1
|
Dai B, Liu H, Juan D, Wu K, Cao R. The role of miRNA-29b1 on the hypoxia-induced apoptosis in mammalian cardiomyocytes. Eur J Histochem 2024; 68:4021. [PMID: 38934067 PMCID: PMC11228570 DOI: 10.4081/ejh.2024.4021] [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: 03/12/2024] [Accepted: 05/06/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiomyocyte apoptosis is a complex biological process involving the interaction of many factors and signaling pathways. In hypoxic environment, cardiomyocytes may trigger apoptosis due to insufficient energy supply, increased production of oxygen free radicals, and disturbance of intracellular calcium ion balance. The present research aimed to investigate the role of microRNA-29b1 (miR-29b1) in hypoxia-treated cardiomyocytes and its potential mechanism involved. We established an in vitro ischemia model using AC16 and H9C2 cardiomyocytes through hypoxia treatment (1% O2, 48 h). Cell apoptosis was evaluated by flow cytometry using Annexin V FITC-PI staining assay. Moreover, we used Western blot and immunofluorescence analysis to determine the expression of Bcl-2, Bax caspase-3 and Cx43 proteins. We found that miR-29b1 protected AC16 and H9C2 cells from hypoxia-induced injury as evidence that miR-29b1 attenuated the effects of hypoxia treatment on AC16 and H9C2 cell apoptosis after hypoxia treatment. In conclusion, our findings suggest that miR-29b1 may have potential cardiovascular protective effects during ischemia-related myocardial injury.
Collapse
Affiliation(s)
- Bo Dai
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan.
| | - Hailin Liu
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan.
| | - Dingmin Juan
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan.
| | - Kaize Wu
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan.
| | - Ruhao Cao
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan.
| |
Collapse
|
2
|
Zhao D, Riquelme MA, Guda T, Tu C, Xu H, Gu S, Jiang JX. Connexin hemichannels with prostaglandin release in anabolic function of bone to mechanical loading. eLife 2022; 11:74365. [PMID: 35132953 PMCID: PMC8824479 DOI: 10.7554/elife.74365] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/10/2022] [Indexed: 01/18/2023] Open
Abstract
Mechanical stimulation, such as physical exercise, is essential for bone formation and health. Here, we demonstrate the critical role of osteocytic Cx43 hemichannels in anabolic function of bone in response to mechanical loading. Two transgenic mouse models, R76W and Δ130–136, expressing dominant-negative Cx43 mutants in osteocytes were adopted. Mechanical loading of tibial bone increased cortical bone mass and mechanical properties in wild-type and gap junction-impaired R76W mice through increased PGE2, endosteal osteoblast activity, and decreased sclerostin. These anabolic responses were impeded in gap junction/hemichannel-impaired Δ130–136 mice and accompanied by increased endosteal osteoclast activity. Specific inhibition of Cx43 hemichannels by Cx43(M1) antibody suppressed PGE2 secretion and impeded loading-induced endosteal osteoblast activity, bone formation and anabolic gene expression. PGE2 administration rescued the osteogenic response to mechanical loading impeded by impaired hemichannels. Together, osteocytic Cx43 hemichannels could be a potential new therapeutic target for treating bone loss and osteoporosis.
Collapse
Affiliation(s)
- Dezhi Zhao
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, United States.,School of Life Sciences, Northwestern Polytechnical University, Xian, China
| | - Manuel A Riquelme
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, United States
| | - Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, United States
| | - Chao Tu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, United States.,Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Huiyun Xu
- School of Life Sciences, Northwestern Polytechnical University, Xian, China
| | - Sumin Gu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, United States
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, United States
| |
Collapse
|
3
|
Ramachandra Rao S, Skelton LA, Wu F, Onysk A, Spolnik G, Danikiewicz W, Butler MC, Stacks DA, Surmacz L, Mu X, Swiezewska E, Pittler SJ, Fliesler SJ. Retinal Degeneration Caused by Rod-Specific Dhdds Ablation Occurs without Concomitant Inhibition of Protein N-Glycosylation. iScience 2020; 23:101198. [PMID: 32526701 PMCID: PMC7287266 DOI: 10.1016/j.isci.2020.101198] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/10/2020] [Accepted: 05/20/2020] [Indexed: 12/16/2022] Open
Abstract
Dehydrodolichyl diphosphate synthase (DHDDS) catalyzes the committed step in dolichol synthesis. Recessive mutations in DHDDS cause retinitis pigmentosa (RP59), resulting in blindness. We hypothesized that rod photoreceptor-specific ablation of Dhdds would cause retinal degeneration due to diminished dolichol-dependent protein N-glycosylation. Dhddsflx/flx mice were crossed with rod-specific Cre recombinase-expressing (Rho-iCre75) mice to generate rod-specific Dhdds knockout mice (Dhddsflx/flx iCre+). In vivo morphological and electrophysiological evaluation of Dhddsflx/flx iCre+ retinas revealed mild retinal dysfunction at postnatal (PN) 4 weeks, compared with age-matched controls; however, rapid photoreceptor degeneration ensued, resulting in almost complete loss of rods and cones by PN 6 weeks. Retina dolichol levels were markedly decreased by PN 4 weeks in Dhddsflx/flx iCre+ mice, relative to controls; despite this, N-glycosylation of retinal proteins, including opsin (the dominant rod-specific glycoprotein), persisted in Dhddsflx/flx iCre+ mice. These findings challenge the conventional mechanistic view of RP59 as a congenital disorder of glycosylation. Deletion of Dhdds in rod cells caused rapid retinal degeneration in mice Retinal dolichol levels markedly decreased before onset of degeneration Protein N-glycosylation was uncompromised despite Dhdds deletion Degeneration also involved gliosis, microglial activation, and phagoptosis
Collapse
Affiliation(s)
- Sriganesh Ramachandra Rao
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14209, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY 142015, USA; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14203, USA
| | - Lara A Skelton
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14209, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY 142015, USA; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14203, USA
| | - Fuguo Wu
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14209, USA; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14203, USA; New York State Center of Excellence in Bioinformatics and Life Sciences, State University of New York- University at Buffalo, Buffalo, NY 14203, USA
| | - Agnieszka Onysk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Grzegorz Spolnik
- Institute of Organic Chemistry, Polish Academy of Sciences, Warsaw 02106, Poland
| | - Witold Danikiewicz
- Institute of Organic Chemistry, Polish Academy of Sciences, Warsaw 02106, Poland
| | - Mark C Butler
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14209, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY 142015, USA
| | - Delores A Stacks
- Department of Optometry and Vision Science, Vision Science Research Center, School of Optometry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Liliana Surmacz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Xiuqian Mu
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14209, USA; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14203, USA; New York State Center of Excellence in Bioinformatics and Life Sciences, State University of New York- University at Buffalo, Buffalo, NY 14203, USA
| | - Ewa Swiezewska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Steven J Pittler
- Department of Optometry and Vision Science, Vision Science Research Center, School of Optometry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Steven J Fliesler
- Department of Ophthalmology/Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14209, USA; Research Service, VA Western NY Healthcare System, Buffalo, NY 142015, USA; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York- University at Buffalo, Buffalo, NY 14203, USA.
| |
Collapse
|
4
|
Wang J, Yang ZY, Guo YF, Kuang JY, Bian XW, Yu SC. Targeting different domains of gap junction protein to control malignant glioma. Neuro Oncol 2019; 20:885-896. [PMID: 29106645 DOI: 10.1093/neuonc/nox207] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
A rational treatment strategy for glioma, the most common primary central nervous system tumor, should focus on early invasive growth and resistance to current therapeutics. Connexin 43 (Cx43), a gap junction protein, plays important roles not only in the development of the central nervous system and but also in the progression of glioma. The different structural domains of Cx43, including extracellular loops, transmembrane domains, and an intracellular carboxyl terminal, have distinct functions in the invasion and proliferation of gliomas. Targeting these domains of Cx43, which is expressed in distinct patterns in the heterogeneous glioma cell population, can inhibit tumor cell invasion and new tumor formation. Thus, this review summarizes the structural characteristics of Cx43, the effects of regulating different Cx43 domains on the biological characteristics of glioma cells, intervention strategies targeting different domains of Cx43, and future research directions.
