1
|
de Sousa JC, Santos SACS, Kurtenbach E. Multiple approaches for the evaluation of connexin-43 expression and function in macrophages. J Immunol Methods 2024; 533:113741. [PMID: 39111361 DOI: 10.1016/j.jim.2024.113741] [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: 02/22/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
Connexins are essential gap junction proteins that play pivotal roles in intercellular communication in various organs of mammals. Connexin-43 (Cx43) is expressed in various components of the immune system, and there is extensive evidence of its participation in inflammation responses. The involvement of Cx43 in macrophage functionality involves the purinergic signaling pathway. Macrophages contribute to defenses against inflammatory reactions such as bacterial sepsis and peritonitis. Several assays can identify the presence and activity of Cx43 in macrophages. Real-time polymerase chain reaction (PCR) can measure the relative mRNA expression of Cx43, whereas western blotting can detect protein expression levels. Using immunofluorescence assays, it is possible to analyze the expression and observe the localization of Cx43 in cells or tissues. Moreover, connexin-mediated gap junction intercellular communication can be evaluated using functional assays such as microinjection of fluorescent dyes or scrape loading-dye transfer. The use of selective inhibitors contributes to this understanding and reinforces the role of connexins in various processes. Here, we discuss these methods to evaluate Cx43 and macrophage gap junctions.
Collapse
Affiliation(s)
- Júlia Costa de Sousa
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil; Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil.
| | | | - Eleonora Kurtenbach
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil; Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| |
Collapse
|
2
|
Luo Y, Zheng S, Xiao W, Zhang H, Li Y. Pannexins in the musculoskeletal system: new targets for development and disease progression. Bone Res 2024; 12:26. [PMID: 38705887 PMCID: PMC11070431 DOI: 10.1038/s41413-024-00334-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/04/2024] [Accepted: 04/01/2024] [Indexed: 05/07/2024] Open
Abstract
During cell differentiation, growth, and development, cells can respond to extracellular stimuli through communication channels. Pannexin (Panx) family and connexin (Cx) family are two important types of channel-forming proteins. Panx family contains three members (Panx1-3) and is expressed widely in bone, cartilage and muscle. Although there is no sequence homology between Panx family and Cx family, they exhibit similar configurations and functions. Similar to Cxs, the key roles of Panxs in the maintenance of physiological functions of the musculoskeletal system and disease progression were gradually revealed later. Here, we seek to elucidate the structure of Panxs and their roles in regulating processes such as osteogenesis, chondrogenesis, and muscle growth. We also focus on the comparison between Cx and Panx. As a new key target, Panxs expression imbalance and dysfunction in muscle and the therapeutic potentials of Panxs in joint diseases are also discussed.
Collapse
Affiliation(s)
- Yan Luo
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, 410008, China
| | - Shengyuan Zheng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, 410008, China
| | - Wenfeng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hang Zhang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
3
|
Pendleton E, Ketner A, Ransick P, Ardekani D, Bodenstine T, Chandar N. Loss of Function of the Retinoblastoma Gene Affects Gap Junctional Intercellular Communication and Cell Fate in Osteoblasts. BIOLOGY 2024; 13:39. [PMID: 38248470 PMCID: PMC10813623 DOI: 10.3390/biology13010039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/08/2023] [Accepted: 01/05/2024] [Indexed: 01/23/2024]
Abstract
Loss of function of the Retinoblastoma gene (RB1) due to mutations is commonly seen in human osteosarcomas. One of the Rb1 gene functions is to facilitate cell fate from mesenchymal stem cells to osteoblasts and prevent adipocyte differentiations. In this study, we demonstrate that a stable reduction of Rb1 expression (RbKD) in murine osteoblasts causes them to express higher levels of PPAR-ɣ and other adipocyte-specific transcription factors while retaining high expression of osteoblast-specific transcription factors, Runx2/Cbfa1 and SP7/Osterix. Inhibition of gap junctional intercellular communication (GJIC) in osteoblasts is another mechanism that causes osteoblasts to transdifferentiate to adipocytes. We found that preosteoblasts exposed to osteoblast differentiating media (DP media) increased GJIC. RbKD cells showed reduced GJIC along with a reduction in expression of Cx43, the protein that mediates GJIC. Other membrane associated adhesion protein Cadherin 11 (Cad11) was also decreased. Since PPAR-ɣ is increased with Rb1 loss, we wondered if the reduction of this transcription factor would reverse the changes observed. Reduction of PPAR-ɣ in control osteoblasts slightly increased bone-specific expression and reduced adipocytic expression as expected along with an increase in Cad11 and Cx43 expression. GJIC remained high and was unaffected by a reduction in PPAR-ɣ in control cells. Knockdown of PPAR-ɣ in RbKD cells reduced adipocyte gene expression, while osteoblast-specific expression showed improvement. Cx43, Cad11 and GJIC remained unaffected by PPAR-ɣ reduction. Our observations suggest that increased PPAR-ɣ that happens with Rb1 loss only affects osteoblast-adipocyte-specific gene expression but does not completely reverse Cx43 gene expression or GJIC. Therefore, these effects may represent independent events triggered by Rb1loss and/or the differentiation process.
