1
|
Pun R, Thapa A, Takafuji SR, Suzuki RM, Kay GF, Howard TD, Kim MH, North BJ. BubR1 Controls Heart Development by Promoting Expression of Cardiogenesis Regulators. J Am Heart Assoc 2025; 14:e038286. [PMID: 40055864 DOI: 10.1161/jaha.124.038286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 01/22/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Congenital heart defects are structural anomalies present at birth that can affect the function of the heart. Aneuploidy is a significant risk factor for congenital heart defects. Mosaic variegated aneuploidy syndrome, caused by mutations in Bub1b (encoding BubR1, a mitotic checkpoint protein), leads to congenital heart defects such as septal defects. However, the molecular rationale for how Bub1b mutations promote congenital heart defects associated with mosaic variegated aneuploidy syndrome remains unresolved. METHODS To study morphological, structural, and cellular consequences of BubR1 deletion in the heart, we crossed mice carrying conditional alleles of Bub1b with Nkx2.5-cre mice. Single-cell RNA sequencing was carried out to determine differentially expressed genes and biological processes in various cell types present in the developing heart. Trajectory analysis was carried out to determine the differentiation trajectory of BubR1 knockout embryonic hearts. Finally, CellChat analysis provided details on the major signaling interactions that were either absent or hyperactive in the BubR1 knockout heart. RESULTS Here, we show that cardiac-specific BubR1 deletion causes embryonic lethality due to developmental stalling after cardiac looping with defects in cardiac maturation including chamber wall thickness, septation, and trabeculation. Single-cell transcriptomic profiling further revealed that the differentiation trajectory of cardiomyocytes is severely impacted with suppression of critical cardiogenesis genes. Hyperactivation of Wnt signaling in BubR1 knockout hearts indicated a disturbed homeostasis in cellular pathways essential for proper tissue morphogenesis of the heart. CONCLUSIONS Taken together, these findings reveal that BubR1 is a crucial regulator of cardiac development in vivo, which ensures the proper timing of heart morphogenesis.
Collapse
Affiliation(s)
- Renju Pun
- Biomedical Sciences Department Creighton University School of Medicine Omaha NE USA
| | - Aradhana Thapa
- Biomedical Sciences Department Creighton University School of Medicine Omaha NE USA
| | - Sylar R Takafuji
- Biomedical Sciences Department Creighton University School of Medicine Omaha NE USA
| | - Rexton M Suzuki
- Biomedical Sciences Department Creighton University School of Medicine Omaha NE USA
| | - Gabrielle F Kay
- Biomedical Sciences Department Creighton University School of Medicine Omaha NE USA
| | - Toni D Howard
- Biomedical Sciences Department Creighton University School of Medicine Omaha NE USA
| | - Michael H Kim
- CHI Heart Institute and Department of Medicine Creighton University School of Medicine Omaha NE USA
| | - Brian J North
- Biomedical Sciences Department Creighton University School of Medicine Omaha NE USA
| |
Collapse
|
2
|
Jara O, Maripillán J, Momboisse F, Cárdenas AM, García IE, Martínez AD. Differential Regulation of Hemichannels and Gap Junction Channels by RhoA GTPase and Actin Cytoskeleton: A Comparative Analysis of Cx43 and Cx26. Int J Mol Sci 2024; 25:7246. [PMID: 39000353 PMCID: PMC11242593 DOI: 10.3390/ijms25137246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Connexins (Cxs) are transmembrane proteins that assemble into gap junction channels (GJCs) and hemichannels (HCs). Previous researches support the involvement of Rho GTPases and actin microfilaments in the trafficking of Cxs, formation of GJCs plaques, and regulation of channel activity. Nonetheless, it remains uncertain whether distinct types of Cxs HCs and GJCs respond differently to Rho GTPases or changes in actin polymerization/depolymerization dynamics. Our investigation revealed that inhibiting RhoA, a small GTPase that controls actin polymerization, or disrupting actin microfilaments with cytochalasin B (Cyto-B), resulted in reduced GJCs plaque size at appositional membranes and increased transport of HCs to non-appositional plasma membrane regions. Notably, these effects were consistent across different Cx types, since Cx26 and Cx43 exhibited similar responses, despite having distinct trafficking routes to the plasma membrane. Functional assessments showed that RhoA inhibition and actin depolymerization decreased the activity of Cx43 GJCs while significantly increasing HC activity. However, the functional status of GJCs and HCs composed of Cx26 remained unaffected. These results support the hypothesis that RhoA, through its control of the actin cytoskeleton, facilitates the transport of HCs to appositional cell membranes for GJCs formation while simultaneously limiting the positioning of free HCs at non-appositional cell membranes, independently of Cx type. This dynamic regulation promotes intercellular communications and reduces non-selective plasma membrane permeability through a Cx-type dependent mechanism, whereby the activity of Cx43 HCs and GJCs are differentially affected but Cx26 channels remain unchanged.
Collapse
Affiliation(s)
- Oscar Jara
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia, Universidad de Valparaíso, Valparaíso 2362807, Chile
- Department of Pediatrics, University of Chicago, Chicago, IL 60637, USA
| | - Jaime Maripillán
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia, Universidad de Valparaíso, Valparaíso 2362807, Chile
| | - Fanny Momboisse
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia, Universidad de Valparaíso, Valparaíso 2362807, Chile
- Virus and Immunity Unit, Institut Pasteur, Université Paris Cité, CNRS UMR3569, 75013 Paris, France
| | - Ana María Cárdenas
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia, Universidad de Valparaíso, Valparaíso 2362807, Chile
| | - Isaac E García
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia, Universidad de Valparaíso, Valparaíso 2362807, Chile
- Laboratorio de Fisiología Molecular y Biofísica, Facultad de Odontología, Universidad de Valparaíso, Valparaíso 2360004, Chile
- Centro de Investigación en Ciencias Odontológicas y Médicas, CICOM, Universidad de Valparaíso, Valparaíso 2360004, Chile
| | - Agustín D Martínez
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencia, Universidad de Valparaíso, Valparaíso 2362807, Chile
| |
Collapse
|
3
|
Badaoui M, Chanson M. Intercellular Communication in Airway Epithelial Cell Regeneration: Potential Roles of Connexins and Pannexins. Int J Mol Sci 2023; 24:16160. [PMID: 38003349 PMCID: PMC10671439 DOI: 10.3390/ijms242216160] [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: 09/26/2023] [Revised: 10/19/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Connexins and pannexins are transmembrane proteins that can form direct (gap junctions) or indirect (connexons, pannexons) intercellular communication channels. By propagating ions, metabolites, sugars, nucleotides, miRNAs, and/or second messengers, they participate in a variety of physiological functions, such as tissue homeostasis and host defense. There is solid evidence supporting a role for intercellular signaling in various pulmonary inflammatory diseases where alteration of connexin/pannexin channel functional expression occurs, thus leading to abnormal intercellular communication pathways and contributing to pathophysiological aspects, such as innate immune defense and remodeling. The integrity of the airway epithelium, which is the first line of defense against invading microbes, is established and maintained by a repair mechanism that involves processes such as proliferation, migration, and differentiation. Here, we briefly summarize current knowledge on the contribution of connexins and pannexins to necessary processes of tissue repair and speculate on their possible involvement in the shaping of the airway epithelium integrity.
Collapse
Affiliation(s)
| | - Marc Chanson
- Department of Cell Physiology & Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland;
| |
Collapse
|
4
|
Moazzen H, Bolaji MD, Leube RE. Desmosomes in Cell Fate Determination: From Cardiogenesis to Cardiomyopathy. Cells 2023; 12:2122. [PMID: 37681854 PMCID: PMC10487268 DOI: 10.3390/cells12172122] [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: 07/18/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Abstract
Desmosomes play a vital role in providing structural integrity to tissues that experience significant mechanical tension, including the heart. Deficiencies in desmosomal proteins lead to the development of arrhythmogenic cardiomyopathy (AC). The limited availability of preventative measures in clinical settings underscores the pressing need to gain a comprehensive understanding of desmosomal proteins not only in cardiomyocytes but also in non-myocyte residents of the heart, as they actively contribute to the progression of cardiomyopathy. This review focuses specifically on the impact of desmosome deficiency on epi- and endocardial cells. We highlight the intricate cross-talk between desmosomal proteins mutations and signaling pathways involved in the regulation of epicardial cell fate transition. We further emphasize that the consequences of desmosome deficiency differ between the embryonic and adult heart leading to enhanced erythropoiesis during heart development and enhanced fibrogenesis in the mature heart. We suggest that triggering epi-/endocardial cells and fibroblasts that are in different "states" involve the same pathways but lead to different pathological outcomes. Understanding the details of the different responses must be considered when developing interventions and therapeutic strategies.
Collapse
Affiliation(s)
- Hoda Moazzen
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany; (M.D.B.); (R.E.L.)
| | | | | |
Collapse
|
5
|
Millesi F, Mero S, Semmler L, Rad A, Stadlmayr S, Borger A, Supper P, Haertinger M, Ploszczanski L, Windberger U, Weiss T, Naghilou A, Radtke C. Systematic Comparison of Commercial Hydrogels Revealed That a Synergy of Laminin and Strain-Stiffening Promotes Directed Migration of Neural Cells. ACS APPLIED MATERIALS & INTERFACES 2023; 15:12678-12695. [PMID: 36876876 PMCID: PMC10020957 DOI: 10.1021/acsami.2c20040] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/23/2023] [Indexed: 05/19/2023]
Abstract
Hydrogels have shown potential in replacing damaged nerve tissue, but the ideal hydrogel is yet to be found. In this study, various commercially available hydrogels were compared. Schwann cells, fibroblasts, and dorsal root ganglia neurons were seeded on the hydrogels, and their morphology, viability, proliferation, and migration were examined. Additionally, detailed analyses of the gels' rheological properties and topography were conducted. Our results demonstrate vast differences on cell elongation and directed migration on the hydrogels. Laminin was identified as the driver behind cell elongation and in combination with a porous, fibrous, and strain-stiffening matrix structure responsible for oriented cell motility. This study improves our understanding of cell-matrix interactions and thereby facilitates tailored fabrication of hydrogels in the future.
Collapse
Affiliation(s)
- Flavia Millesi
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Sascha Mero
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Lorenz Semmler
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Anda Rad
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Sarah Stadlmayr
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Anton Borger
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Paul Supper
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Maximilian Haertinger
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Leon Ploszczanski
- Institute
for Physics and Materials Science, University
of Natural Resources and Life Sciences, Vienna 1190, Austria
| | - Ursula Windberger
- Decentralized
Biomedical Facilities, Core Unit Laboratory Animal Breeding and Husbandry, Medical University Vienna, Vienna 1090, Austria
| | - Tamara Weiss
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
| | - Aida Naghilou
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
- Department
of Physical Chemistry, University of Vienna, Vienna 1090, Austria
| | - Christine Radtke
- Research
Laboratory of the Department of Plastic, Reconstructive and Aesthetic
Surgery, Medical University of Vienna, Vienna 1090, Austria
- Austrian
Cluster for Tissue Regeneration, Vienna 1200, Austria
- Department
of Plastic, Reconstructive
and Aesthetic Surgery, Medical University
of Vienna, Vienna 1090, Austria
| |
Collapse
|
6
|
A new host-targeted antiviral cyclolignan (SAU-22.107) for Dengue Virus infection in cell cultures. Potential action mechanisms based on cell imaging. Virus Res 2023; 323:198995. [PMID: 36336130 DOI: 10.1016/j.virusres.2022.198995] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/31/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Dengue virus (DENV) infection is the most arbovirosis in the world. However, medications have not been approved for its treatment. Drug discovery based on the host-targeted antiviral (HTA) constitutes a new promising strategy, considering their high genetic barrier to resistance and the low probability of selecting drug resistance strains. In this study, we have tested fifty-seven podophyllotoxin-related cyclolignans on DENV-2 infected cells and found the most promising compound was S.71. Using cellular and molecular biology experiments, we have discovered that the new lignan altered the distribution of microtubules, induced changes in cell morphology, and caused retraction of the rough endoplasmic reticulum. In addition, the compound alters the viral envelope protein and the double-stranded RNA, while there is a decrease in negative-strand RNA synthesis; especially when the compound was added between 6- and 12-hours post-infection. Altogether, S.71 decreases the viral yield through an HTA-related mechanism of action, possibly altering the DENV genome replication and/or polyprotein translation, through the alteration of microtubule distribution and endoplasmic reticulum deterioration. Finally, pharmacokinetic predictors show that S.71 falls within the standard ranges established for drugs.