Collapse
Affiliation(s)
- Jun Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of the Ministry of Education, Chongqing, China
| | - Ze-Yu Yang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of the Ministry of Education, Chongqing, China
| | - Yu-Feng Guo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of the Ministry of Education, Chongqing, China
| | - Jing-Ya Kuang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of the Ministry of Education, Chongqing, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of the Ministry of Education, Chongqing, China
| | - Shi-Cang Yu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, The Third Military Medical University, Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of the Ministry of Education, Chongqing, China
| |
Collapse
|
5
|
Cotter ML, Boitano S, Lampe PD, Solan JL, Vagner J, Ek-Vitorin JF, Burt JM. The lipidated connexin mimetic peptide SRPTEKT- Hdc is a potent inhibitor of Cx43 channels with specificity for the pS368 phospho-isoform. Am J Physiol Cell Physiol 2019; 317:C825-C842. [PMID: 31365296 PMCID: PMC6850999 DOI: 10.1152/ajpcell.00160.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/16/2019] [Accepted: 07/19/2019] [Indexed: 11/22/2022]
Abstract
Connexin (Cx) mimetic peptides derived from extracellular loop II sequences (e.g., Gap27: SRPTEKTIFII; Peptide5: VDCFLSRPTEKT) have been used as reversible, Cx-specific blockers of hemichannel (HCh) and gap junction channel (GJCh) function. These blockers typically require high concentrations (~5 µM, <1 h for HCh; ~100 µM, >1 h for GJCh) to achieve inhibition. We have shown that addition of a hexadecyl (Hdc) lipid tail to the conserved SRPTEKT peptide sequence (SRPTEKT-Hdc) results in a novel, highly efficacious, and potent inhibitor of mechanically induced Ca2+-wave propagation (IC50 64.8 pM) and HCh-mediated dye uptake (IC50 45.0 pM) in Madin-Darby canine kidney cells expressing rat Cx43 (MDCK43). The lack of similar effect on dye coupling (NBD-MTMA) suggested channel conformation-specific inhibition. Here we report that SRPTEKT-Hdc inhibition of Ca2+-wave propagation, dye coupling, and dye uptake depended on the functional configuration of Cx43 as determined by phosphorylation at serine 368 (S368). Ca2+-wave propagation was enhanced in MDCK cells expressing single-site mutants of Cx43 that mimicked (MDCK43-S368D) or favored (MDCK43-S365A) phosphorylation at S368. Furthermore, SRPTEKT-Hdc potently inhibited GJCh-mediated Ca2+-wave propagation (IC50 230.4 pM), dye coupling, and HCh-mediated dye uptake in MDCK43-S368D and -S365A cells. In contrast, Ca2+-wave propagation, dye coupling, and dye uptake were largely unaffected (IC50 12.3 μM) by SRPTEKT-Hdc in MDCK43-S368A and -S365D cells, mutations that mimic or favor dephosphorylation at S368. Together, these data indicate that SRPTEKT-Hdc is a potent inhibitor of physiological Ca2+-wave signaling mediated specifically by the pS368 phosphorylated form of Cx43.
Collapse
Affiliation(s)
- Maura L Cotter
- Department of Physiology, University of Arizona, Tucson, Arizona
| | - Scott Boitano
- Department of Physiology, University of Arizona, Tucson, Arizona
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona
- Bio5 Institute, University of Arizona, Tucson, Arizona
| | - Paul D Lampe
- Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Joell L Solan
- Translational Research Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Josef Vagner
- Bio5 Institute, University of Arizona, Tucson, Arizona
- Department of Pharmacology, University of Arizona, Tucson, Arizona
| | | | - Janis M Burt
- Department of Physiology, University of Arizona, Tucson, Arizona
| |
Collapse
|
6
|
Zhou Y, Chen Y, Hu X, Guo J, Shi H, Yu G, Tang Z. Icariin attenuate microcystin-LR-induced gap junction injury in Sertoli cells through suppression of Akt pathways. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 251:328-337. [PMID: 31091496 DOI: 10.1016/j.envpol.2019.04.114] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 03/27/2019] [Accepted: 04/24/2019] [Indexed: 06/09/2023]
Abstract
Microcystin-leucine-arginine (MC-LR) can cause male reproductive disorder. However, the underlying mechanism are not yet entirely elucidated. In this study, we aimed to investigated the effects of MC-LR on the integrity of blood-testis barrier (BTB) and the related molecular mechanisms. Both in vivo and in vitro experiments revealed that MC-LR caused disruption of BTB and gap junctions between Sertoli cells respectively, which was paralleled by the alteration of connexin43 (Cx43). Our data demonstrated that MC-LR decreased gap junction intercellular communication (GJIC) and impaired Cx43 expression by activating the phosphatidylinositol 3-kinase/Akt cascades. In addition, a possible protective effect of Icariin (ICA), a flavonoid isolated from Chinese medicinal herb, against MC-LR toxicity was investigated. The ICA prevented the degradation of GJIC and impairment of Cx43 induced by MC-LR via suppressing the Akt pathway. Together, our results confirmed that the expression of Cx43 induced by MC-LR was regulated in vivo and in vitro, which was involved in the destruction of BTB. Additionally, ICA seems to be able to mitigate the MC-LR toxic effects.
Collapse
Affiliation(s)
- Yuan Zhou
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Yu Chen
- Research Center of Endocrine and Metabolic Diseases, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Xueqin Hu
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Jun Guo
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Hao Shi
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Guang Yu
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Zongxiang Tang
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China.
| |
Collapse
|
7
|
Zhang Q, Jia GJ, Zhang GB, Wang L, Wu Z, Jia W, Hao SY, Ni M, Li D, Wang K, Zhang JT. A Logistic Regression Model for Detecting the Presence of Malignant Progression in Atypical Meningiomas. World Neurosurg 2019; 126:e392-e401. [PMID: 30822595 DOI: 10.1016/j.wneu.2019.02.062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 02/04/2019] [Accepted: 02/06/2019] [Indexed: 01/07/2023]
Abstract
OBJECTIVE To develop a method to distinguish atypical meningiomas (AMs) with malignant progression (MP) from primary AMs without a clinical history. METHODS The clinical, radiologic, and pathologic data of 33 previously Simpson grade I resected (if any) as well as no radiotherapy treated intracranial AMs between January 2008 and December 2015 were reviewed. Immunohistochemical staining for connexin 43 (Cx43) and Ki-67 was performed. Descriptive analysis and univariate and multivariate logistic regression analyses were used to explore independent predictors of MP. A multivariable logistic model was developed to estimate the risk of MP, and its diagnostic value was determined from a receiver operating characteristic curve. RESULTS There were 11 AMs (33.3%) with histopathologically confirmed MP from benign meningiomas. The other 22 (66.7%) were initially diagnosed AMs with no histopathologically confirmed MP during a median 60.5 months (range, 42-126 months) of follow-up. Univariate and multivariate logistic analyses showed that irregular tumor shape (P = 0.010) and low Cx43 expression (P = 0.010) were independent predictors of the presence of MP, and the predicted probability was calculated by the following formula: P = 1/[1+exp.{1.218-(3.202×Shape)+(3.814×Cx43)}]. P > 0.5 for an irregularly shaped (score 1) AM with low Cx43 expression (score 0) indicated a high probability of MP. The sensitivity, specificity, positive predictive value, negative predictive value, and overall predictive accuracy were 63.6, 95.6, 87.5, 84.0, and 84.8%, respectively. CONCLUSIONS Low Cx43 expression and irregular tumor shape were independent predictors of the presence of MP. The relevant logistic regression model was found to be effective in distinguishing MP-AMs from primary AMs.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Gui-Jun Jia
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Guo-Bin Zhang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Liang Wang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Wang Jia
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Shu-Yu Hao
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Ming Ni
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Da Li
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Ke Wang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China
| | - Jun-Ting Zhang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China; China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China; Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China; Beijing Key Laboratory of Brain Tumor, Beijing, People's Republic of China.
