh-mediated Gli1 expression led to integrin β1 up-regulation, followed by FAK and Src phosphorylation. Furthermore, among the Rho-GTPases, Rac1 and Cdc42 were activated in a Shh-dependent manner while F-actin expression was suppressed by Rac1 and Cdc42 siRNA transfection. Consistent with the in vitro results, the skin wound healing assay revealed that Shh-treated mESCs increased angiogenesis and skin wound repair compared to that in Shh-treated mESCs transfected with integrin β1 siRNA in vivo. CONCLUSIONS AND IMPLICATIONS Our results imply that Shh induces adherens junction disruption and integrin β1-dependent F-actin formation by a mechanism involving FAK/Src and Rac1/Cdc42 signalling pathways in mESCs.
Collapse
Affiliation(s)
- Ji Young Oh
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea.,Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| | - Han Na Suh
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea.,Minipig Model Group, Animal Model Center, Korea Institute of Toxicology, Jeongeup-si, Jeollabuk-do, Korea
| | - Gee Euhn Choi
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| | - Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| | - So Hee Ko
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| | - Jun Sung Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| | - Chang Woo Chae
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| | - Chang-Kyu Lee
- Department of Agricultural Biotechnology, Animal Biotechnology Major, and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Korea.,Institute of Green Bio Science and Technology, Seoul National University, Pyeong Chang, Kangwon do, Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 PLUS Program for Creative Veterinary Science Research, Seoul National University, Seoul, Korea
| |
Collapse
|