Collapse
|
7
|
Cruz Del Puerto M, Rojas ML, Racca AC, Kourdova LT, Miranda AL, Panzetta-Dutari G, Genti-Raimondi S, Flores-Martín JB. StarD7 deficiency hinders cell motility through p-ERK1/2/Cx43 reduction. PLoS One 2022; 17:e0279912. [PMID: 36584213 PMCID: PMC9803278 DOI: 10.1371/journal.pone.0279912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/18/2022] [Indexed: 01/01/2023] Open
Abstract
StarD7 belongs to START protein family involved in lipid traffic, metabolism, and signaling events. Its precursor, StarD7.I which is important for mitochondrial homeostasis, is processed to the StarD7.II isoform that lacks the mitochondrial targeting sequence and is mainly released to the cytosol. StarD7 knockdown interferes with cell migration by an unknown mechanism. Here, we demonstrate that StarD7 silencing decreased connexin 43 (Cx43), integrin β1, and p-ERK1/2 expression in the non-tumoral migratory HTR-8/SVneo cells. StarD7-deficient cells exhibited Golgi disruption and reduced competence to reorient the microtubule-organizing center. The migratory capacity of StarD7-silenced cells was reestablished when Cx43 level was resettled, while p-ERK1/2 expression remained low. Importantly, ectopic expression of the StarD7.II isoform not only restored cell migration but also ERK1/2, Cx43, and integrin β1 expression. Thus, StarD7 is implicated in cell migration through an ERK1/2/Cx43 dependent mechanism but independent of the StarD7.I function in the mitochondria.
Collapse
Affiliation(s)
- Mariano Cruz Del Puerto
- Departamento de Bioquímica Clínica, Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Ciudad Universitaria, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, Córdoba, Argentina
| | - María Laura Rojas
- Departamento de Bioquímica Clínica, Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Ciudad Universitaria, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, Córdoba, Argentina
| | - Ana Cristina Racca
- Departamento de Bioquímica Clínica, Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Ciudad Universitaria, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, Córdoba, Argentina
| | - Lucille Tihomirova Kourdova
- Departamento de Bioquímica Clínica, Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Ciudad Universitaria, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, Córdoba, Argentina
| | - Andrea Lis Miranda
- Departamento de Bioquímica Clínica, Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Ciudad Universitaria, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, Córdoba, Argentina
| | - Graciela Panzetta-Dutari
- Departamento de Bioquímica Clínica, Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Ciudad Universitaria, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, Córdoba, Argentina
| | - Susana Genti-Raimondi
- Departamento de Bioquímica Clínica, Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Ciudad Universitaria, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, Córdoba, Argentina
| | - Jésica Belén Flores-Martín
- Departamento de Bioquímica Clínica, Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Ciudad Universitaria, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, Córdoba, Argentina
- * E-mail:
| |
Collapse
|
8
|
Peng Y, Ye J, Xu Y, Huang J, Wu Y, Liu W, Bai K, Chen S, Lu Y. Two genetic variants in NEXN and ABCC6 genes found in a patient with right coronary artery to right ventricle fistula combined with giant coronary aneurysm and patent ductus arteriosus. Front Cardiovasc Med 2022; 9:1048795. [DOI: 10.3389/fcvm.2022.1048795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/28/2022] [Indexed: 11/18/2022] Open
Abstract
ObjectiveCoronary artery fistula, defined as communication between a coronary artery and a great vessel or a cardiac chamber, is a relatively rare anomaly with an estimated incidence of 0.002% in the general population. It could be combined with a giant coronary artery aneurysm, with an incidence of 5.9% of the total incidence rate of CAF in the general population. The pathogenesis of these two combined anomalies is not clear, and we aimed to detect whether genetic abnormalities underlie the pathogenesis of these rarely combined anomalies.Materials and methodsA 6-year-old patient with a diagnosis of the right coronary artery to right ventricle fistula combined with a giant right coronary artery aneurysm and patent ductus arteriosus underwent a surgical repair at our center. The diagnosis was confirmed by echocardiography, CT, and surgery. DNA was extracted from the peripheral venous blood samples of the patient and his mother after informed consent was obtained. Hematoxylin and Eosin (HE) and Alizarin red staining were performed on the excised coronary artery aneurysm. Exome sequencing and in silico analyses were performed to detect detrimental genetic variants.ResultsNo obvious abnormalities were found in the excised coronary artery aneurysm. A heterozygous truncated variant (NM_144573: c.G298T; p.G100X) in the NEXN gene and a missense variant (NM_001171: c.G1312A; p.V438M) in the ABCC6 gene were carried by the patient but not by his mother.ConclusionThe NEXN-truncated variant, NEXN-G100X, is associated with the development of coronary arteries and congenital coronary artery anomalies.
Collapse
|
9
|
Jang DG, Kwon KY, Kweon YC, Kim BG, Myung K, Lee HS, Young Park C, Kwon T, Park TJ. GJA1 depletion causes ciliary defects by affecting Rab11 trafficking to the ciliary base. eLife 2022; 11:81016. [PMID: 36004726 PMCID: PMC9448326 DOI: 10.7554/elife.81016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
The gap junction complex functions as a transport channel across the membrane. Among gap junction subunits, gap junction protein α1 (GJA1) is the most commonly expressed subunit. A recent study showed that GJA1 is necessary for the maintenance of motile cilia; however, the molecular mechanism and function of GJA1 in ciliogenesis remain unknown. Here, we examined the functions of GJA1 during ciliogenesis in human retinal pigment epithelium-1 and Xenopus laevis embryonic multiciliated-cells. GJA1 localizes to the motile ciliary axonemes or pericentriolar regions beneath the primary cilium. GJA1 depletion caused malformation of both the primary cilium and motile cilia. Further study revealed that GJA1 depletion affected several ciliary proteins such as BBS4, CP110, and Rab11 in the pericentriolar region and basal body. Interestingly, CP110 removal from the mother centriole was significantly reduced by GJA1 depletion. Importantly, Rab11, a key regulator during ciliogenesis, was immunoprecipitated with GJA1, and GJA1 knockdown caused the mislocalization of Rab11. These findings suggest that GJA1 regulates ciliogenesis by interacting with the Rab11-Rab8 ciliary trafficking pathway.
Collapse
Affiliation(s)
- Dong Gil Jang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Keun Yeong Kwon
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Yeong Cheon Kweon
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Byung-Gyu Kim
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Republic of Korea
| | - Hyun-Shik Lee
- School of Life Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Chan Young Park
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Taejoon Kwon
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Tae Joo Park
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| |
Collapse
|
10
|
The Role of Connexin in Ophthalmic Neovascularization and the Interaction between Connexin and Proangiogenic Factors. J Ophthalmol 2022; 2022:8105229. [PMID: 35783340 PMCID: PMC9242797 DOI: 10.1155/2022/8105229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 06/11/2022] [Indexed: 12/02/2022] Open
Abstract
The formation of new blood vessels is an important physiological process that occurs during development. When the body is injured, new blood vessel formation helps the body recuperate by supplying more oxygen and nutrients. However, this mechanism can have a negative effect. In ophthalmologic diseases, such as corneal new blood vessels, neonatal vascular glaucoma, and diabetes retinopathy, the formation of new blood vessels has become a critical component in patient survival. Connexin is a protein that regulates the cellular and molecular material carried by cells. It has been demonstrated that it is widely expressed in vascular endothelial cells, where it forms a slit connection between adjacent cells to promote cell-cell communication via hemichannels, as well as substance exchange into intracellular environments. Numerous studies have demonstrated that connexin in vascular endothelial cells plays an important role in angiogenesis and vascular leakage. The purpose of this study was to investigate the effect between the angiogenesis-associated factor and the connexin. It also reveals the effect of connexin on ophthalmic neovascularization.
Collapse
|
11
|
Beth Payne L, Tewari BP, Dunkenberger L, Bond S, Savelli A, Darden J, Zhao H, Willi C, Kanodia R, Gude R, Powell MD, Oestreich KJ, Sontheimer H, Dal-Pra S, Chappell JC. Pericyte Progenitor Coupling to the Emerging Endothelium During Vasculogenesis via Connexin 43. Arterioscler Thromb Vasc Biol 2022; 42:e96-e114. [PMID: 35139658 PMCID: PMC8957572 DOI: 10.1161/atvbaha.121.317324] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/24/2022] [Indexed: 01/23/2023]
Abstract
BACKGROUND Vascular pericytes stabilize blood vessels and contribute to their maturation, while playing other key roles in microvascular function. Nevertheless, relatively little is known about involvement of their precursors in the earliest stages of vascular development, specifically during vasculogenesis. METHODS We combined high-power, time-lapse imaging with transcriptional profiling of emerging pericytes and endothelial cells in reporter mouse and cell lines. We also analyzed conditional transgenic animals deficient in Cx43/Gja1 (connexin 43/gap junction alpha-1) expression within Ng2+ cells. RESULTS A subset of Ng2-DsRed+ cells, likely pericyte/mural cell precursors, arose alongside endothelial cell differentiation and organization and physically engaged vasculogenic endothelium in vivo and in vitro. We found no overlap between this population of differentiating pericyte/mural progenitors and other lineages including hemangiogenic and neuronal/glial cell types. We also observed cell-cell coupling and identified Cx43-based gap junctions contributing to pericyte-endothelial cell precursor communication during vascular assembly. Genetic loss of Cx43/Gja1 in Ng2+ pericyte progenitors compromised embryonic blood vessel formation in a subset of animals, while surviving mutants displayed little-to-no vessel abnormalities, suggesting a resilience to Cx43/Gja1 loss in Ng2+ cells or potential compensation by additional connexin isoforms. CONCLUSIONS Together, our data suggest that a distinct pericyte lineage emerges alongside vasculogenesis and directly communicates with the nascent endothelium via Cx43 during early vessel formation. Cx43/Gja1 loss in pericyte/mural cell progenitors can induce embryonic vessel dysmorphogenesis, but alternate connexin isoforms may be able to compensate. These data provide insight that may reshape the current framework of vascular development and may also inform tissue revascularization/vascularization strategies.
Collapse
Affiliation(s)
- Laura Beth Payne
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
| | - Bhanu P. Tewari
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903, USA
| | - Logan Dunkenberger
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
| | - Samantha Bond
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
| | - Alyssa Savelli
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Jordan Darden
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
- Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Blacksburg, VA 24061, USA
| | - Huaning Zhao
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA
| | - Caroline Willi
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
| | - Ronak Kanodia
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
| | - Rosalie Gude
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
| | - Michael D. Powell
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kenneth J. Oestreich
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Harald Sontheimer
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903, USA
| | - Sophie Dal-Pra
- Division of Cardiovascular Medicine and Mandel Center for Hypertension Research and Division of Cardiovascular Medicine, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - John C. Chappell
- Center for Vascular and Heart Research, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
12
|
Han H, Zhao X, Liao M, Song Y, You C, Dong X, Yang X, Wang X, Huang B, Du M, Yan H. Activated Blood Coagulation Factor X (FXa) Contributes to the Development of Traumatic PVR Through Promoting RPE Epithelial-Mesenchymal Transition. Invest Ophthalmol Vis Sci 2021; 62:29. [PMID: 34283209 PMCID: PMC8300057 DOI: 10.1167/iovs.62.9.29] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Purpose Uncontrolled coagulation reactions contribute to pathological fibroproliferation in several organs, and yet their role in proliferative vitreoretinopathy (PVR) remains to be elucidated. In this study, we evaluated the profibrotic effects of FXa in RPE cells and in a mouse model of PVR. Methods FXa levels in the eyes of traumatic PVR patients and rabbit models of mechanical ocular trauma was measured by ELISA and immunohistochemistry. FXa-induced RPE EMT was assessed by examining cell proliferation, migration, tight junction changes, and expression of fibrotic markers. For in vivo study, FXa was injected into dispase-injured eyes, then intraocular fibrosis was evaluated by histological analysis and Western blotting. The therapeutic effect of FXa inhibitor was also examined in PVR mouse models. Results Vitreous FXa were higher in patients with traumatic PVR compared to patients with macular hole. Moreover, expressions of FXa and PAR1 were found in the epiretinal membranes from traumatic PVR patients. Vitreous FXa were markedly increased after mechanical ocular trauma in rabbits. In vitro, FXa stimulated RPE EMT characterized as ZO-1 disruption, compromised cell polarity, and increased fibronectin expressions. Co-injection of FXa and dispase in mice induced more severely damaged retinal structures, and increased α-SMA expressions than FXa or dispase treatment alone. Oral FXa or thrombin inhibitors significantly blocked intraocular fibrosis in PVR mouse models. FXa promoted phospho-activation of p38 in ARPE19 cells, which was dependent on PAR1. Moreover, TGF-βR inhibitor also significantly alleviated FXa-induced intraocular fibrosis in mice. Conclusions FXa promotes intraocular fibrosis in mice via mechanisms involving RPE activation.