| |
Collapse
|
8
|
Phosphorylation-Dependent Intra-Domain Interaction of the Cx37 Carboxyl-Terminus Controls Cell Survival. Cancers (Basel) 2019; 11:cancers11020188. [PMID: 30736283 PMCID: PMC6406260 DOI: 10.3390/cancers11020188] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/01/2019] [Accepted: 02/02/2019] [Indexed: 02/07/2023] Open
Abstract
Differential phosphorylation of the carboxyl-terminus of connexin 37 (Cx37-CT) regulates phenotypic switching between cell growth phenotypes (cell death, cell cycle arrest, proliferation). The specific phosphorylation events in the Cx37-CT that are necessary for these growth regulatory effects are currently unknown. Through the combined use of deletion and site specific (de)phospho-mimetic Cx37-CT mutants, our data suggest a phosphorylation-dependent interaction between the mid-tail (aa 273⁻317) and end-tail (aa 318⁻333) portions of the Cx37-CT that regulates cell survival. As detected by mass spectrometry, Cx37 was phosphorylated at serines 275, 321, and 328; phosphomimetic mutations of these sites resulted in cell death when expressed in rat insulinoma cells. Alanine substitution at S328, but not at S275 or S321, also triggered cell death. Cx37-S275D uniquely induced the death of only low density, non-contact forming cells, but neither hemichannel open probability nor channel conductance distinguished death-inducing mutants. As channel function is necessary for cell death, together the data suggest that the phosphorylation state of the Cx37-CT controls an intra-domain interaction within the CT that modifies channel function and induces cell death.
Collapse
|
9
|
O'Brien J, Bloomfield SA. Plasticity of Retinal Gap Junctions: Roles in Synaptic Physiology and Disease. Annu Rev Vis Sci 2018; 4:79-100. [DOI: 10.1146/annurev-vision-091517-034133] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Electrical synaptic transmission via gap junctions underlies direct and rapid neuronal communication in the central nervous system. The diversity of functional roles played by electrical synapses is perhaps best exemplified in the vertebrate retina, in which gap junctions are expressed by each of the five major neuronal types. These junctions are highly plastic; they are dynamically regulated by ambient illumination and circadian rhythms acting through light-activated neuromodulators. The networks formed by electrically coupled neurons provide plastic, reconfigurable circuits positioned to play key and diverse roles in the transmission and processing of visual information at every retinal level. Recent work indicates gap junctions also play a role in the progressive cell death and aberrant activity seen in various pathological conditions of the retina. Gap junctions thus form potential targets for novel neuroprotective therapies in the treatment of neurodegenerative retinal diseases such as glaucoma and ischemic retinopathies.
Collapse
Affiliation(s)
- John O'Brien
- Department of Ophthalmology and Visual Science, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Stewart A. Bloomfield
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, NY 10036, USA
| |
Collapse
|
10
|
Ma Y, Han X, de Castro RB, Zhang P, Zhang K, Hu Z, Qin L. Analysis of the bystander effect in cone photoreceptors via a guided neural network platform. SCIENCE ADVANCES 2018; 4:eaas9274. [PMID: 29750200 PMCID: PMC5942910 DOI: 10.1126/sciadv.aas9274] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/21/2018] [Indexed: 06/08/2023]
Abstract
The mammalian retina system consists of a complicated photoreceptor structure, which exhibits extensive random synaptic connections. To study retinal development and degeneration, various experimental models have been used previously, but these models are often uncontrollable, are difficult to manipulate, and do not provide sufficient similarity or precision. Therefore, the mechanisms in many retinal diseases remain unclear because of the limited capability in observing the progression and molecular driving forces. For example, photoreceptor degeneration can spread to surrounding healthy photoreceptors via a phenomenon known as the bystander effect; however, no in-depth observations can be made to decipher the molecular mechanisms or the pathways that contribute to the spreading. It is then necessary to build dissociated neural networks to investigate the communications with controllability of cells and their treatment. We developed a neural network chip (NN-Chip) to load single neurons into highly ordered microwells connected by microchannels for synapse formation to build the neural network. By observing the distribution of apoptosis spreading from light-induced apoptotic cones to the surrounding cones, we demonstrated convincing evidence of the existence of a cone-to-cone bystander killing effect. Combining the NN-Chip with microinjection technology, we also found that the gap junction protein connexin 36 (Cx36) is critical for apoptosis spreading and the bystander effect in cones. In addition, our unique NN-Chip platform provides a quantitative, high-throughput tool for investigating signaling mechanisms and behaviors in neurons and opens a new avenue for screening potential drug targets to cure retinal diseases.
Collapse
Affiliation(s)
- Yuan Ma
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- College of Materials Sciences and Optoelectronics, University of Chinese Academy of Sciences, Beijing 100049, China
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Xin Han
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ricardo Bessa de Castro
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
- College of Engineering, Swansea University, Singleton Park, Swansea SA2 8PP, UK
| | - Pengchao Zhang
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kai Zhang
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Zhongbo Hu
- College of Materials Sciences and Optoelectronics, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lidong Qin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
11
|
Multiple and complex influences of connexins and pannexins on cell death. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017. [PMID: 28625689 DOI: 10.1016/j.bbamem.2017.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cell death is a fundamental process for organogenesis, immunity and cell renewal. During the last decades a broad range of molecular tools were identified as important players for several different cell death pathways (apoptosis, pyroptosis, necrosis, autosis…). Aside from these direct regulators of cell death programs, several lines of evidence proposed connexins and pannexins as potent effectors of cell death. In the present review we discussed the potential roles played by connexins, pannexins and innexins in the different cell death programs at different scales from gap junction intercellular communication to protein-protein interactions. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
|
12
|
Belousov AB, Fontes JD, Freitas-Andrade M, Naus CC. Gap junctions and hemichannels: communicating cell death in neurodevelopment and disease. BMC Cell Biol 2017; 18:4. [PMID: 28124625 PMCID: PMC5267333 DOI: 10.1186/s12860-016-0120-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Gap junctions are unique membrane channels that play a significant role in intercellular communication in the developing and mature central nervous system (CNS). These channels are composed of connexin proteins that oligomerize into hexamers to form connexons or hemichannels. Many different connexins are expressed in the CNS, with some specificity with regard to the cell types in which distinct connexins are found, as well as the timepoints when they are expressed in the developing and mature CNS. Both the main neuronal Cx36 and glial Cx43 play critical roles in neurodevelopment. These connexins also mediate distinct aspects of the CNS response to pathological conditions. An imbalance in the expression, translation, trafficking and turnover of connexins, as well as mutations of connexins, can impact their function in the context of cell death in neurodevelopment and disease. With the ever-increasing understanding of connexins in the brain, therapeutic strategies could be developed to target these membrane channels in various neurological disorders.
Collapse
Affiliation(s)
- Andrei B Belousov
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, The University of Kansas, Kansas City, KS, 66160, USA
| | - Joseph D Fontes
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, The University of Kansas, Kansas City, KS, 66160, USA
| | - Moises Freitas-Andrade
- Department of Cellular & Physiological Sciences, Faculty of Medicine, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Christian C Naus
- Department of Cellular & Physiological Sciences, Faculty of Medicine, Life Sciences Institute, The University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
13
|
A reaction-diffusion model for radiation-induced bystander effects. J Math Biol 2016; 75:341-372. [PMID: 28035423 DOI: 10.1007/s00285-016-1090-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 12/08/2016] [Indexed: 12/29/2022]
Abstract
We develop and analyze a reaction-diffusion model to investigate the dynamics of the lifespan of a bystander signal emitted when cells are exposed to radiation. Experimental studies by Mothersill and Seymour 1997, using malignant epithelial cell lines, found that an emitted bystander signal can still cause bystander effects in cells even 60 h after its emission. Several other experiments have also shown that the signal can persist for months and even years. Also, bystander effects have been hypothesized as one of the factors responsible for the phenomenon of low-dose hyper-radiosensitivity and increased radioresistance (HRS/IRR). Here, we confirm this hypothesis with a mathematical model, which we fit to Joiner's data on HRS/IRR in a T98G glioma cell line. Furthermore, we use phase plane analysis to understand the full dynamics of the signal's lifespan. We find that both single and multiple radiation exposure can lead to bystander signals that either persist temporarily or permanently. We also found that, in an heterogeneous environment, the size of the domain exposed to radiation and the number of radiation exposures can determine whether a signal will persist temporarily or permanently. Finally, we use sensitivity analysis to identify those cell parameters that affect the signal's lifespan and the signal-induced cell death the most.
Collapse
|
14
|
Belousov AB, Fontes JD. Role of neuronal gap junctions in NMDA receptor-mediated excitotoxicity and ischemic neuronal death. Neural Regen Res 2016; 11:75-6. [PMID: 26981086 PMCID: PMC4774234 DOI: 10.4103/1673-5374.169630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Affiliation(s)
- Andrei B Belousov
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Joseph D Fontes
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
15
|
Gao K, Chi Y, Zhang X, Zhang H, Li G, Sun W, Takeda M, Yao J. A novel TXNIP-based mechanism for Cx43-mediated regulation of oxidative drug injury. J Cell Mol Med 2015; 19:2469-80. [PMID: 26154105 PMCID: PMC4594688 DOI: 10.1111/jcmm.12641] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 05/28/2015] [Indexed: 01/28/2023] Open
Abstract
Gap junctions (GJs) play an important role in the regulation of cell response to many drugs. However, little is known about their mechanisms. Using an in vitro model of cytotoxicity induced by geneticin (G418), we explored the potential signalling mechanisms involved. Incubation of cells with G418 resulted in cell death, as indicated by the change in cell morphology, loss of cell viability and activation of caspase-3. Before the onset of cell injury, G418 induced reactive oxygen species (ROS) generation, activated oxidative sensitive kinase P38 and caused a shift of connexin 43 (Cx43) from non-phosphorylated form to hyperphosphorylated form. These changes were largely prevented by antioxidants, suggesting an implication of oxidative stress. Downregulation of Cx43 with inhibitors or siRNA suppressed the expression of thioredoxin-interacting protein (TXNIP), activated Akt and protected cells against the toxicity of G418. Further analysis revealed that inhibition of TXNIP with siRNA activated Akt and reproduced the protective effect of Cx43-inhibiting agents, whereas suppression of Akt sensitized cells to the toxicity of G418. Furthermore, interference of TXNIP/Akt also affected puromycin- and adriamycin-induced cell injury. Our study thus characterized TXNIP as a presently unrecognized molecule implicated in the regulatory actions of Cx43 on oxidative drug injury. Targeting Cx43/TXNIP/Akt signalling cascade might be a promising approach to modulate cell response to drugs.
Collapse
Affiliation(s)
- Kun Gao
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan.,Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yuan Chi
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Xiling Zhang
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Hui Zhang
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Gang Li
- Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan.,Department of Urology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Wei Sun
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Masayuki Takeda
- Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Jian Yao
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| |
Collapse
|
16
|
Fontes JD, Ramsey J, Polk JM, Koop A, Denisova JV, Belousov AB. Death of Neurons following Injury Requires Conductive Neuronal Gap Junction Channels but Not a Specific Connexin. PLoS One 2015; 10:e0125395. [PMID: 26017008 PMCID: PMC4446213 DOI: 10.1371/journal.pone.0125395] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 03/24/2015] [Indexed: 01/01/2023] Open
Abstract
Pharmacological blockade or genetic knockout of neuronal connexin 36 (Cx36)-containing gap junctions reduces neuronal death caused by ischemia, traumatic brain injury and NMDA receptor (NMDAR)-mediated excitotoxicity. However, whether Cx36 gap junctions contribute to neuronal death via channel-dependent or channel-independent mechanism remains an open question. To address this, we manipulated connexin protein expression via lentiviral transduction of mouse neuronal cortical cultures and analyzed neuronal death twenty-four hours following administration of NMDA (a model of NMDAR excitotoxicity) or oxygen-glucose deprivation (a model of ischemic injury). In cultures prepared from wild-type mice, over-expression and knockdown of Cx36-containing gap junctions augmented and prevented, respectively, neuronal death from NMDAR-mediated excitotoxicity and ischemia. In cultures obtained form from Cx36 knockout mice, re-expression of functional gap junction channels, containing either neuronal Cx36 or non-neuronal Cx43 or Cx31, resulted in increased neuronal death following insult. In contrast, the expression of communication-deficient gap junctions (containing mutated connexins) did not have this effect. Finally, the absence of ethidium bromide uptake in non-transduced wild-type neurons two hours following NMDAR excitotoxicity or ischemia suggested the absence of active endogenous hemichannels in those neurons. Taken together, these results suggest a role for neuronal gap junctions in cell death via a connexin type-independent mechanism that likely relies on channel activities of gap junctional complexes among neurons. A possible contribution of gap junction channel-permeable death signals in neuronal death is discussed.
Collapse
Affiliation(s)
- Joseph D. Fontes
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Jon Ramsey
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Jeremy M Polk
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Andre Koop
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Janna V. Denisova
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Andrei B. Belousov
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| |
Collapse
|
17
|
Akopian A, Atlasz T, Pan F, Wong S, Zhang Y, Völgyi B, Paul DL, Bloomfield SA. Gap junction-mediated death of retinal neurons is connexin and insult specific: a potential target for neuroprotection. J Neurosci 2014; 34:10582-91. [PMID: 25100592 PMCID: PMC4200109 DOI: 10.1523/jneurosci.1912-14.2014] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 06/06/2014] [Accepted: 06/27/2014] [Indexed: 01/02/2023] Open
Abstract
Secondary cell death via gap junctions (GJs) plays a role in the propagation of neuronal loss under a number of degenerative disorders. Here, we examined the role of GJs in neuronal death in the retina, which has arguably the most diverse expression of GJs in the CNS. Initially, we induced apoptotic death by injecting single retinal ganglion cells and glia with cytochrome C and found that this resulted in the loss of neighboring cells to which they were coupled via GJs. We next found that pharmacological blockade of GJs eradicated nearly all amacrine cell loss and reduced retinal ganglion cell loss by ∼70% after induction of either excitotoxic or ischemic insult conditions. These data indicate that the GJ-mediated secondary cell death was responsible for the death of most cells. Whereas genetic deletion of the GJ subunit Cx36 increased cell survivability by ∼50% under excitotoxic condition, cell loss in Cx45 knock-out mouse retinas was similar to that seen in wild-type mice. In contrast, ablation of Cx45 reduced neuronal loss by ∼50% under ischemic insult, but ablation of Cx36 offered no protection. Immunolabeling of the connexins showed differential changes in protein expression consistent with their differing roles in propagating death signals under the two insults. These data indicate that secondary cell death is mediated by different cohorts of GJs dependent on the connexins they express and the type of initial insult. Our results suggest that targeting specific connexins offers a novel therapeutic strategy to reduce progressive cell loss under different neurodegenerative conditions.
Collapse
Affiliation(s)
- Abram Akopian
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, New York 10036, Department of Physiology & Neuroscience, New York University School of Medicine, New York, New York 10016
| | - Tamas Atlasz
- Department of Ophthalmology, New York University School of Medicine, New York, New York 10016, and
| | - Feng Pan
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, New York 10036, Department of Physiology & Neuroscience, New York University School of Medicine, New York, New York 10016
| | - Sze Wong
- Department of Physiology & Neuroscience, New York University School of Medicine, New York, New York 10016
| | - Yi Zhang
- Department of Physiology & Neuroscience, New York University School of Medicine, New York, New York 10016
| | - Béla Völgyi
- Department of Ophthalmology, New York University School of Medicine, New York, New York 10016, and
| | - David L Paul
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115
| | - Stewart A Bloomfield
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, New York 10036, Department of Physiology & Neuroscience, New York University School of Medicine, New York, New York 10016, Department of Ophthalmology, New York University School of Medicine, New York, New York 10016, and
| |
Collapse
|
18
|
Carette D, Gilleron J, Chevallier D, Segretain D, Pointis G. Connexin a check-point component of cell apoptosis in normal and physiopathological conditions. Biochimie 2014; 101:1-9. [DOI: 10.1016/j.biochi.2013.11.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 11/18/2013] [Indexed: 12/16/2022]
|
19
|
Warzych E, Cieslak A, Madeja ZE, Pawlak P, Wolc A, Lechniak D. Multifactorial analysis of the follicular environment is predictive of oocyte morphology in cattle. J Reprod Dev 2013; 60:1-8. [PMID: 24256920 PMCID: PMC3963297 DOI: 10.1262/jrd.2013-086] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Numerous attempts have been recently made in the search for a reliable, fast and
noninvasive assay for selection of oocytes suitable for in vitro embryo
production. Potential markers have been described in the follicle such as follicular fluid
(FF) or cumulus cells (CCs). However, the reported findings are contradictory, which may
reflect the complexity of metabolism of the ovarian follicle. In the present experiment, a
data set from individual follicles of known diameter was obtained: cumulus-oocyte complex
(COC) morphology, fatty acid composition and glucose concentration in FF as well as
apoptotic index in CCs. The obtained data was statistically analyzed either separately
(univariate analysis) or simultaneously (multivariate analysis) to examine its predictive
value in morphology assessment of bovine COCs. Although the univariate analysis yielded a
complex relation system of the selected parameters, no clear outcome could be established.