Collapse
Affiliation(s)
- Han Han
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiao Zhao
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Mengyu Liao
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yinting Song
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Caiyun You
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xue Dong
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Molecular Ophthalmology, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xueli Yang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China.,Department of Ophthalmology, The First Affiliated Hospital of Dali University, Yunnan, China
| | - Xiaohong Wang
- Laboratory of Molecular Ophthalmology, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Bo Huang
- Department of Ophthalmology, University of Mississippi Medical Center, Jackson, Mississippi, United States
| | - Mei Du
- Laboratory of Molecular Ophthalmology, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
13
|
Meerschaut I, Vergult S, Dheedene A, Menten B, De Groote K, De Wilde H, Muiño Mosquera L, Panzer J, Vandekerckhove K, Coucke PJ, De Wolf D, Callewaert B. A Reassessment of Copy Number Variations in Congenital Heart Defects: Picturing the Whole Genome. Genes (Basel) 2021; 12:genes12071048. [PMID: 34356064 PMCID: PMC8304049 DOI: 10.3390/genes12071048] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/03/2021] [Accepted: 07/05/2021] [Indexed: 11/17/2022] Open
Abstract
Copy number variations (CNVs) can modulate phenotypes by affecting protein-coding sequences directly or through interference of gene expression. Recent studies in cancer and limb defects pinpointed the relevance of non-coding gene regulatory elements such as long non-coding RNAs (lncRNAs) and topologically associated domain (TAD)-related gene-enhancer interactions. The contribution of such non-coding elements is largely unexplored in congenital heart defects (CHD). We performed a retrospective analysis of CNVs reported in a cohort of 270 CHD patients. We reviewed the diagnostic yield of pathogenic CNVs, and performed a comprehensive reassessment of 138 CNVs of unknown significance (CNV-US), evaluating protein-coding genes, lncRNA genes, and potential interferences with TAD-related gene-enhancer interactions. Fifty-two of the 138 CNV-US may relate to CHD, revealing three candidate CHD regions, 19 candidate CHD genes, 80 lncRNA genes of interest, and six potentially CHD-related TAD interferences. Our study thus indicates a potential relevance of non-coding gene regulatory elements in CNV-related CHD pathogenesis. Shortcomings in our current knowledge on genomic variation call for continuous reporting of CNV-US in international databases, careful patient counseling, and additional functional studies to confirm these preliminary findings.
Collapse
Affiliation(s)
- Ilse Meerschaut
- Center for Medical Genetics, Ghent University Hospital, Belgium and Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; (I.M.); (S.V.); (A.D.); (B.M.); (L.M.M.); (P.J.C.)
- Department of Pediatric Cardiology, Ghent University Hospital, 9000 Ghent, Belgium; (K.D.G.); (H.D.W.); (J.P.); (K.V.); (D.D.W.)
| | - Sarah Vergult
- Center for Medical Genetics, Ghent University Hospital, Belgium and Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; (I.M.); (S.V.); (A.D.); (B.M.); (L.M.M.); (P.J.C.)
| | - Annelies Dheedene
- Center for Medical Genetics, Ghent University Hospital, Belgium and Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; (I.M.); (S.V.); (A.D.); (B.M.); (L.M.M.); (P.J.C.)
| | - Björn Menten
- Center for Medical Genetics, Ghent University Hospital, Belgium and Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; (I.M.); (S.V.); (A.D.); (B.M.); (L.M.M.); (P.J.C.)
| | - Katya De Groote
- Department of Pediatric Cardiology, Ghent University Hospital, 9000 Ghent, Belgium; (K.D.G.); (H.D.W.); (J.P.); (K.V.); (D.D.W.)
| | - Hans De Wilde
- Department of Pediatric Cardiology, Ghent University Hospital, 9000 Ghent, Belgium; (K.D.G.); (H.D.W.); (J.P.); (K.V.); (D.D.W.)
| | - Laura Muiño Mosquera
- Center for Medical Genetics, Ghent University Hospital, Belgium and Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; (I.M.); (S.V.); (A.D.); (B.M.); (L.M.M.); (P.J.C.)
- Department of Pediatric Cardiology, Ghent University Hospital, 9000 Ghent, Belgium; (K.D.G.); (H.D.W.); (J.P.); (K.V.); (D.D.W.)
| | - Joseph Panzer
- Department of Pediatric Cardiology, Ghent University Hospital, 9000 Ghent, Belgium; (K.D.G.); (H.D.W.); (J.P.); (K.V.); (D.D.W.)
| | - Kristof Vandekerckhove
- Department of Pediatric Cardiology, Ghent University Hospital, 9000 Ghent, Belgium; (K.D.G.); (H.D.W.); (J.P.); (K.V.); (D.D.W.)
| | - Paul J. Coucke
- Center for Medical Genetics, Ghent University Hospital, Belgium and Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; (I.M.); (S.V.); (A.D.); (B.M.); (L.M.M.); (P.J.C.)
| | - Daniël De Wolf
- Department of Pediatric Cardiology, Ghent University Hospital, 9000 Ghent, Belgium; (K.D.G.); (H.D.W.); (J.P.); (K.V.); (D.D.W.)
- Department of Pediatric Cardiology, Brussels University Hospital, 1090 Brussels, Belgium
| | - Bert Callewaert
- Center for Medical Genetics, Ghent University Hospital, Belgium and Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium; (I.M.); (S.V.); (A.D.); (B.M.); (L.M.M.); (P.J.C.)
- Correspondence: ; Tel.: +32-9-332-3603
| |
Collapse
|
14
|
Connexins in the Heart: Regulation, Function and Involvement in Cardiac Disease. Int J Mol Sci 2021; 22:ijms22094413. [PMID: 33922534 PMCID: PMC8122935 DOI: 10.3390/ijms22094413] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 12/20/2022] Open
Abstract
Connexins are a family of transmembrane proteins that play a key role in cardiac physiology. Gap junctional channels put into contact the cytoplasms of connected cardiomyocytes, allowing the existence of electrical coupling. However, in addition to this fundamental role, connexins are also involved in cardiomyocyte death and survival. Thus, chemical coupling through gap junctions plays a key role in the spreading of injury between connected cells. Moreover, in addition to their involvement in cell-to-cell communication, mounting evidence indicates that connexins have additional gap junction-independent functions. Opening of unopposed hemichannels, located at the lateral surface of cardiomyocytes, may compromise cell homeostasis and may be involved in ischemia/reperfusion injury. In addition, connexins located at non-canonical cell structures, including mitochondria and the nucleus, have been demonstrated to be involved in cardioprotection and in regulation of cell growth and differentiation. In this review, we will provide, first, an overview on connexin biology, including their synthesis and degradation, their regulation and their interactions. Then, we will conduct an in-depth examination of the role of connexins in cardiac pathophysiology, including new findings regarding their involvement in myocardial ischemia/reperfusion injury, cardiac fibrosis, gene transcription or signaling regulation.
Collapse
|
15
|
Wang CY, Liu SY, Kuo FH, Lin HF, Liu CY, Yang YP, Tsai FT, Huang WC, Tarng YW, Lin HC, Lu KH, Yu WC, Yang MY. Using multiple-steps bioinformatic analysis to predict the potential microRNA targets by cardiogenic HoxA11. J Chin Med Assoc 2021; 84:68-72. [PMID: 32773582 DOI: 10.1097/jcma.0000000000000397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND In this study, our major aim is to using multiple-steps bioinformatic analysis to predict cardiogenic genes with targeting mRNA profiling for predicting cardiogenic HoxA11 gene. METHODS We first analyzed the microarray data with bioinformatic measurement, including combining with panel module 1 (mouse embryonic stem cells), panel module 2 (mouse induced pluripotent stem cells), and panel module 3 (gene list form literature of heart development). A literature-based comparison of the two microarrays and a software-based (Targetscan program, www.targetscan.org) comparative analysis of the two datasets. Furthermore, we select the common central pathways and potential candidate genes involved in the cardiomyocyte-lineaged differentiation and development. RESULTS Schematic presentation of a putative miR181a target site in Hox-A11 3'UTR. The bioinformatic result showed that potential interacted cardiogenic targets of Tbx5, Tbx20, Mal2c, Nkx2.5, cTNT, Cx43, MHC, and MCK in different treatment groups of pluripotent stem cells by using a literature-based comparison of the two microarrays and a software-based gene-lineage system. CONCLUSION Our findings support that mir181a is an up-stream regulating microRNA to target the 3'UTR of HoxA11 mRNA during the process of cardiomyocyte differentiation.
Collapse
Affiliation(s)
- Chien-Ying Wang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Division of Trauma, Department of Emergency Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Szu-Yuan Liu
- Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
- College of Nursing, Central Taiwan University of Science and Technology, Taichung, Taiwan, ROC
| | - Fu-Hsuan Kuo
- Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
- Department of Neurosurgery, Tri-service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Heng-Fu Lin
- Division of Thoracic Surgery, Department of Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan, ROC
| | - Chao-Yu Liu
- Division of Traumatology, Department of Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan, ROC
| | - Yi-Ping Yang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Pharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan, ROC
| | - Fu-Ting Tsai
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Wei-Chun Huang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Critical Care Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
- Department of Physical Therapy, Fooyin University, Kaohsiung, Taiwan, ROC
| | - Yih-Wen Tarng
- Department of Orthopedics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
- Department of Emergency, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Hsin-Chi Lin
- Department of General Internal Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| | - Kai-Hsi Lu
- Department of Medical Research and Education, Cheng-Hsin General Hospital, Taipei, Taiwan, ROC
| | - Wen-Chung Yu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Division of Cardiology, Taipei Veterans General Hospital; Taipei, Taiwan, ROC
| | - Meng-Yin Yang
- Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
- College of Nursing, Central Taiwan University of Science and Technology, Taichung, Taiwan, ROC
- Department of Neurosurgery, Da-Li Jan-Ai Hospital, Taichung, Taiwan, ROC
- Department of Surgery/Neurosurgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| |
Collapse
|
16
|
Strauss RE, Gourdie RG. Cx43 and the Actin Cytoskeleton: Novel Roles and Implications for Cell-Cell Junction-Based Barrier Function Regulation. Biomolecules 2020; 10:E1656. [PMID: 33321985 PMCID: PMC7764618 DOI: 10.3390/biom10121656] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Barrier function is a vital homeostatic mechanism employed by epithelial and endothelial tissue. Diseases across a wide range of tissue types involve dynamic changes in transcellular junctional complexes and the actin cytoskeleton in the regulation of substance exchange across tissue compartments. In this review, we focus on the contribution of the gap junction protein, Cx43, to the biophysical and biochemical regulation of barrier function. First, we introduce the structure and canonical channel-dependent functions of Cx43. Second, we define barrier function and examine the key molecular structures fundamental to its regulation. Third, we survey the literature on the channel-dependent roles of connexins in barrier function, with an emphasis on the role of Cx43 and the actin cytoskeleton. Lastly, we discuss findings on the channel-independent roles of Cx43 in its associations with the actin cytoskeleton and focal adhesion structures highlighted by PI3K signaling, in the potential modulation of cellular barriers. Mounting evidence of crosstalk between connexins, the cytoskeleton, focal adhesion complexes, and junctional structures has led to a growing appreciation of how barrier-modulating mechanisms may work together to effect solute and cellular flux across tissue boundaries. This new understanding could translate into improved therapeutic outcomes in the treatment of barrier-associated diseases.