In multivariate analysis, the concentration of the four fatty acids (C16:0, C16:1,
C18:1cis9, C22:5n3) and Δ9-desaturase (16) as well as elongase activities were
selected as covariates. This allowed prediction of the morphology of a COC with an
accuracy of 72%, which is the most interesting finding of the experiment. The present
study indicates that the multifactorial model comprising of selected parameters related to
the follicle appeared more effective in predicting the morphology of a bovine COC, which
may improve the effectiveness of in vitro production systems.
Collapse
Affiliation(s)
- Ewelina Warzych
- Department of Genetics and Animal Breeding, Poznan University of Life Sciences, 60-637 Poznan, Poland
| | | | | | | | | | | |
Collapse
|
20
|
Neuronal gap junction coupling as the primary determinant of the extent of glutamate-mediated excitotoxicity. J Neural Transm (Vienna) 2013; 121:837-46. [PMID: 24178243 DOI: 10.1007/s00702-013-1109-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 10/16/2013] [Indexed: 01/12/2023]
Abstract
In the mammalian central nervous system (CNS), coupling of neurons by gap junctions (electrical synapses) increases during early postnatal development, then decreases, but increases in the mature CNS following neuronal injury, such as ischemia, traumatic brain injury and epilepsy. Glutamate-dependent neuronal death also occurs in the CNS during development and neuronal injury, i.e., at the time when neuronal gap junction coupling is increased. Here, we review our recent studies on regulation of neuronal gap junction coupling by glutamate in developing and injured neurons and on the role of gap junctions in neuronal cell death. A modified model of the mechanisms of glutamate-dependent neuronal death is discussed, which includes neuronal gap junction coupling as a critical part of these mechanisms.
Collapse
|
21
|
Kameritsch P, Khandoga N, Pohl U, Pogoda K. Gap junctional communication promotes apoptosis in a connexin-type-dependent manner. Cell Death Dis 2013; 4:e584. [PMID: 23579271 PMCID: PMC3641328 DOI: 10.1038/cddis.2013.105] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 01/31/2013] [Accepted: 02/21/2013] [Indexed: 12/28/2022]
Abstract
Gap junctions (GJs) have been described to modulate cell death and survival. It still remains unclear whether this effect requires functional GJ channels or depends on channel-independent effects of connexins (Cx), the constituents of GJs. Therefore, we analysed the apoptotic response to streptonigrin (SN, intrinsic apoptotic pathway) or to α-Fas (extrinsic apoptotic pathway) in HeLa cells expressing Cx43 as compared with empty vector-transfected (CTL) cells. Apoptosis assessed by annexin V-fluorescein isothiocyanate/propidium iodide staining was significantly higher in HeLa-Cx43 compared with HeLa-CTL cells. Moreover, the cleavage of caspase-7 or Parp occurred earlier in HeLa-Cx43 than in HeLa-CTL cells. Comparative analysis of the effect of two further (endothelial) Cx (Cx37 and Cx40) on apoptosis revealed that apoptosis was highest in HeLa-Cx43 and lowest in HeLa-Cx37 cells, and correlated with the GJ permeability (assessed by spreading of a GJ-permeable dye and locally induced Ca(2+) signals). Pharmacologic inhibition of GJ formation in HeLa-Cx43 cells reduced apoptosis significantly. The role of GJ communication was further analysed by the expression of truncated Cx43 proteins with and without channel-forming capacity. Activation of caspases was higher in cells expressing the channel-building part (HeLa-Cx43NT-GFP) than in cells expressing the channel-incompetent C-terminal part of Cx43 (HeLa-Cx43CT-GFP) only. A hemichannel-dependent release and, hence, paracrine effect of proapoptotic signals could be excluded since the addition of a peptide (Pep)-blocking Cx43-dependent hemichannels (but not GJs) did not reduce apoptosis in HeLa-Cx43 cells. Treatment with SN resulted in a significant higher increase of the intracellular free Ca(2+) concentration in HeLa-Cx43 and HeLa-Cx43NT-GFP cells compared with HeLa-CTL or HeLa-Cx43CT-GFP cells, suggesting that Ca(2+) or a Ca(2+)-releasing agent could play a signalling role. Blocking of inositol triphosphate receptors reduced the SN-induced Ca(2+) increase as well as the increase in apoptosis. Our observations suggest that Cx43 and Cx40 but not Cx37 promote apoptosis via gap junctional transfer of pro-apoptotic signals between cells.
Collapse
Affiliation(s)
- P Kameritsch
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, Munich, Germany.
| | | | | | | |
Collapse
|
22
|
Wang X, Ma A, Zhu W, Zhu L, Zhao Y, Xi J, Zhang X, Zhao B, Becker DL. The role of connexin 43 and hemichannels correlated with the astrocytic death following ischemia/reperfusion insult. Cell Mol Neurobiol 2013; 33:401-10. [PMID: 23328809 PMCID: PMC11498013 DOI: 10.1007/s10571-013-9906-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 01/03/2013] [Indexed: 11/24/2022]
Abstract
The aim of this study was to investigate the role of connexin 43 (Cx43) and its hemichannel (HC1) in the death of astrocytes following ischemia/reperfusion (IR) or oxygen-glucose deprivation/reoxygenation (OGDR) insult. Wistar rats had their bilateral common carotid artery clamped for 1.5 h followed by 0, 4, and 24 h of reperfusion (n = 8 for each time point), respectively. All rats were sacrificed and Cx43, HC1, and caspase 3 (Casp3) in cerebral ischemic tissues were examined by immunohistochemistry and western blotting. Astrocytes cell line, astrocytes transduced with a retroviral empty vector (Psup astrocyte), or a Cx43-specific shRNA construct (shRNA astrocytes) were treated with OGDR insult for various periods. The viability of astrocytes was assessed by MTT assay. The expression of Cx43, HC1, and Casp3 was detected with western blotting. The results showed that the expression of Cx43, HC1, and Casp3 in rats' brain, astrocytes, and Psup astrocytes was significantly increased after 4 h of IR/OGDR and recovered on 24 h of the insult. Cell viability decreased after 4 h of the insult whereas the cell viability increased on 24 h after the insult. In contrast, the expression of Cx43, HC1, Casp3, and cell viability had no statistical differences in the null Cx43 gene-shRNA transfected astrocytes after the treatment of OGDR. The results suggest that Cx43 and HC1 are likely to play the pivotal roles in the mediation of the astrocytic death.