Collapse
Affiliation(s)
- Randy E. Strauss
- Virginia Tech, Translational Biology Medicine and Health (TBMH) Program, Roanoke, VA 24016, USA
| | - Robert G. Gourdie
- Center for Heart and Reparative Medicine Research, Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA 24016, USA
- Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| |
Collapse
|
17
|
van den Hoff MJB, Wessels A. Muscularization of the Mesenchymal Outlet Septum during Cardiac Development. J Cardiovasc Dev Dis 2020; 7:jcdd7040051. [PMID: 33158304 PMCID: PMC7711588 DOI: 10.3390/jcdd7040051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/02/2020] [Accepted: 11/02/2020] [Indexed: 12/11/2022] Open
Abstract
After the formation of the linear heart tube, it becomes divided into right and left components by the process of septation. Relatively late during this process, within the developing outflow tract, the initially mesenchymal outlet septum becomes muscularized as the result of myocardialization. Myocardialization is defined as the process in which existing cardiomyocytes migrate into flanking mesenchyme. Studies using genetically modified mice, as well as experimental approaches using in vitro models, demonstrate that Wnt and TGFβ signaling play an essential role in the regulation of myocardialization. They also show the significance of the interaction between cardiomyocytes, endocardial derived cells, neural crest cells, and the extracellular matrix. Interestingly, Wnt-mediated non-canonical planar cell polarity signaling was found to be a crucial regulator of myocardialization in the outlet septum and Wnt-mediated canonical β-catenin signaling is an essential regulator of the expansion of mesenchymal cells populating the outflow tract cushions.
Collapse
Affiliation(s)
- Maurice J. B. van den Hoff
- Department of Medical Biology, AmsterdamUMC, Location AMC, 1105AZ Amsterdam, The Netherlands
- Correspondence: ; Tel.: +1-3120-5665-405
| | - Andy Wessels
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA;
| |
Collapse
|
18
|
Picoli C, Soleilhac E, Journet A, Barette C, Comte M, Giaume C, Mouthon F, Fauvarque MO, Charvériat M. High-Content Screening Identifies New Inhibitors of Connexin 43 Gap Junctions. Assay Drug Dev Technol 2020; 17:240-248. [PMID: 31314551 DOI: 10.1089/adt.2019.927] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Gap junctions (GJs) are dynamic structures composed of hexamers of connexins (Cxs), a class of transmembrane proteins enabling channel-mediated direct intercellular communication through cell-cell diffusion of ions and small metabolites. In defined conditions, Cxs also work as hemichannels allowing exchanges between the cytoplasm and the extracellular medium. The most common GJ channel is formed by connexin 43 (Cx43) and plays an important role in physiological and pathological processes in excitable tissues, such as heart and brain. Hence, Cx43 has been largely envisioned as a new therapeutic target in cancer, neurological and psychiatric indications, or cardiovascular diseases. Identifying new pharmacological inhibitors of Cx43 GJs with different mechanisms of action and from diverse chemical classes is thus highly challenging. We present here a high-content screening method, based on the evaluation of fluorescent dye transfer rates between adjacent cells to monitor the function of GJs in U251 glioblastoma cells expressing high levels of Cx43. This assay was validated using well-described pharmacological GJ inhibitors such as mefloquine. The method was adapted to screen a library of 1,280 Food and Drug Administration- and European Medicines Agency-approved drugs that led to the selection of both known and new inhibitors of GJ channel function. We further focused on a specific class of microtubule-targeting agents, confirming that a proper tubulin network is required for functional Cx43 GJ channels.
Collapse
Affiliation(s)
| | - Emmanuelle Soleilhac
- 2Univ. Grenoble Alpes, CEA, Inserm, IRIG BGE-Genetics and Chemogenomics, Grenoble, France
| | - Agnès Journet
- 2Univ. Grenoble Alpes, CEA, Inserm, IRIG BGE-Genetics and Chemogenomics, Grenoble, France
| | - Caroline Barette
- 2Univ. Grenoble Alpes, CEA, Inserm, IRIG BGE-Genetics and Chemogenomics, Grenoble, France
| | - Marjorie Comte
- 2Univ. Grenoble Alpes, CEA, Inserm, IRIG BGE-Genetics and Chemogenomics, Grenoble, France
| | | | | | - Marie-Odile Fauvarque
- 2Univ. Grenoble Alpes, CEA, Inserm, IRIG BGE-Genetics and Chemogenomics, Grenoble, France
| | | |
Collapse
|
19
|
Abstract
Of the 21 members of the connexin family, 4 (Cx37, Cx40, Cx43, and Cx45) are expressed in the endothelium and/or smooth muscle of intact blood vessels to a variable and dynamically regulated degree. Full-length connexins oligomerize and form channel structures connecting the cytosol of adjacent cells (gap junctions) or the cytosol with the extracellular space (hemichannels). The different connexins vary mainly with regard to length and sequence of their cytosolic COOH-terminal tails. These COOH-terminal parts, which in the case of Cx43 are also translated as independent short isoforms, are involved in various cellular signaling cascades and regulate cell functions. This review focuses on channel-dependent and -independent effects of connexins in vascular cells. Channels play an essential role in coordinating and synchronizing endothelial and smooth muscle activity and in their interplay, in the control of vasomotor actions of blood vessels including endothelial cell reactivity to agonist stimulation, nitric oxide-dependent dilation, and endothelial-derived hyperpolarizing factor-type responses. Further channel-dependent and -independent roles of connexins in blood vessel function range from basic processes of vascular remodeling and angiogenesis to vascular permeability and interactions with leukocytes with the vessel wall. Together, these connexin functions constitute an often underestimated basis for the enormous plasticity of vascular morphology and function enabling the required dynamic adaptation of the vascular system to varying tissue demands.
Collapse
Affiliation(s)
- Ulrich Pohl
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Planegg-Martinsried, Germany; Biomedical Centre, Cardiovascular Physiology, LMU Munich, Planegg-Martinsried, Germany; German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany; and Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
20
|
Jourdeuil K, Taneyhill LA. The gap junction protein connexin 43 controls multiple aspects of cranial neural crest cell development. J Cell Sci 2020; 133:jcs235440. [PMID: 31964703 PMCID: PMC7044449 DOI: 10.1242/jcs.235440] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 01/10/2020] [Indexed: 12/14/2022] Open
Abstract
Gap junctions are intercellular channels between cells that facilitate cell-cell communication. Connexin 43 (Cx43; also known as GJA1), the predominant gap junction protein in vertebrates, is expressed in premigratory cranial neural crest cells and is maintained throughout the neural crest cell epithelial-to-mesenchymal transition (EMT), but its function in these cells is unknown. To this end, we used a combination of in vivo and ex vivo experiments to assess gap junction formation, and Cx43 function, in chick cranial neural crest cells. Our results demonstrate that gap junctions exist between premigratory and migratory cranial neural crest cells and depend on Cx43 for their function. In the embryo, Cx43 knockdown just prior to EMT delays the emergence of Cx43-depleted neural crest cells from the neural tube, but these cells eventually successfully emigrate and join the migratory stream. This delay can be rescued by introduction of full-length Cx43 into Cx43-depleted cells. Furthermore, Cx43 depletion reduces the size of the premigratory neural crest cell domain through an early effect on neural crest cell specification. Collectively, these data identify new roles for Cx43 in chick cranial neural crest cell development.
Collapse
Affiliation(s)
- Karyn Jourdeuil
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Lisa A Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
21
|
Abstract
The vertebrate heart tube forms from epithelial progenitor cells in the early embryo and subsequently elongates by progressive addition of second heart field (SHF) progenitor cells from adjacent splanchnic mesoderm. Failure to maximally elongate the heart results in a spectrum of morphological defects affecting the cardiac poles, including outflow tract alignment and atrioventricular septal defects, among the most common congenital birth anomalies. SHF cells constitute an atypical apicobasally polarized epithelium with dynamic basal filopodia, located in the dorsal wall of the pericardial cavity. Recent studies have highlighted the importance of epithelial architecture and cell adhesion in the SHF, particularly for signaling events that control the progenitor cell niche during heart tube elongation. The 22q11.2 deletion syndrome gene Tbx1 regulates progenitor cell status through modulating cell shape and filopodial activity and is required for SHF contributions to both cardiac poles. Noncanonical Wnt signaling and planar cell polarity pathway genes control epithelial polarity in the dorsal pericardial wall, as progenitor cells differentiate in a transition zone at the arterial pole. Defects in these pathways lead to outflow tract shortening. Moreover, new biomechanical models of heart tube elongation have been proposed based on analysis of tissue-wide forces driving epithelial morphogenesis in the SHF, including regional cell intercalation, cell cohesion, and epithelial tension. Regulation of the epithelial properties of SHF cells is thus emerging as a key step during heart tube elongation, adding a new facet to our understanding of the mechanisms underlying both heart morphogenesis and congenital heart defects.
Collapse
Affiliation(s)
- Claudio Cortes
- From Aix-Marseille University, CNRS UMR 7288, Developmental Biology Institute of Marseille, France
| | - Alexandre Francou
- From Aix-Marseille University, CNRS UMR 7288, Developmental Biology Institute of Marseille, France
| | - Christopher De Bono
- From Aix-Marseille University, CNRS UMR 7288, Developmental Biology Institute of Marseille, France
| | - Robert G Kelly
- From Aix-Marseille University, CNRS UMR 7288, Developmental Biology Institute of Marseille, France.
| |
Collapse
|
22
|
The Functional Implications of Endothelial Gap Junctions and Cellular Mechanics in Vascular Angiogenesis. Cancers (Basel) 2019; 11:cancers11020237. [PMID: 30781714 PMCID: PMC6406946 DOI: 10.3390/cancers11020237] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 02/08/2019] [Accepted: 02/13/2019] [Indexed: 12/27/2022] Open
Abstract
Angiogenesis—the sprouting and growth of new blood vessels from the existing vasculature—is an important contributor to tumor development, since it facilitates the supply of oxygen and nutrients to cancer cells. Endothelial cells are critically affected during the angiogenic process as their proliferation, motility, and morphology are modulated by pro-angiogenic and environmental factors associated with tumor tissues and cancer cells. Recent in vivo and in vitro studies have revealed that the gap junctions of endothelial cells also participate in the promotion of angiogenesis. Pro-angiogenic factors modulate gap junction function and connexin expression in endothelial cells, whereas endothelial connexins are involved in angiogenic tube formation and in the cell migration of endothelial cells. Several mechanisms, including gap junction function-dependent or -independent pathways, have been proposed. In particular, connexins might have the potential to regulate cell mechanics such as cell morphology, cell migration, and cellular stiffness that are dynamically changed during the angiogenic processes. Here, we review the implication for endothelial gap junctions and cellular mechanics in vascular angiogenesis.
Collapse
|
23
|
Wu M. Mechanisms of Trabecular Formation and Specification During Cardiogenesis. Pediatr Cardiol 2018; 39:1082-1089. [PMID: 29594501 PMCID: PMC6164162 DOI: 10.1007/s00246-018-1868-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/14/2018] [Indexed: 01/08/2023]
Abstract
Trabecular morphogenesis is a key morphologic event during cardiogenesis and contributes to the formation of a competent ventricular wall. Lack of trabeculation results in embryonic lethality. The trabecular morphogenesis is a multistep process that includes, but is not limited to, trabecular initiation, proliferation/growth, specification, and compaction. Although a number of signaling molecules have been implicated in regulating trabeculation, the cellular processes underlying mammalian trabecular formation are not fully understood. Recent works show that the myocardium displays polarity, and oriented cell division (OCD) and directional migration of the cardiomyocytes in the monolayer myocardium are required for trabecular initiation and formation. Furthermore, perpendicular OCD is an extrinsic asymmetric cell division that contributes to trabecular specification, and is a mechanism that causes the trabecular cardiomyocytes to be distinct from the cardiomyocytes in compact zone. Once the coronary vasculature system starts to function in the embryonic heart, the trabeculae will coalesce with the compact zone to thicken the heart wall, and abnormal compaction will lead to left ventricular non-compaction (LVNC) and heart failure. There are many reviews about compaction and LVNC. In this review, we will focus on the roles of myocardial polarity and OCD in trabecular initiation, formation, and specification.
Collapse
Affiliation(s)
- Mingfu Wu
- Department of Molecular and Cellular Physiology, Albany Medical College, 43 New Scotland Ave, Albany, NY, 12208, USA.
| |
Collapse
|
24
|
Ramai D, Lai J, Monzidelis C, Reddy S. Coronary Artery Development: Origin, Malformations, and Translational Vascular Reparative Therapy. J Cardiovasc Pharmacol Ther 2018; 23:292-300. [PMID: 29642708 DOI: 10.1177/1074248418769633] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
After thickening of the cardiac chamber walls during embryogenesis, oxygen and nutrients can no longer be adequately supplied to cardiac cells via passive diffusion; therefore, a primitive vascular network develops to supply these vital structures. This plexus further matures into coronary arteries and veins, which ensures continued development of the heart. Various models have been proposed to account for the growth of the coronary arteries. However, lineage-tracing studies in the last decade have identified 3 major sources, namely, the proepicardium, the sinus venosus, and endocardium. Although the exact contribution of each source remains unknown, the emerging model depicts alternative pathways and progenitor cells, which ensure successful coronary angiogenesis. We aim to explore the current trends in coronary artery development, the cellular and molecular signals regulating heart vascularization, and its implications for heart disease and vascular regeneration.
Collapse
Affiliation(s)
- Daryl Ramai
- Department of Medicine, The Brooklyn Hospital Center, Academic Affiliate of The Icahn School of Medicine at Mount Sinai, Clinical Affiliate of The Mount Sinai Hospital, Brooklyn, NY, USA
- Department of Anatomical Sciences, School of Medicine, St George’s University, Grenada, West Indies
| | - Jonathan Lai
- Department of Anatomical Sciences, School of Medicine, St George’s University, Grenada, West Indies
| | - Constantine Monzidelis
- Department of Medicine, The Brooklyn Hospital Center, Academic Affiliate of The Icahn School of Medicine at Mount Sinai, Clinical Affiliate of The Mount Sinai Hospital, Brooklyn, NY, USA
| | - Sarath Reddy
- Division of Cardiology, The Brooklyn Hospital Center, Academic Affiliate of The Icahn School of Medicine at Mount Sinai, Clinical Affiliate of The Mount Sinai Hospital, Brooklyn, NY, USA
| |
Collapse
|
25
|
Liu X, Wang Y, Liu F, Zhang M, Song H, Zhou B, Lo CW, Tong S, Hu Z, Zhang Z. Wdpcp promotes epicardial EMT and epicardium-derived cell migration to facilitate coronary artery remodeling. Sci Signal 2018; 11:11/519/eaah5770. [PMID: 29487191 DOI: 10.1126/scisignal.aah5770] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
During coronary vasculature development, endothelial cells enclose the embryonic heart to form the primitive coronary plexus. This structure is remodeled upon recruitment of epicardial cells that may undergo epithelial-mesenchymal transition (EMT) to enable migration and that give rise to smooth muscle cells. In mice expressing a loss-of-function mutant form of Wdpcp, a gene involved in ciliogenesis, the enclosure of the surface of the heart by the subepicardial coronary plexus was accelerated because of enhanced chemotactic responses to Shh. Coronary arteries, but not coronary veins in Wdpcp mutant mice, showed reduced smooth muscle cell coverage. In addition, Wdpcp mutant hearts had reduced expression of EMT and mesenchymal markers and had fewer epicardium-derived cells (EPDCs) that showed impaired migration. Epicardium-specific deletion of Wdpcp recapitulated the coronary artery defect of the Wdpcp mutant. Thus, Wdpcp promotes epithelial EMT and EPDC migration, processes that are required for remodeling of the coronary primitive plexus. The Wdpcp mutant mice will be a useful tool to dissect the molecular mechanisms that govern the remodeling of the primitive plexus during coronary development.
Collapse
Affiliation(s)
- Xiangyang Liu
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Ye Wang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Feng Liu
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Min Zhang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Hejie Song
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Bin Zhou
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA 15201, USA
| | - Shilu Tong
- Department of Clinical Epidemiology and Biostatistics, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Zhenlei Hu
- Department of Cardiovascular Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China.
| | - Zhen Zhang
- Shanghai Pediatric Congenital Heart Disease Institute and Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China.
| |
Collapse
|
26
|
Nyffeler J, Chovancova P, Dolde X, Holzer AK, Purvanov V, Kindinger I, Kerins A, Higton D, Silvester S, van Vugt-Lussenburg BMA, Glaab E, van der Burg B, Maclennan R, Legler DF, Leist M. A structure-activity relationship linking non-planar PCBs to functional deficits of neural crest cells: new roles for connexins. Arch Toxicol 2017; 92:1225-1247. [PMID: 29164306 DOI: 10.1007/s00204-017-2125-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 11/15/2017] [Indexed: 12/14/2022]
Abstract
Migration of neural crest cells (NCC) is a fundamental developmental process, and test methods to identify interfering toxicants have been developed. By examining cell function endpoints, as in the 'migration-inhibition of NCC (cMINC)' assay, a large number of toxicity mechanisms and protein targets can be covered. However, the key events that lead to the adverse effects of a given chemical or group of related compounds are hard to elucidate. To address this issue, we explored here, whether the establishment of two overlapping structure-activity relationships (SAR)-linking chemical structure on the one hand to a phenotypic test outcome, and on the other hand to a mechanistic endpoint-was useful as strategy to identify relevant toxicity mechanisms. For this purpose, we chose polychlorinated biphenyls (PCB) as a large group of related, but still toxicologically and physicochemically diverse structures. We obtained concentration-dependent data for 26 PCBs in the cMINC assay. Moreover, the test chemicals were evaluated by a new high-content imaging method for their effect on cellular re-distribution of connexin43 and for their capacity to inhibit gap junctions. Non-planar PCBs inhibited NCC migration. The potency (1-10 µM) correlated with the number of ortho-chlorine substituents; non-ortho-chloro (planar) PCBs were non-toxic. The toxicity to NCC partially correlated with gap junction inhibition, while it fully correlated (p < 0.0004) with connexin43 cellular re-distribution. Thus, our double-SAR strategy revealed a mechanistic step tightly linked to NCC toxicity of PCBs. Connexin43 patterns in NCC may be explored as a new endpoint relevant to developmental toxicity screening.
Collapse
Affiliation(s)
- Johanna Nyffeler
- In vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Konstanz, Germany.,Research Training Group RTG1331, 78457, Konstanz, Germany
| | - Petra Chovancova
- In vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Konstanz, Germany.,Konstanz Research School Chemical Biology (KoRS-CB), 78457, Konstanz, Germany
| | - Xenia Dolde
- In vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Konstanz, Germany.,Konstanz Research School Chemical Biology (KoRS-CB), 78457, Konstanz, Germany
| | - Anna-Katharina Holzer
- In vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Konstanz, Germany
| | - Vladimir Purvanov
- Biotechnology Institute Thurgau at the University of Konstanz, 8280, Kreuzlingen, Switzerland
| | - Ilona Kindinger
- Biotechnology Institute Thurgau at the University of Konstanz, 8280, Kreuzlingen, Switzerland
| | - Anna Kerins
- Cyprotex Discovery, No 24 Mereside, Alderley Park, Cheshire, SK10 4TG, UK
| | - David Higton
- Cyprotex Discovery, No 24 Mereside, Alderley Park, Cheshire, SK10 4TG, UK
| | - Steve Silvester
- Cyprotex Discovery, No 24 Mereside, Alderley Park, Cheshire, SK10 4TG, UK
| | | | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 4362, Esch-sur-Alzette, Luxembourg
| | - Bart van der Burg
- BioDetection Systems bv, Science Park 406, 1098XH, Amsterdam, The Netherlands
| | - Richard Maclennan
- Cyprotex Discovery, No 24 Mereside, Alderley Park, Cheshire, SK10 4TG, UK
| | - Daniel F Legler
- Research Training Group RTG1331, 78457, Konstanz, Germany.,Konstanz Research School Chemical Biology (KoRS-CB), 78457, Konstanz, Germany.,Biotechnology Institute Thurgau at the University of Konstanz, 8280, Kreuzlingen, Switzerland
| | - Marcel Leist
- In vitro Toxicology and Biomedicine, Department Inaugurated by the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Konstanz, Germany. .,Research Training Group RTG1331, 78457, Konstanz, Germany. .,Konstanz Research School Chemical Biology (KoRS-CB), 78457, Konstanz, Germany.
| |
Collapse
|
27
|
Venhuizen JH, Zegers MM. Making Heads or Tails of It: Cell-Cell Adhesion in Cellular and Supracellular Polarity in Collective Migration. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a027854. [PMID: 28246177 DOI: 10.1101/cshperspect.a027854] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Collective cell migration is paramount to morphogenesis and contributes to the pathogenesis of cancer. To migrate directionally and reach their site of destination, migrating cells must distinguish a front and a rear. In addition to polarizing individually, cell-cell interactions in collectively migrating cells give rise to a higher order of polarity, which allows them to move as a supracellular unit. Rather than just conferring adhesion, emerging evidence indicates that cadherin-based adherens junctions intrinsically polarize the cluster and relay mechanical signals to establish both intracellular and supracellular polarity. In this review, we discuss the various functions of adherens junctions in polarity of migrating cohorts.
Collapse
Affiliation(s)
- Jan-Hendrik Venhuizen
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands
| | - Mirjam M Zegers
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
28
|
Elashry MI, Heimann M, Wenisch S, Patel K, Arnhold S. Multipotency of skeletal muscle stem cells on their native substrate and the expression of Connexin 43 during adoption of adipogenic and osteogenic fate. Acta Histochem 2017; 119:786-794. [PMID: 29037777 DOI: 10.1016/j.acthis.2017.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/06/2017] [Accepted: 10/06/2017] [Indexed: 12/25/2022]
Abstract
Muscle regeneration is performed by resident muscle stem cells called satellite cells (SC). However they are multipotent, being able to adopt adipogenic and osteogenic fate under the correct stimuli. Since SC behavior can be regulated by the extra-cellular matrix, we examined the robustness of the myogenic programme of SC on their native substrate-the surface of a myofiber. We show that the native substrate supports myogenic differentiation judged by the expression of members of the Myogenic Determination Factor (MRF) family. However SC even on their native substrate can be induced into adopting adipogenic or osteogenic fate. Furthermore conditions that support adipose or bone formation inhibit the proliferation of SC progeny as well as their migration. We show that Connexin43 (Cx43), a gap junction complex protein, is only expressed by activated and not quiescent SC. Furthermore, it is not expressed by SC that are in the process of changing their fate. Lastly we show that intact adult mouse muscle contains numerous cells expressing Cx43 and that the density of these cells seems to be related to capillary density. We suggest the Cx43 expression is localized to angioblasts and is more prominent in oxidative slow muscle compared to glycolytic fast muscle.
Collapse
|
29
|
Rhett JM, Calder BW, Fann SA, Bainbridge H, Gourdie RG, Yost MJ. Mechanism of action of the anti-inflammatory connexin43 mimetic peptide JM2. Am J Physiol Cell Physiol 2017; 313:C314-C326. [PMID: 28701358 PMCID: PMC5625091 DOI: 10.1152/ajpcell.00229.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 07/05/2017] [Accepted: 07/05/2017] [Indexed: 12/31/2022]
Abstract
Connexin-based therapeutics have shown the potential for therapeutic efficacy in improving wound healing. Our previous work demonstrated that the connexin43 (Cx43) mimetic peptide juxtamembrane 2 (JM2) reduced the acute inflammatory response to a submuscular implant model by inhibiting purinergic signaling. Given the prospective application in improving tissue-engineered construct tolerance that these results indicated, we sought to determine the mechanism of action for JM2 in the present study. Using confocal microscopy, a gap-FRAP cell communication assay, and an ethidium bromide uptake assay of hemichannel function we found that the peptide reduced cell surface Cx43 levels, Cx43 gap junction (GJ) size, GJ communication, and hemichannel activity. JM2 is based on the sequence of the Cx43 microtubule binding domain, and microtubules have a confirmed role in intracellular trafficking of Cx43 vesicles. Therefore, we tested the effect of JM2 on Cx43-microtubule interaction and microtubule polymerization. We found that JM2 enhanced Cx43-microtubule interaction and that microtubule polymerization was significantly enhanced. Taken together, these data suggest that JM2 inhibits trafficking of Cx43 to the cell surface by promoting irrelevant microtubule polymerization and thereby reduces the number of hemichannels in the plasma membrane available to participate in proinflammatory purinergic signaling. Importantly, this work indicates that JM2 may have therapeutic value in the treatment of proliferative diseases such as cancer. We conclude that the targeted action of JM2 on Cx43 channels may improve the tolerance of implanted tissue-engineered constructs against the innate inflammatory response.