Collapse
Affiliation(s)
- Xueyu Wang
- Division of Neurology, Department of Pediatrics, Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, 250021 China
| | - Aihua Ma
- Division of Neurology, Department of Pediatrics, Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, 250021 China
| | - Weiwei Zhu
- Department of Pediatrics, Central Hospital of Jinan Affiliated to Shandong University, Jinan, China
| | - Liping Zhu
- Division of Neurology, Department of Pediatrics, Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, 250021 China
| | - Yutian Zhao
- School of Pharmacy, Cardiff University, Cardiff, UK
| | - Jiashui Xi
- Division of Neurology, Department of Pediatrics, Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, 250021 China
| | - Xinying Zhang
- Division of Neurology, Department of Pediatrics, Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, 250021 China
| | - Bojun Zhao
- Division of Neurology, Department of Pediatrics, Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, 250021 China
| | - David L. Becker
- Department of Cell & Development Biology, University College London, London, UK
| |
Collapse
|
23
|
Decrock E, De Bock M, Wang N, Gadicherla AK, Bol M, Delvaeye T, Vandenabeele P, Vinken M, Bultynck G, Krysko DV, Leybaert L. IP3, a small molecule with a powerful message. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:1772-86. [PMID: 23291251 DOI: 10.1016/j.bbamcr.2012.12.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 12/18/2012] [Accepted: 12/19/2012] [Indexed: 12/22/2022]
Abstract
Research conducted over the past two decades has provided convincing evidence that cell death, and more specifically apoptosis, can exceed single cell boundaries and can be strongly influenced by intercellular communication networks. We recently reported that gap junctions (i.e. channels directly connecting the cytoplasm of neighboring cells) composed of connexin43 or connexin26 provide a direct pathway to promote and expand cell death, and that inositol 1,4,5-trisphosphate (IP3) diffusion via these channels is crucial to provoke apoptosis in adjacent healthy cells. However, IP3 itself is not sufficient to induce cell death and additional factors appear to be necessary to create conditions in which IP3 will exert proapoptotic effects. Although IP3-evoked Ca(2+) signaling is known to be required for normal cell survival, it is also actively involved in apoptosis induction and progression. As such, it is evident that an accurate fine-tuning of this signaling mechanism is crucial for normal cell physiology, while a malfunction can lead to cell death. Here, we review the role of IP3 as an intracellular and intercellular cell death messenger, focusing on the endoplasmic reticulum-mitochondrial synapse, followed by a discussion of plausible elements that can convert IP3 from a physiological molecule to a killer substance. Finally, we highlight several pathological conditions in which anomalous intercellular IP3/Ca(2+) signaling might play a role. This article is part of a Special Issue entitled:12th European Symposium on Calcium.
Collapse
Affiliation(s)
- Elke Decrock
- Department of Basic Medical Sciences, Ghent University, Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Spray DC, Hanstein R, Lopez-Quintero SV, Stout RF, Suadicani SO, Thi MM. Gap junctions and Bystander Effects: Good Samaritans and executioners. ACTA ACUST UNITED AC 2012; 2:1-15. [PMID: 23565352 DOI: 10.1002/wmts.72] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The "Bystander" and "Good Samaritan" effects involve the transfer of toxic or beneficial compounds from one cell to a generally adjacent other through gap junction channels and through extracellular routes. The variety of injuries in which bystander cell killing or protection occurs has greatly expanded in the last decade to include infectious agents and therapeutic compounds, radiation injury, chaperones in cell therapy and apoptosis in development. This has been accompanied by the appreciation that both gap junction mediated and paracrine routes are used for the signaling of the "kiss of life" and the "kiss of death" and that manipulations of these pathways and the molecules that use them may find therapeutic utility in treatment of a variety of pathological conditions.
Collapse
Affiliation(s)
- David C Spray
- Dominick P. Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461 ; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461
| | | | | | | | | | | |
Collapse
|
25
|
Spatiotemporal pattern of rod degeneration in the S334ter-line-3 rat model of retinitis pigmentosa. Cell Tissue Res 2012; 351:29-40. [DOI: 10.1007/s00441-012-1522-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 10/18/2012] [Indexed: 11/25/2022]
|
26
|
Abstract
In the mammalian CNS, excessive release of glutamate and overactivation of glutamate receptors are responsible for the secondary (delayed) neuronal death following neuronal injury, including ischemia, traumatic brain injury (TBI) and epilepsy. The coupling of neurons by gap junctions (electrical synapses) increases during neuronal injury. In a recent study with the use of in vivo and in vitro models of cortical ischemia in mice, we have demonstrated that the ischemic increase in neuronal gap junction coupling is regulated by glutamate via group II metabotropic glutamate receptors (mGluR). Specifically, we found that activation of group II mGluRs increases background levels of neuronal gap junction coupling and expression of connexin 36 (Cx36; neuronal gap junction protein), whereas inactivation of group II mGluRs prevents the ischemia-mediated increases in the coupling and Cx36 expression. Using the analysis of neuronal death, we also established that inactivation of group II mGluRs or genetic elimination of Cx36 both dramatically reduce ischemic neuronal death in vitro and in vivo. Similar results were obtained using in vitro models of TBI and epilepsy. Our study demonstrated that mechanisms for the injury-mediated increase in neuronal gap junction coupling are part of the mechanisms for glutamate-dependent neuronal death.
Collapse
Affiliation(s)
- Andrei B Belousov
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
27
|
Kar R, Batra N, Riquelme MA, Jiang JX. Biological role of connexin intercellular channels and hemichannels. Arch Biochem Biophys 2012; 524:2-15. [PMID: 22430362 PMCID: PMC3376239 DOI: 10.1016/j.abb.2012.03.008] [Citation(s) in RCA: 174] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 02/16/2012] [Accepted: 03/06/2012] [Indexed: 12/11/2022]
Abstract
Gap junctions (GJ) and hemichannels (HC) formed from the protein subunits called connexins are transmembrane conduits for the exchange of small molecules and ions. Connexins and another group of HC-forming proteins, pannexins comprise the two families of transmembrane proteins ubiquitously distributed in vertebrates. Most cell types express more than one connexin or pannexin. While connexin expression and channel activity may vary as a function of physiological and pathological states of the cell and tissue, only a few studies suggest the involvement of pannexin HC in acquired pathological conditions. Importantly, genetic mutations in connexin appear to interfere with GJ and HC function which results in several diseases. Thus connexins could serve as potential drug target for therapeutic intervention. Growing evidence suggests that diseases resulting from HC dysfunction might open a new direction for development of specific HC reagents. This review provides a comprehensive overview of the current studies of GJ and HC formed by connexins and pannexins in various tissue and organ systems including heart, central nervous system, kidney, mammary glands, ovary, testis, lens, retina, inner ear, bone, cartilage, lung and liver. In addition, present knowledge of the role of GJ and HC in cell cycle progression, carcinogenesis and stem cell development is also discussed.
Collapse
Affiliation(s)
| | | | - Manuel A Riquelme
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229-3900
| | - Jean X. Jiang
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229-3900
| |
Collapse
|
28
|
Belousov AB. Novel model for the mechanisms of glutamate-dependent excitotoxicity: role of neuronal gap junctions. Brain Res 2012; 1487:123-30. [PMID: 22771704 DOI: 10.1016/j.brainres.2012.05.063] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 05/23/2012] [Accepted: 05/31/2012] [Indexed: 11/16/2022]
Abstract
In the mammalian central nervous system (CNS), coupling of neurons by gap junctions (electrical synapses) increases during early post-natal development, then decreases, but increases in the mature CNS following neuronal injury, such as ischemia, traumatic brain injury and epilepsy. Glutamate-dependent neuronal death also occurs in the CNS during development and neuronal injury, i.e., at the time when neuronal gap junction coupling is increased. Here, we review our recent studies on the regulation of neuronal gap junction coupling by glutamate during development and injury and on the role of gap junctions in neuronal cell death. A novel model of the mechanisms of glutamate-dependent neuronal death is discussed, which includes neuronal gap junction coupling as a critical part of these mechanisms.