Collapse
Affiliation(s)
- J Matthew Rhett
- Department of Surgery, General Surgery Division, Medical University of South Carolina, Charleston, South Carolina;
| | - Bennett W Calder
- Department of Surgery, General Surgery Division, Medical University of South Carolina, Charleston, South Carolina
| | - Stephen A Fann
- Department of Surgery, General Surgery Division, Medical University of South Carolina, Charleston, South Carolina
| | - Heather Bainbridge
- Department of Surgery, General Surgery Division, Medical University of South Carolina, Charleston, South Carolina
| | - Robert G Gourdie
- Center for Heart and Regenerative Medicine Research, Virginia Tech Carilion Research Institute, Roanoke, Virginia; and
| | - Michael J Yost
- Department of Surgery, General Surgery Division, Medical University of South Carolina, Charleston, South Carolina.,Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
30
|
Leithe E, Mesnil M, Aasen T. The connexin 43 C-terminus: A tail of many tales. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:48-64. [PMID: 28526583 DOI: 10.1016/j.bbamem.2017.05.008] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/10/2017] [Accepted: 05/12/2017] [Indexed: 10/19/2022]
Abstract
Connexins are chordate gap junction channel proteins that, by enabling direct communication between the cytosols of adjacent cells, create a unique cell signalling network. Gap junctional intercellular communication (GJIC) has important roles in controlling cell growth and differentiation and in tissue development and homeostasis. Moreover, several non-canonical connexin functions unrelated to GJIC have been discovered. Of the 21 members of the human connexin family, connexin 43 (Cx43) is the most widely expressed and studied. The long cytosolic C-terminus (CT) of Cx43 is subject to extensive post-translational modifications that modulate its intracellular trafficking and gap junction channel gating. Moreover, the Cx43 CT contains multiple domains involved in protein interactions that permit crosstalk between Cx43 and cytoskeletal and regulatory proteins. These domains endow Cx43 with the capacity to affect cell growth and differentiation independently of GJIC. Here, we review the current understanding of the regulation and unique functions of the Cx43 CT, both as an essential component of full-length Cx43 and as an independent signalling hub. We highlight the complex regulatory and signalling networks controlled by the Cx43 CT, including the extensive protein interactome that underlies both gap junction channel-dependent and -independent functions. We discuss these data in relation to the recent discovery of the direct translation of specific truncated forms of Cx43. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Edward Leithe
- Department of Molecular Oncology, Institute for Cancer Research, University of Oslo, NO-0424 Oslo, Norway; Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, NO-0424 Oslo, Norway
| | - Marc Mesnil
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers 86073, France
| | - Trond Aasen
- Translational Molecular Pathology, Vall d'Hebron Institute of Research (VHIR), Autonomous University of Barcelona, CIBERONC, 08035 Barcelona, Spain.
| |
Collapse
|
31
|
Trembley MA, Velasquez LS, Small EM. Epicardial Outgrowth Culture Assay and Ex Vivo Assessment of Epicardial-derived Cell Migration. J Vis Exp 2016:53750. [PMID: 27023710 PMCID: PMC4829037 DOI: 10.3791/53750] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
A single layer of epicardial cells lines the heart, providing paracrine factors that stimulate cardiomyocyte proliferation and directly contributing cardiovascular progenitors during development and disease. While a number of factors have been implicated in epicardium-derived cell (EPDC) mobilization, the mechanisms governing their subsequent migration and differentiation are poorly understood. Here, we present in vitro and ex vivo strategies to study EPDC motility and differentiation. First, we describe a method of obtaining primary epicardial cells by outgrowth culture from the embryonic mouse heart. We also introduce a detailed protocol to assess three-dimensional migration of labeled EPDC in an organ culture system. We provide evidence using these techniques that genetic deletion of myocardin-related transcription factors in the epicardium attenuates EPDC migration. This approach serves as a platform to evaluate candidate modifiers of EPDC biology and could be used to develop genetic or chemical screens to identify novel regulators of EPDC mobilization that might be useful for cardiac repair.
Collapse
Affiliation(s)
- Michael A Trembley
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry; Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry
| | - Lissette S Velasquez
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry
| | - Eric M Small
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry; Department of Medicine, University of Rochester School of Medicine and Dentistry; Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry;
| |
Collapse
|
32
|
Yuan D, Sun G, Zhang R, Luo C, Ge M, Luo G, Hei Z. Connexin 43 expressed in endothelial cells modulates monocyte‑endothelial adhesion by regulating cell adhesion proteins. Mol Med Rep 2015; 12:7146-52. [PMID: 26324251 DOI: 10.3892/mmr.2015.4273] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 08/04/2015] [Indexed: 11/05/2022] Open
Abstract
Adhesion between circulating monocytes and vascular endothelial cells is a key initiator of atherosclerosis. In our previous studies, it was demonstrated that the expression of connexin (Cx)43 in monocytes modulates cell adhesion, however, the effects of the expression of Cx43 in endothelial cells remains to be elucidated. Therefore, the present study investigated the role of the expression of Cx43 in endothelial cells in the process of cell adhesion. A total of four different methods with distinct mechanisms were used to change the function and expression of Cx43 channels in human umbilical vein endothelial cells: Cx43 channel inhibitor (oleamide), enhancer (retinoic acid), overexpression of Cx43 by transfection with pcDNA‑Cx43 and knock‑down of the expression of Cx43 by small interfering RNA against Cx43. The results indicated that the upregulation of the expression of Cx43 enhanced monocyte‑endothelial adhesion and this was markedly decreased by downregulation of Cx43. This mechanism was associated with Cx43‑induced expression of vascular cell adhesion molecule‑1 and intercellular cell adhesion molecule‑1. The effects of Cx43 in endothelial cells was independent of Cx37 or Cx40. These experiments suggested that local regulation of endothelial Cx43 expression within the vasculature regulates monocyte‑endothelial adhesion, a critical event in the development of atherosclerosis and other inflammatory pathologies, with baseline adhesion set by the expression of Cx43. This balance may be crucial in controlling leukocyte involvement in inflammatory cascades.
Collapse
Affiliation(s)
- Dongdong Yuan
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Guoliang Sun
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Rui Zhang
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Chenfang Luo
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Mian Ge
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Gangjian Luo
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Ziqing Hei
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat‑Sen University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
33
|
Kameritsch P, Kiemer F, Beck H, Pohl U, Pogoda K. Cx43 increases serum induced filopodia formation via activation of p21-activated protein kinase 1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2907-17. [PMID: 26255026 DOI: 10.1016/j.bbamcr.2015.08.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/13/2015] [Accepted: 08/04/2015] [Indexed: 01/02/2023]
Abstract
In a previous study we could show that connexin 43 (Cx43) expression increased the migration of cells in a channel-independent manner involving the MAPK p38. We analyzed here the mechanism by which Cx43 enhanced p38 activation and migration related changes of the actin cytoskeleton. HeLa cells were used as a model system for the controlled expression of Cx43 and truncated Cx43 proteins. The expression of Cx43 altered the actin cytoskeleton organization in response to serum stimulation. Cx43 expressing HeLa cells had significantly more filopodial protrusions per cell than empty-vector transfected control cells. The expression of the channel incompetent carboxyl tail of Cx43 was sufficient to enhance the filopodia formation whereas the N-terminal, channel-building part, had no such effect. The enhanced filopodia formation was p38 dependent since the p38 blocker SB203580 significantly diminished it. Immunoprecipitation revealed an interaction of the upstream regulator of p38, p21-activated protein kinase 1 (PAK1), with Cx43 resulting in an enhanced phosphorylation of PAK1. Moreover, p38 activation, filopodia formation and cell migration were significantly reduced by blocking the PAK1 activity with its pharmacological inhibitor, IPA-3. The p38 target Hsp27, which favors the actin polymerization in its phosphorylated form, was significantly more phosphorylated characterizing it as a potential candidate molecule to enhance the serum-induced actin polymerization in Cx43 expressing cells. Our results provide a novel mechanism by which Cx43 can modify actin cytoskeletal dynamics and may thereby enhance cell migration.
Collapse
Affiliation(s)
- Petra Kameritsch
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, Marchioninistr. 27, 81377 München, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Biedersteiner Str. 29, 80802 München, Germany.
| | - Felizitas Kiemer
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, Marchioninistr. 27, 81377 München, Germany.
| | - Heike Beck
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, Marchioninistr. 27, 81377 München, Germany.
| | - Ulrich Pohl
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, Marchioninistr. 27, 81377 München, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Biedersteiner Str. 29, 80802 München, Germany; Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 München, Germany.
| | - Kristin Pogoda
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, Marchioninistr. 27, 81377 München, Germany; DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Biedersteiner Str. 29, 80802 München, Germany.
| |
Collapse
|
34
|
Sin WC, Aftab Q, Bechberger JF, Leung JH, Chen H, Naus CC. Astrocytes promote glioma invasion via the gap junction protein connexin43. Oncogene 2015; 35:1504-16. [DOI: 10.1038/onc.2015.210] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 04/22/2015] [Accepted: 04/24/2015] [Indexed: 01/03/2023]
|
35
|
Tabernero A, Gangoso E, Jaraíz-Rodríguez M, Medina JM. The role of connexin43-Src interaction in astrocytomas: A molecular puzzle. Neuroscience 2015; 323:183-94. [PMID: 25711938 DOI: 10.1016/j.neuroscience.2015.02.029] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 02/10/2015] [Accepted: 02/13/2015] [Indexed: 12/18/2022]
Abstract
Connexin43 (Cx43) as a building block of gap junction channels and hemichannels exerts important functions in astrocytes. When these cells acquire a malignant phenotype Cx43 protein but not mRNA levels are downregulated, being negligible in high-grade astrocytoma or glioblastoma multiforme, the most common and deadliest of malignant primary brain tumors in adults. Some microRNAs associated to glioma target Cx43 and could explain the lack of correlation between mRNA and protein levels of Cx43 found in some high-grade astrocytomas. More importantly, these microRNAs could be a promising therapeutic target. A great number of studies have confirmed the relationship between cancer and connexins that was proposed by Loewenstein more than 40years ago, but these studies have also revealed that this is a very complex relationship. Indeed, restoring Cx43 to glioma cells reduces their rate of proliferation and their tumorigenicity but this tumor suppressor effect could be counterbalanced by its effects on invasiveness, adhesion and migration. The mechanisms underlying these effects suggest the participation of a great variety of proteins that bind to different regions of Cx43. The present review focuses on an intrinsically disordered region of the C-terminal domain of Cx43 in which converges the interaction of several proteins, including the proto-oncogene Src. We summarize data that indicate that Cx43-Src interaction inhibits the oncogenic activity of Src and promotes a conformational change in the structure of Cx43 that allosterically modifies the binding to other important signaling proteins. As a consequence, crucial cell functions, such as proliferation or migration, could be strongly affected. We propose that the knowledge of the structural basis of the antitumorigenic effect of Cx43 on astrocytomas could help to design new therapies against this incurable disease.
Collapse
Affiliation(s)
- A Tabernero
- Departamento de Bioquímica y Biología Molecular, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Spain.
| | - E Gangoso
- Departamento de Bioquímica y Biología Molecular, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Spain
| | - M Jaraíz-Rodríguez
- Departamento de Bioquímica y Biología Molecular, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Spain
| | - J M Medina
- Departamento de Bioquímica y Biología Molecular, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Spain
| |
Collapse
|
36
|
Kelly JJ, Simek J, Laird DW. Mechanisms linking connexin mutations to human diseases. Cell Tissue Res 2014; 360:701-21. [DOI: 10.1007/s00441-014-2024-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 09/26/2014] [Indexed: 11/30/2022]
|
37
|
Diman NYSG, Brooks G, Kruithof BPT, Elemento O, Seidman JG, Seidman CE, Basson CT, Hatcher CJ. Tbx5 is required for avian and Mammalian epicardial formation and coronary vasculogenesis. Circ Res 2014; 115:834-44. [PMID: 25245104 DOI: 10.1161/circresaha.115.304379] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Holt-Oram syndrome is an autosomal dominant heart-hand syndrome caused by mutations in the TBX5 gene. Overexpression of Tbx5 in the chick proepicardial organ impaired coronary blood vessel formation. However, the potential activity of Tbx5 in the epicardium itself, and the role of Tbx5 in mammalian coronary vasculogenesis, remains largely unknown. OBJECTIVE To evaluate the consequences of altered Tbx5 gene dosage during proepicardial organ and epicardial development in the embryonic chick and mouse. METHODS AND RESULTS Retroviral-mediated knockdown or upregulation of Tbx5 expression in the embryonic chick proepicardial organ and proepicardial-specific deletion of Tbx5 in the embryonic mouse (Tbx5(epi-/)) impaired normal proepicardial organ cell development, inhibited epicardial and coronary blood vessel formation, and altered developmental gene expression. The generation of epicardial-derived cells and their migration into the myocardium were impaired between embryonic day (E) 13.5 to 15.5 in mutant hearts because of delayed epicardial attachment to the myocardium and subepicardial accumulation of epicardial-derived cells. This caused defective coronary vasculogenesis associated with impaired vascular smooth muscle cell recruitment and reduced invasion of cardiac fibroblasts and endothelial cells into myocardium. In contrast to wild-type hearts that exhibited an elaborate ventricular vascular network, Tbx5(epi-/-) hearts displayed a marked decrease in vascular density that was associated with myocardial hypoxia as exemplified by hypoxia inducible factor-1α upregulation and increased binding of hypoxyprobe-1. Tbx5(epi-/-) mice with such myocardial hypoxia exhibited reduced exercise capacity when compared with wild-type mice. CONCLUSIONS Our findings support a conserved Tbx5 dose-dependent requirement for both proepicardial and epicardial progenitor cell development in chick and in mouse coronary vascular formation.