Collapse
Affiliation(s)
- Andrei B Belousov
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
29
|
Wang Y, Song JH, Denisova JV, Park WM, Fontes JD, Belousov AB. Neuronal gap junction coupling is regulated by glutamate and plays critical role in cell death during neuronal injury. J Neurosci 2012; 32:713-25. [PMID: 22238107 PMCID: PMC3567463 DOI: 10.1523/jneurosci.3872-11.2012] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 10/10/2011] [Accepted: 10/31/2011] [Indexed: 11/21/2022] Open
Abstract
In the mammalian CNS, excessive release of glutamate and overactivation of glutamate receptors are responsible for the secondary (delayed) neuronal death following neuronal injury, including ischemia, traumatic brain injury (TBI), and epilepsy. The coupling of neurons by gap junctions (electrical synapses) increases during neuronal injury. We report here that the ischemic increase in neuronal gap junction coupling is regulated by glutamate via group II metabotropic glutamate receptors (mGluRs). Specifically, using electrotonic coupling, Western blots, and siRNA in the mouse somatosensory cortex in vivo and in vitro, we demonstrate that activation of group II mGluRs increases background levels of neuronal gap junction coupling and expression of connexin 36 (Cx36) (neuronal gap junction protein), and inactivation of group II mGluRs prevents the ischemia-mediated increases in the coupling and Cx36 expression. We also show that the regulation is via cAMP/PKA (cAMP-dependent protein kinase)-dependent signaling and posttranscriptional control of Cx36 expression and that other glutamate receptors are not involved in these regulatory mechanisms. Furthermore, using the analysis of neuronal death, we show that inactivation of group II mGluRs or genetic elimination of Cx36 both dramatically reduce ischemia-mediated neuronal death in vitro and in vivo. Similar results are obtained using in vitro models of TBI and epilepsy. Our results indicate that neuronal gap junction coupling is a critical component of glutamate-dependent neuronal death. They also suggest that causal link among group II mGluR function, neuronal gap junction coupling, and neuronal death has a universal character and operates in different types of neuronal injuries.
Collapse
Affiliation(s)
- Yongfu Wang
- Departments of Molecular and Integrative Physiology and
| | - Ji-Hoon Song
- Departments of Molecular and Integrative Physiology and
| | | | - Won-Mee Park
- Departments of Molecular and Integrative Physiology and
| | - Joseph D. Fontes
- Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas 66160
| | | |
Collapse
|
30
|
Transfer of IP₃ through gap junctions is critical, but not sufficient, for the spread of apoptosis. Cell Death Differ 2011; 19:947-57. [PMID: 22117194 DOI: 10.1038/cdd.2011.176] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Decades of research have indicated that gap junction channels contribute to the propagation of apoptosis between neighboring cells. Inositol 1,4,5-trisphosphate (IP₃) has been proposed as the responsible molecule conveying the apoptotic message, although conclusive results are still missing. We investigated the role of IP₃ in a model of gap junction-mediated spreading of cytochrome C-induced apoptosis. We used targeted loading of high-molecular-weight agents interfering with the IP₃ signaling cascade in the apoptosis trigger zone and cell death communication zone of C6-glioma cells heterologously expressing connexin (Cx)43 or Cx26. Blocking IP₃ receptors or stimulating IP₃ degradation both diminished the propagation of apoptosis. Apoptosis spread was also reduced in cells expressing mutant Cx26, which forms gap junctions with an impaired IP₃ permeability. However, IP₃ by itself was not able to induce cell death, but only potentiated cell death propagation when the apoptosis trigger was applied. We conclude that IP₃ is a key necessary messenger for communicating apoptotic cell death via gap junctions, but needs to team up with other factors to become a fully pro-apoptotic messenger.
Collapse
|
31
|
Decrock E, Vinken M, Bol M, D'Herde K, Rogiers V, Vandenabeele P, Krysko DV, Bultynck G, Leybaert L. Calcium and connexin-based intercellular communication, a deadly catch? Cell Calcium 2011; 50:310-21. [PMID: 21621840 DOI: 10.1016/j.ceca.2011.05.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 05/03/2011] [Accepted: 05/05/2011] [Indexed: 10/18/2022]
Abstract
Ca(2+) is known as a universal messenger mediating a wide variety of cellular processes, including cell death. In fact, this ion has been proposed as the 'cell death master', not only at the intracellular but also at the intercellular level. The most direct form of intercellular spread of cell death is mediated by gap junction channels. These channels have been shown to propagate cell death as well as cell survival signals between the cytoplasm of neighbouring cells, reflecting the dual role of Ca(2+) signals, i.e. cell death versus survival. Its precursor, the unopposed hemichannel (half of a gap junction channel), has recently joined in as a toxic pore connecting the intracellular with the extracellular environment and allowing the passage of a range of substances. The biochemical nature of the so-called intercellular cell death molecule, transferred through gap junctions or released/taken up via hemichannels, remains elusive but several studies pinpoint Ca(2+) itself or its messenger inositol trisphosphate as the responsible masters in crime. Although direct evidence is still lacking, indirect data including Ca(2+) involvement in intercellular communication and cell death, and effects of intercellular communication on intracellular Ca(2+) homeostasis, support this hypothesis. In addition, hemichannels and their molecular building blocks, connexin or pannexin proteins, may exert their effects on Ca(2+)-dependent cell death at the intracellular level, independently from their channel functions. This review provides a cutting edge overview of the current knowledge and underscores the intimate connection between intercellular communication, Ca(2+) signalling and cell death.
Collapse
Affiliation(s)
- Elke Decrock
- Department of Basic Medical Sciences - Physiology Group, Faculty of Medicine and Health Sciences, Ghent University, B-9000 Ghent, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Peixoto PM, Lue JK, Ryu SY, Wroble BN, Sible JC, Kinnally KW. Mitochondrial apoptosis-induced channel (MAC) function triggers a Bax/Bak-dependent bystander effect. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 178:48-54. [PMID: 21224042 DOI: 10.1016/j.ajpath.2010.11.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 08/20/2010] [Accepted: 09/14/2010] [Indexed: 02/08/2023]
Abstract
Collateral spread of apoptosis to nearby cells is referred to as the bystander effect, a process that is integral to tissue homeostasis and a challenge to anticancer therapies. In many systems, apoptosis relies on permeabilization of the mitochondrial outer membrane to factors such as cytochrome c and Smac/DIABLO. This permeabilization occurs via formation of a mitochondrial apoptosis-induced channel (MAC) and was mimicked here by single-cell microinjection of cytochrome c into Xenopus laevis embryos. Waves of apoptosis were observed in vivo from the injected to the neighboring cells. This finding indicates that a death signal generated downstream of cytochrome c release diffused to neighboring cells and ultimately killed the animals. The role of MAC in bystander effects was then assessed in mouse embryonic fibroblasts that did or did not express its main components, Bax and/or Bak. Exogenous expression of green fluorescent protein-Bax triggered permeabilization of the outer membrane and apoptosis in these cells. Time-lapse videos showed that neighboring cells also underwent apoptosis, but expression of Bax and/or Bak was essential to this effect, because no bystanders were observed in cells lacking both of these MAC components. These results may guide development of novel therapeutic strategies to selectively eliminate tumors or minimize the size of tissue injury in degenerative or traumatic cell death.
Collapse
Affiliation(s)
- Pablo M Peixoto
- Department of Basic Sciences, New York University College of Dentistry, New York, New York, USA
| | | | | | | | | | | |
Collapse
|
33
|
Kandouz M, Batist G. Gap junctions and connexins as therapeutic targets in cancer. Expert Opin Ther Targets 2010; 14:681-92. [PMID: 20446866 DOI: 10.1517/14728222.2010.487866] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
IMPORTANCE OF THE FIELD Connexins (Cxs) and gap junctional intercellular communications (GJICs) play roles in cancer development, growth and metastasis. Experimental studies suggest that targeting Cxs may be a novel technique, either to inhibit tumor cell growth directly or to sensitize to various therapies. AREAS COVERED IN THIS REVIEW A brief introduction to the role of Cxs in cancer. The focus is mainly on data available in the literature regarding therapeutic aspects. WHAT THE READER WILL GAIN This article reviews the various strategies that take advantage of gap junctions and connexins to eliminate cancer cells, including use of the bystander effect (BE) in gene therapy, the effect of connexins on chemosensitization, the role of apoptotic processes and interactions with the microenvironment. Attempts to restore connexin expression at the transcriptional and post-transcriptional levels are described, as well as promising strategies recently explored. The potential and limitations of the approaches are discussed. TAKE HOME MESSAGE Connexins have multiple facets, singly, in hemichannel complexes, in gap junctions or interacting with different proteins. The regulation of their expression is not fully resolved and selective manipulation of Cxs expression is therefore a challenge. Although the therapeutic potential of connexins is undeniable, more effort is needed to study the regulation and functions of these proteins.