Collapse
Affiliation(s)
- Nata Y S-G Diman
- From the Center for Molecular Cardiology, Greenberg Division of Cardiology (N.Y.S.-G.D., G.B., B.P.T.K., C.T.B., C.J.H.) and Department of Physiology and Biophysics (O.E.), Weill Cornell Medical College, New York, NY; Department of Genetics, Harvard Medical School, Boston, MA (J.G.S., C.E.S.); and Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, PA (C.J.H.)
| | - Gabriel Brooks
- From the Center for Molecular Cardiology, Greenberg Division of Cardiology (N.Y.S.-G.D., G.B., B.P.T.K., C.T.B., C.J.H.) and Department of Physiology and Biophysics (O.E.), Weill Cornell Medical College, New York, NY; Department of Genetics, Harvard Medical School, Boston, MA (J.G.S., C.E.S.); and Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, PA (C.J.H.)
| | - Boudewijn P T Kruithof
- From the Center for Molecular Cardiology, Greenberg Division of Cardiology (N.Y.S.-G.D., G.B., B.P.T.K., C.T.B., C.J.H.) and Department of Physiology and Biophysics (O.E.), Weill Cornell Medical College, New York, NY; Department of Genetics, Harvard Medical School, Boston, MA (J.G.S., C.E.S.); and Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, PA (C.J.H.)
| | - Olivier Elemento
- From the Center for Molecular Cardiology, Greenberg Division of Cardiology (N.Y.S.-G.D., G.B., B.P.T.K., C.T.B., C.J.H.) and Department of Physiology and Biophysics (O.E.), Weill Cornell Medical College, New York, NY; Department of Genetics, Harvard Medical School, Boston, MA (J.G.S., C.E.S.); and Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, PA (C.J.H.)
| | - J G Seidman
- From the Center for Molecular Cardiology, Greenberg Division of Cardiology (N.Y.S.-G.D., G.B., B.P.T.K., C.T.B., C.J.H.) and Department of Physiology and Biophysics (O.E.), Weill Cornell Medical College, New York, NY; Department of Genetics, Harvard Medical School, Boston, MA (J.G.S., C.E.S.); and Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, PA (C.J.H.)
| | - Christine E Seidman
- From the Center for Molecular Cardiology, Greenberg Division of Cardiology (N.Y.S.-G.D., G.B., B.P.T.K., C.T.B., C.J.H.) and Department of Physiology and Biophysics (O.E.), Weill Cornell Medical College, New York, NY; Department of Genetics, Harvard Medical School, Boston, MA (J.G.S., C.E.S.); and Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, PA (C.J.H.)
| | - Craig T Basson
- From the Center for Molecular Cardiology, Greenberg Division of Cardiology (N.Y.S.-G.D., G.B., B.P.T.K., C.T.B., C.J.H.) and Department of Physiology and Biophysics (O.E.), Weill Cornell Medical College, New York, NY; Department of Genetics, Harvard Medical School, Boston, MA (J.G.S., C.E.S.); and Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, PA (C.J.H.).
| | - Cathy J Hatcher
- From the Center for Molecular Cardiology, Greenberg Division of Cardiology (N.Y.S.-G.D., G.B., B.P.T.K., C.T.B., C.J.H.) and Department of Physiology and Biophysics (O.E.), Weill Cornell Medical College, New York, NY; Department of Genetics, Harvard Medical School, Boston, MA (J.G.S., C.E.S.); and Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, PA (C.J.H.).
| |
Collapse
|
38
|
Zhang SS, Shaw RM. Trafficking highways to the intercalated disc: new insights unlocking the specificity of connexin 43 localization. ACTA ACUST UNITED AC 2014; 21:43-54. [PMID: 24460200 DOI: 10.3109/15419061.2013.876014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
With each heartbeat, billions of cardiomyocytes work in concert to propagate the electrical excitation needed to effectively circulate blood. Regulated expression and timely delivery of connexin proteins to form gap junctions at the specialized cell-cell contact region, known as the intercalated disc, is essential to ventricular cardiomyocyte coupling. We focus this review on several regulatory mechanisms that have been recently found to govern the lifecycle of connexin 43 (Cx43), the short-lived and most abundantly expressed connexin in cardiac ventricular muscle. The Cx43 lifecycle begins with gene expression, followed by oligomerization into hexameric channels, and then cytoskeletal-based transport toward the disc region. Once delivered, hemichannels interact with resident disc proteins and are organized to effect intercellular coupling. We highlight recent studies exploring regulation of Cx43 localization to the intercalated disc, with emphasis on alternatively translated Cx43 isoforms and cytoskeletal transport machinery that together regulate Cx43 gap junction coupling between cardiomyocytes.
Collapse
|
39
|
Dyce PW, Li D, Barr KJ, Kidder GM. Connexin43 is required for the maintenance of multipotency in skin-derived stem cells. Stem Cells Dev 2014; 23:1636-46. [PMID: 24694074 DOI: 10.1089/scd.2013.0459] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Expression of the gap junction protein, connexin43 (Cx43), begins early during embryogenesis and is maintained in many different cell types. Several stem cell populations have been shown to express Cx43 and to form functional gap junctions. While it is clear that Cx43 is critical to the function of many organs, whether the same is true for stem cells has not been clearly demonstrated. Recently, stem cells isolated from newborn mouse skin were shown to form oocyte-like cells (OLCs) in vitro, hence the present study focussed on the role Cx43 plays in the proliferation and differentiation of these cells. The stem cells express Cx43 and those from knockout mice (Cx43 KO) exhibited significantly reduced cell-cell coupling. Loss of Cx43 reduced the rate of cellular migration [Cx43 KO, 1.57±0.65 radial cell units (RCU); wildtype (WT), 5.57±0.37 RCU] but increased the proliferation rate of the stem cells (Cx43 KO, 29.40%±2.02%; WT, 12.76%±1.50%). The expression of the pluripotency markers OCT4 and Nanog were found to be reduced in the Cx43 KO population, suggesting an inhibition of differentiation potential. To test the differentiation ability, the stem cells were induced to form neuronal cell types in vitro. While both the WT and KO cells were able to form GFAP-positive astrocytic cells, only WT stem cells were able to form βIII tubulin-positive neurons. Similarly, the ability of the stem cells to form OLCs was ablated by the loss of Cx43. These data reveal a role for Cx43 in maintaining multipotency within the skin-derived stem cell population.
Collapse
Affiliation(s)
- Paul W Dyce
- 1 Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario , London, Ontario, Canada
| | | | | | | |
Collapse
|
40
|
Billaud M, Lohman AW, Johnstone SR, Biwer LA, Mutchler S, Isakson BE. Regulation of cellular communication by signaling microdomains in the blood vessel wall. Pharmacol Rev 2014; 66:513-69. [PMID: 24671377 PMCID: PMC3973613 DOI: 10.1124/pr.112.007351] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It has become increasingly clear that the accumulation of proteins in specific regions of the plasma membrane can facilitate cellular communication. These regions, termed signaling microdomains, are found throughout the blood vessel wall where cellular communication, both within and between cell types, must be tightly regulated to maintain proper vascular function. We will define a cellular signaling microdomain and apply this definition to the plethora of means by which cellular communication has been hypothesized to occur in the blood vessel wall. To that end, we make a case for three broad areas of cellular communication where signaling microdomains could play an important role: 1) paracrine release of free radicals and gaseous molecules such as nitric oxide and reactive oxygen species; 2) role of ion channels including gap junctions and potassium channels, especially those associated with the endothelium-derived hyperpolarization mediated signaling, and lastly, 3) mechanism of exocytosis that has considerable oversight by signaling microdomains, especially those associated with the release of von Willebrand factor. When summed, we believe that it is clear that the organization and regulation of signaling microdomains is an essential component to vessel wall function.
Collapse
Affiliation(s)
- Marie Billaud
- Dept. of Molecular Physiology and Biophysics, University of Virginia School of Medicine, PO Box 801394, Charlottesville, VA 22902.
| | | | | | | | | | | |
Collapse
|
41
|
Zhou J, Dong X, Zhou Q, Wang H, Qian Y, Tian W, Ma D, Li X. microRNA expression profiling of heart tissue during fetal development. Int J Mol Med 2014; 33:1250-60. [PMID: 24604530 DOI: 10.3892/ijmm.2014.1691] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 02/18/2014] [Indexed: 01/08/2023] Open
Abstract
microRNAs (miRNAs) are important both in early cardiogenesis and in the process of heart maturation. The aim of this study was to determine the stage-specific expression of miRNAs in human fetal heart in order to identify valuable targets for further study of heart defects. Affymetrix microarrays were used to obtain miRNA expression profiles from human fetal heart tissue at 5, 7, 9 and 23 weeks of gestation. To identify differentially expressed miRNAs at each time-point, linear regression analysis by the R limma algorithm was employed. Hierarchical clustering analysis was conducted with Cluster 3.0 software. Gene Ontology analysis was carried out for miRNAs from different clusters. Commonalities in miRNA families and genomic localization were identified, and the differential expression of selected miRNAs from different clusters was verified by quantitative polymerase chain reaction (qPCR). A total of 703 miRNAs were expressed in human fetal heart. Of these, 288 differentially expressed miRNAs represented 5 clusters with different expression trends. Several clustered miRNAs also shared classification within miRNA families or proximal genomic localization. qPCR confirmed the expression patterns of selected miRNAs. miRNAs within the 5 clusters were predicted to target genes vital for heart development and to be involved in cellular signaling pathways that affect heart structure formation and heart-associated cellular events. In conclusion, to the best of our knowledge, this is the first miRNA expression profiling study of human fetal heart tissue. The stage-specific expression of specific miRNAs suggests potential roles at distinct time-points during fetal heart development.
Collapse
Affiliation(s)
- Jizi Zhou
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P.R. China
| | - Xinran Dong
- Department of Biostatistics and Computational Biology, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Qiongjie Zhou
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P.R. China
| | - Huijun Wang
- Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yanyan Qian
- Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Weidong Tian
- Department of Biostatistics and Computational Biology, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Duan Ma
- Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Xiaotian Li
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
42
|
Burns TA, Dours-Zimmermann MT, Zimmermann DR, Krug EL, Comte-Walters S, Reyes L, Davis MA, Schey KL, Schwacke JH, Kern CB, Mjaatvedt CH. Imbalanced expression of Vcan mRNA splice form proteins alters heart morphology and cellular protein profiles. PLoS One 2014; 9:e89133. [PMID: 24586547 PMCID: PMC3930639 DOI: 10.1371/journal.pone.0089133] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 01/20/2014] [Indexed: 01/09/2023] Open
Abstract
The fundamental importance of the proteoglycan versican to early heart formation was clearly demonstrated by the Vcan null mouse called heart defect (hdf). Total absence of the Vcan gene halts heart development at a stage prior to the heart’s pulmonary/aortic outlet segment growth. This creates a problem for determining the significance of versican’s expression in the forming valve precursors and vascular wall of the pulmonary and aortic roots. This study presents data from a mouse model, Vcan(tm1Zim), of heart defects that results from deletion of exon 7 in the Vcan gene. Loss of exon 7 prevents expression of two of the four alternative splice forms of the Vcan gene. Mice homozygous for the exon 7 deletion survive into adulthood, however, the inability to express the V2 or V0 forms of versican results in ventricular septal defects, smaller cushions/valve leaflets with diminished myocardialization and altered pulmonary and aortic outflow tracts. We correlate these phenotypic findings with a large-scale differential protein expression profiling to identify compensatory alterations in cardiac protein expression at E13.5 post coitus that result from the absence of Vcan exon 7. The Vcan(tm1Zim) hearts show significant changes in the relative abundance of several cytoskeletal and muscle contraction proteins including some previously associated with heart disease. These alterations define a protein fingerprint that provides insight to the observed deficiencies in pre-valvular/septal cushion mesenchyme and the stability of the myocardial phenotype required for alignment of the outflow tract with the heart ventricles.
Collapse
Affiliation(s)
- Tara A. Burns
- Departments of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | | | - Dieter R. Zimmermann
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Edward L. Krug
- Departments of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Susana Comte-Walters
- Department of Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Leticia Reyes
- Departments of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Monica A. Davis
- Departments of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Kevin L. Schey
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - John H. Schwacke
- Departments of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Christine B. Kern
- Departments of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Corey H. Mjaatvedt
- Departments of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
43
|
Schmidt VJ, Hilgert JG, Covi JM, Weis C, Wietbrock JO, de Wit C, Horch RE, Kneser U. High flow conditions increase connexin43 expression in a rat arteriovenous and angioinductive loop model. PLoS One 2013; 8:e78782. [PMID: 24236049 PMCID: PMC3827249 DOI: 10.1371/journal.pone.0078782] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 09/23/2013] [Indexed: 11/25/2022] Open
Abstract
Gap junctions are involved in vascular growth and their expression pattern is modulated in response to hemodynamic conditions. They are clusters of intercellular channels formed by connexins (Cx) of which four subtypes are expressed in the cardiovascular system, namely Cx37, Cx40, Cx43 and Cx45. We hypothesize that high flow conditions affect vascular expression of Cx in vivo. To test this hypothesis, flow hemodynamics and subsequent changes in vascular expression of Cx were studied in an angioinductive rat arteriovenous (AV) loop model. Fifteen days after interposition of a femoral vein graft between femoral artery and vein encased in a fibrin-filled chamber strong neovascularization was evident that emerged predominantly from the graft. Blood flow through the grafted vessel was enhanced ∼4.5-fold accompanied by increased pulsatility exceeding arterial levels. Whereas Cx43 protein expression in the femoral vein is negligible at physiologic flow conditions as judged by immunostaining its expression was enhanced in the endothelium of the venous graft exposed to these hemodynamic changes for 5 days. This was most likely due to enhanced transcription since Cx43 mRNA increased likewise, whereas Cx37 mRNA expression remained unaffected and Cx40 mRNA was reduced. Although enhanced Cx43 expression in regions of high flow in vivo has already been demonstrated, the arteriovenous graft used in the present study provides a reliable model to verify an association between Cx43 expression and high flow conditions in vivo that was selective for this Cx. We conclude that enhancement of blood flow and its oscillation possibly associated with the transition from laminar to more turbulent flow induces Cx43 expression in a vein serving as an AV loop. It is tempting to speculate that this upregulation is involved in the vessel formation occuring in this model as Cx43 was suggested to be involved in angiogenesis.
Collapse
Affiliation(s)
- Volker J. Schmidt
- Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Hand-, Plastic- and Reconstructive Surgery, BG Unfallklinik Ludwigshafen, Universität Heidelberg, Heidelberg, Germany
| | - Johannes G. Hilgert
- Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jennifer M. Covi
- Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Weis
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Johanna O. Wietbrock
- Department of Hand-, Plastic- and Reconstructive Surgery, BG Unfallklinik Ludwigshafen, Universität Heidelberg, Heidelberg, Germany
| | - Cor de Wit
- Department of Physiology, Universität zu Lübeck, Lübeck, Germany
| | - Raymund E. Horch
- Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ulrich Kneser
- Department of Plastic and Hand Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Hand-, Plastic- and Reconstructive Surgery, BG Unfallklinik Ludwigshafen, Universität Heidelberg, Heidelberg, Germany
| |
Collapse
|
44
|
Nam J, Onitsuka I, Hatch J, Uchida Y, Ray S, Huang S, Li W, Zang H, Ruiz-Lozano P, Mukouyama YS. Coronary veins determine the pattern of sympathetic innervation in the developing heart. Development 2013; 140:1475-85. [PMID: 23462468 PMCID: PMC3596991 DOI: 10.1242/dev.087601] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Anatomical congruence of peripheral nerves and blood vessels is well recognized in a variety of tissues. Their physical proximity and similar branching patterns suggest that the development of these networks might be a coordinated process. Here we show that large diameter coronary veins serve as an intermediate template for distal sympathetic axon extension in the subepicardial layer of the dorsal ventricular wall of the developing mouse heart. Vascular smooth muscle cells (VSMCs) associate with large diameter veins during angiogenesis. In vivo and in vitro experiments demonstrate that these cells mediate extension of sympathetic axons via nerve growth factor (NGF). This association enables topological targeting of axons to final targets such as large diameter coronary arteries in the deeper myocardial layer. As axons extend along veins, arterial VSMCs begin to secrete NGF, which allows axons to reach target cells. We propose a sequential mechanism in which initial axon extension in the subepicardium is governed by transient NGF expression by VSMCs as they are recruited to coronary veins; subsequently, VSMCs in the myocardium begin to express NGF as they are recruited by remodeling arteries, attracting axons toward their final targets. The proposed mechanism underlies a distinct, stereotypical pattern of autonomic innervation that is adapted to the complex tissue structure and physiology of the heart.
Collapse
MESH Headings
- Animals
- Axons/physiology
- Cells, Cultured
- Chick Embryo
- Coronary Vessels/embryology
- Coronary Vessels/innervation
- Coronary Vessels/physiology
- Embryo Culture Techniques
- Embryo, Mammalian
- Heart/embryology
- Heart/innervation
- Mice
- Mice, Transgenic
- Models, Biological
- Muscle, Smooth, Vascular/embryology
- Muscle, Smooth, Vascular/innervation
- Muscle, Smooth, Vascular/metabolism
- Pericardium/embryology
- Pericardium/innervation
- Sympathetic Nervous System/embryology
- Sympathetic Nervous System/physiology
Collapse
Affiliation(s)
- Joseph Nam
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA
| | - Izumi Onitsuka
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA
| | - John Hatch
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA
| | - Yutaka Uchida
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA
| | - Saugata Ray
- Development and Aging Program, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Siyi Huang
- Department of Neuroscience, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Wenling Li
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA
| | - Heesuk Zang
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA
| | - Pilar Ruiz-Lozano
- Development and Aging Program, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
- Pediatric Cardiology, Stanford University School of Medicine, 300 Pasteur Drive, Palo Alto, CA 94305, USA
| | - Yoh-suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Building 10/6C103, 10 Center Drive, Bethesda, MD 20892, USA
- Author for correspondence ()
| |
Collapse
|
45
|
Abstract
Epithelial-mesenchymal transition (EMT) is a crucial, evolutionarily conserved process that occurs during development and is essential for shaping embryos. Also implicated in cancer, this morphological transition is executed through multiple mechanisms in different contexts, and studies suggest that the molecular programs governing EMT, albeit still enigmatic, are embedded within developmental programs that regulate specification and differentiation. As we review here, knowledge garnered from studies of EMT during gastrulation, neural crest delamination and heart formation have furthered our understanding of tumor progression and metastasis.
Collapse
Affiliation(s)
- Jormay Lim
- Institute of Molecular Cell Biology, ASTAR, 61 Biopolis Drive, Singapore
| | | |
Collapse
|
46
|
Huang GY, Xie LJ, Linask KL, Zhang C, Zhao XQ, Yang Y, Zhou GM, Wu YJ, Marquez-Rosado L, McElhinney DB, Goldmuntz E, Liu C, Lampe PD, Chatterjee B, Lo CW. Evaluating the role of connexin43 in congenital heart disease: Screening for mutations in patients with outflow tract anomalies and the analysis of knock-in mouse models. J Cardiovasc Dis Res 2012; 2:206-12. [PMID: 22135478 PMCID: PMC3224440 DOI: 10.4103/0975-3583.89804] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background: GJA1 gene encodes a gap junction protein known as connexin 43 (Cx43). Cx43 is abundantly expressed in the ventricular myocardium and in cardiac neural crest cells. Cx43 is proposed to play an important role in human congenital heart disease, as GJA1 knock-out mice die neonatally from outflow tract obstruction. In addition, patients with visceroatrial heterotaxia or hypoplastic left heart syndrome were reported to have point mutations in GJA1 at residues that affect protein kinase phosphorylation and gating of the gap junction channel. However, as these clinical findings were not replicated in subsequent studies, the question remains about the contribution of GJA1 mutations in human congenital heart disease (CHD). Materials and Methods: We analyzed the GJA1 coding sequence in 300 patients with CHD from two clinical centers, focusing on outflow tract anomalies. This included 152 with Tetralogy of Fallot from over 200 patients exhibiting outflow tract anomalies, as well as other structural heart defects including atrioventricular septal defects and other valvar anomalies. Our sequencing analysis revealed only two silent nucleotide substitutions in 8 patients. To further assess the possible role of Cx43 in CHD, we also generated two knock-in mouse models with point mutations at serine residues subject to protein kinase C or casein kinase phosphorylation, sites that are known to regulate gating and trafficking of Cx43, respectively. Results: Both heterozygous and homozygous knock-in mice were long term viable and did not exhibit overt CHD. Conclusion: The combined clinical and knock-in mouse mutant studies indicate GJA1 mutation is not likely a major contributor to CHD, especially those involving outflow tract anomalies.
Collapse
Affiliation(s)
- Guo-Ying Huang
- Children's Hospital of Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Chen CH, Beard RS, Bearden SE. Homocysteine impairs endothelial wound healing by activating metabotropic glutamate receptor 5. Microcirculation 2012; 19:285-95. [PMID: 22221504 DOI: 10.1111/j.1549-8719.2012.00159.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Hcy is an independent risk factor for cerebrovascular disease and cognitive impairment. The purpose of this study was to elucidate the role of mGluR5 in Hcy-mediated impairment of cerebral endothelial wound repair. METHODS Mouse CMVECs (bEnd.3) were used in conjunction with directed pharmacology and shRNA. AutoDock was used to simulate the docking of ligand-receptor interactions. RESULTS Hcy (20 μM) significantly increased Cx43-pS368 by mGluR5- and PKC-dependent mechanisms. Hcy attenuated wound repair by an mGluR5-dependent mechanism over the six-day study period but did not alter cell proliferation in a proliferation assay, suggesting that the attenuation of wound repair may be due to dysfunctional migration in HHcy. Hcy increased the expression of Cx43 and Cx43-pS368 at the wound edge by activating mGluR5. Direct activation of mGluR5, using the specific agonist CHPG, was sufficient to reproduce the results whereas KO of mGluR5 with shRNA, or inhibition with MPEP, blocked the response to Hcy. CONCLUSIONS Inhibition of mGluR5 activation could be a novel strategy for promoting endothelial wound repair in patients with HHcy. Activation of mGluR5 may be a viable strategy for disrupting angiogenesis.
Collapse
Affiliation(s)
- Cheng-Hung Chen
- Department of Biological Sciences, Idaho State University, Pocatello, Idaho 83209-8007, USA
| | | | | |
Collapse
|
48
|
Chen WC, Zhang Y, Ma D, Ma XJ, Shou WN, Huang GY. Bmp2 regulates the interaction between EPDCs and myocytes in cardiac OFT. Med Hypotheses 2012; 79:174-7. [DOI: 10.1016/j.mehy.2012.04.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 04/06/2012] [Accepted: 04/13/2012] [Indexed: 11/30/2022]
|
49
|
Opposing roles of connexin43 in glioma progression. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1818:2058-67. [DOI: 10.1016/j.bbamem.2011.10.022] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 10/17/2011] [Accepted: 10/24/2011] [Indexed: 12/12/2022]
|
50
|
Kovacic JC, Mercader N, Torres M, Boehm M, Fuster V. Epithelial-to-mesenchymal and endothelial-to-mesenchymal transition: from cardiovascular development to disease. Circulation 2012; 125:1795-808. [PMID: 22492947 DOI: 10.1161/circulationaha.111.040352] [Citation(s) in RCA: 339] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Jason C Kovacic
- Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai School of Medicine, One Gustave L. Levy Place, Box 1030, New York, NY 10029, USA.
| | | | | | | | | |
Collapse
|