Collapse
Affiliation(s)
- Mustapha Kandouz
- Wayne State University, Department of Pathology, 5101 Cass Avenue, Chemistry Building, Detroit, Michigan 48202, USA.
| | | |
Collapse
|
34
|
Peixoto PM, Ryu SY, Pruzansky DP, Kuriakose M, Gilmore A, Kinnally KW. Mitochondrial apoptosis is amplified through gap junctions. Biochem Biophys Res Commun 2009; 390:38-43. [PMID: 19766591 DOI: 10.1016/j.bbrc.2009.09.054] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2009] [Accepted: 09/15/2009] [Indexed: 10/20/2022]
Abstract
The death of one cell can precipitate the death of nearby cells in a process referred to as the bystander effect. We investigated whether mitochondrial apoptosis generated a bystander effect and, if so, by which pathway. Microinjection with cytochrome c mimicked function of the mitochondrial apoptosis-induced channel MAC and caused apoptosis of both target and nearby osteoblasts. This effect was suppressed by inhibiting gap junction intercellular communication. A bystander effect was also observed after exogenous expression of tBid, which facilitates MAC formation and cytochrome c release. Interestingly, in connexin-43 deficient osteoblasts, microinjection of cytochrome c induced apoptosis only in the target cell. These findings indicate that a death signal was generated downstream of MAC function and was transmitted through gap junctions to amplify apoptosis in neighboring cells. This concept may have implications in development of new therapeutic approaches.
Collapse
Affiliation(s)
- Pablo M Peixoto
- Dept Basic Sciences, New York University College of Dentistry, New York, NY 10010, USA
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
Evidence is accumulating that some forms of cell death, like apoptosis, are not only governed by the complex interplay between extracellular and intracellular signals but are also strongly influenced by intercellular communicative networks. The latter is provided by arrays of channels consisting of connexin proteins, with gap junctions directly connecting the cytoplasm of neighboring cells and hemichannels positioned as pores that link the cytoplasm to the extracellular environment. The role of gap junctions in cell death communication has received considerable interest and recently hemichannels have joined in as potentially toxic pores adding their part to the cell death process. However, despite a large body of existing evidence, especially for gap junctions, the exact contribution of the connexin channel family still remains controversial, as both gap junctions and hemichannels may furnish cell death as well as cell survival signals. An additional layer of complexity is formed by the fact that connexin proteins as such, beyond their channel function, may influence the cell death process. We here review the current knowledge on connexins and their channels in cell death and specifically address the molecular mechanisms that underlie connexin-related signaling. We also briefly focus on pannexins, a novel set of connexin-like proteins that have been implicated in cellular responses to pathological insults.
Collapse
|
36
|
Decrock E, De Vuyst E, Vinken M, Van Moorhem M, Vranckx K, Wang N, Van Laeken L, De Bock M, D'Herde K, Lai CP, Rogiers V, Evans WH, Naus CC, Leybaert L. Connexin 43 hemichannels contribute to the propagation of apoptotic cell death in a rat C6 glioma cell model. Cell Death Differ 2008; 16:151-63. [PMID: 18820645 DOI: 10.1038/cdd.2008.138] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Gap junctions (GJs) have been demonstrated to communicate cell death signals from apoptotic to healthy cells, thereby spatially extending apoptosis. Before being incorporated into GJs, hemichannels (hemi-GJs) are normally closed but recent evidence suggests that they can be opened by various messengers and conditions, thereby forming a pore through which molecules can enter or leave the cell potentially leading to cell death. The aim of this study was to determine the contribution of GJs and hemichannels in the communication of apoptosis toward surrounding cells. We induced apoptosis in C6 glioma cells stably transfected with connexin (Cx)43, with cytochrome C (cytC) using in situ electroporation and found that healthy surrounding cells underwent apoptotic transformation. Work with various cell death markers, wild-type (WT) and Cx43-expressing cells, inhibitors of GJs and/or hemichannels, and Cx43 gene silencing showed that GJs contribute to the spread of apoptosis in a zone next to where apoptosis was triggered whereas hemichannels also promoted cell death beyond this area. Buffering cytoplasmic Ca(2+) changes inhibited the spread of apoptosis in both cases. We conclude that Cx43 hemichannels, in concert with their GJ counterparts, play a role in communicating cytC-induced apoptotic cell death messages.
Collapse
Affiliation(s)
- E Decrock
- Department of Basic Medical Sciences - Physiology group, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
de Rivero Vaccari JC, Corriveau RA, Belousov AB. Gap junctions are required for NMDA receptor dependent cell death in developing neurons. J Neurophysiol 2007; 98:2878-86. [PMID: 17855590 DOI: 10.1152/jn.00362.2007] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
A number of studies have indicated an important role for N-methyl-D-aspartate (NMDA) receptors in cell survival versus cell death decisions during neuronal development, trauma, and ischemia. Coupling of neurons by electrical synapses (gap junctions) is high or increases in neuronal networks during all three of these conditions. However, whether neuronal gap junctions contribute to NMDA receptor-regulated cell death is not known. Here we address the role of neuronal gap junction coupling in NMDA receptor-regulated cell death in developing neurons. We report that inactivation or hyperactivation of NMDA receptors induces neuronal cell death in primary hypothalamic cultures, specifically during the peak of developmental gap junction coupling. In contrast, increasing or decreasing NMDA receptor function when gap junction coupling is low has no or greatly reduced impact on cell survival. Pharmacological inactivation of gap junctions or knockout of neuronal connexin 36 prevents the cell death caused by NMDA receptor hypofunction or hyperfunction. The results indicate the critical role of neuronal gap junctions in cell death caused by increased or decreased NMDA receptor function in developing neurons. Based on these data, we propose the novel hypothesis that NMDA receptors and gap junctions work in concert to regulate neuronal survival.
Collapse
|
38
|
Talhouk RS, Zeinieh MP, Mikati MA, El-Sabban ME. Gap junctional intercellular communication in hypoxia-ischemia-induced neuronal injury. Prog Neurobiol 2007; 84:57-76. [PMID: 18006137 DOI: 10.1016/j.pneurobio.2007.10.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2006] [Revised: 08/29/2007] [Accepted: 10/04/2007] [Indexed: 01/07/2023]
Abstract
Brain hypoxia-ischemia is a relatively common and serious problem in neonates and in adults. Its consequences include long-term histological and behavioral changes and reduction in seizure threshold. Gap junction intercellular communication is pivotal in the spread of hypoxia-ischemia related injury and in mediating its long-term effects. This review provides a comprehensive and critical review of hypoxia-ischemia and hypoxia in the brain and the potential role of gap junctions in the spread of the neuronal injury induced by these insults. It also presents the effects of hypoxia-ischemia and of hypoxia on the state of gap junctions in vitro and in vivo. Understanding the mechanisms involved in gap junction-mediated neuronal injury due to hypoxia will lead to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Rabih S Talhouk
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | | | | | | |
Collapse
|
39
|
Abstract
"Hemichannels" are defined as the halves of gap junction channels (also termed connexons) that are contributed by one cell; "hemichannels" are considered to be functional if they are open in nonjunctional membranes in the absence of pairing with partners from adjacent cells. Several recent reviews have summarized the blossoming literature regarding functional "hemichannels", in some cases encyclopedically. However, most of these previous reviews have been written with the assumption that all data reporting "hemichannel" involvement really have studied phenomena in which connexons actually form the permeability or conductance pathway. In this review, we have taken a slightly different approach. We review the concept of "hemichannels", summarize properties that might be expected of half gap junctions and evaluate the extent to which the properties of presumptive "hemichannels" match expectations. Then we consider functions attributed to hemichannels, provide an overview of other channel types that might fulfill similar roles and provide sets of criteria that might be applied to verify involvement of connexin hemichannels in cell and tissue function. One firm conclusion is reached. The study of hemichannels is technically challenging and fraught with opportunities for misinterpretation, so that future studies must apply rigorous standards for detection of hemichannel expression and function. At the same time there are reasons to expect surprises, including the possibility that some time honored techniques for studying gap junctions may prove unsuitable for detecting hemichannels. We advise hemichannel researchers to proceed with caution and an open mind.
Collapse
Affiliation(s)
- David C Spray
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| | - Zu-Cheng Ye
- Department of Neurology, University of Washington School of Medicine, Seattle, Washington
| | - Bruce R Ransom
- Department of Neurology, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|