Collapse
Affiliation(s)
- Elisha Pendleton
- Department of Biochemistry and Molecular Genetics, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA; (E.P.); (A.K.); (T.B.)
| | - Anthony Ketner
- Department of Biochemistry and Molecular Genetics, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA; (E.P.); (A.K.); (T.B.)
| | - Phil Ransick
- Chicago College of Osteopathic Medicine, Midwestern University, 555 31st Street, Downers Grove, IL 60515, USA; (P.R.); (D.A.)
| | - Doug Ardekani
- Chicago College of Osteopathic Medicine, Midwestern University, 555 31st Street, Downers Grove, IL 60515, USA; (P.R.); (D.A.)
| | - Thomas Bodenstine
- Department of Biochemistry and Molecular Genetics, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA; (E.P.); (A.K.); (T.B.)
| | - Nalini Chandar
- Department of Biochemistry and Molecular Genetics, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA; (E.P.); (A.K.); (T.B.)
| |
Collapse
|
4
|
An S, Zheng S, Cai Z, Chen S, Wang C, Li Y, Deng Z. Connexin43 in Musculoskeletal System: New Targets for Development and Disease Progression. Aging Dis 2022; 13:1715-1732. [PMID: 36465186 PMCID: PMC9662276 DOI: 10.14336/ad.2022.0421] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/21/2022] [Indexed: 03/16/2025] Open
Abstract
Connexin43, which is the most highly expressed connexin subtype in the musculoskeletal system, exists in a variety of bone cells, synovial tissue, and cartilage tissue. Connexin43 has been suggested to be a key regulator of bone homeostasis. Studies have shown aberrant Connexin43 expression in musculoskeletal disorders, such as osteoporosis, osteoarthritis, and rheumatoid arthritis. During cellular activities, Connexin43 can participate in the formation of functionally specific gap junctions and hemichannels and can exert independent cellular regulatory and signaling functions through special C-termini. The critical role of Connexin43 in physiological development and disease progression has been gradually revealed. In this article, the function of Connexin43 in musculoskeletal tissues is summarized, revealing the potential role of Connexin43 as a key target in the treatment of related bone and muscle disorders and the need for further discovery.
Collapse
Affiliation(s)
- Senbo An
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| | - Shengyuan Zheng
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Clinical Medicine, Xiangya Medicine School, Central South University, Changsha, Hunan, China.
| | - Zijun Cai
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Siyu Chen
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China.
| | - Chen Wang
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Zhenhan Deng
- Department of Sports Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, Guangdong, China.
| |
Collapse
|
5
|
Iyer AR, Sheeba V. A new player in circadian networks: Role of electrical synapses in regulating functions of the circadian clock. Front Physiol 2022; 13:968574. [PMID: 36406999 PMCID: PMC9669436 DOI: 10.3389/fphys.2022.968574] [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: 06/14/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Several studies have indicated that coherent circadian rhythms in behaviour can be manifested only when the underlying circadian oscillators function as a well-coupled network. The current literature suggests that circadian pacemaker neuronal networks rely heavily on communication mediated by chemical synapses comprising neuropeptides and neurotransmitters to regulate several behaviours and physiological processes. It has become increasingly clear that chemical synapses closely interact with electrical synapses and function together in the neuronal networks of most organisms. However, there are only a few studies which have examined the role of electrical synapses in circadian networks and here, we review our current understanding of gap junction proteins in circadian networks of various model systems. We describe the general mechanisms by which electrical synapses function in neural networks, their interactions with chemical neuromodulators and their contributions to the regulation of circadian rhythms. We also discuss the various methods available to characterize functional electrical synapses in these networks and the potential directions that remain to be explored to understand the roles of this relatively understudied mechanism of communication in modulating circadian behaviour.
Collapse
Affiliation(s)
- Aishwarya Ramakrishnan Iyer
- Chronobiology and Behavioural Neurogenetics Laboratory, Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, Karnataka, India
- Department of Neuroscience and Behavior, Barnard College of Columbia University, New York, NY, United States
| | - Vasu Sheeba
- Chronobiology and Behavioural Neurogenetics Laboratory, Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, Karnataka, India
- *Correspondence: Vasu Sheeba,
| |
Collapse
|
6
|
Astragaloside IV attenuated TGF-β1- induced epithelial-mesenchymal transition of renal tubular epithelial cells via connexin 43 and Akt/mTOR signaling pathway. Tissue Cell 2022; 77:101831. [PMID: 35643056 DOI: 10.1016/j.tice.2022.101831] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 01/25/2023]
Abstract
INTRODUCTION The objective of the study was to observe whether connexin 43 (Cx43) could regulate epithelial mesenchymal transformation (EMT) of renal tubular epithelial cells (RTECs) by influencing Akt/mTOR signaling pathway, and whether ASV could inhibit the development of renal interstitial fibrosis by regulating Cx43. METHODS Lentivirus infection was transfected into RTECs with the final concentration of 50 ×PFU/ cell to regulate the expression of Cx43. And RTECs were intervened by different doses of Astragaloside IV (ASV). After synchronous culture of RTECs in each group,the expression levels of EMT-related indicators and Cx43 were detected by fluorescence microscope and Western-Blotting (WB), even the protein expressions and phosphorylation levels of AKT and mTOR in different groups were detected by WB. RESULTS When the expression of Cx43 in RTECs was regulated by lentivirus infection, the degree of EMT induced by TGF‑β1 and the phosphorylation level of Akt and mTOR were changed accordingly, indicating that Akt/mTOR pathway might be a downstream molecular mechanism by which Cx43 could regulate EMT. After intervention with different doses of ASV, the expression level of Cx43 increased with obvious concentration dependence, and the expression levels of p-Akt and p- mTOR were significantly altered, suggesting that ASV could effectively increase the protein expressions of TGF‑β1-induced Cx43 in RTECs and inhibit the phosphorylation levels of Akt and mTOR. CONCLUSION Cx43 were the main material basis of RTECs' injury, and ASV could inhibit TGF-β1- induced RTECs' transdifferentiation. In-depth study of the mechanism might provide a broad application prospect for the treatment of renal interstitial fibrosis.
Collapse
|
7
|
Yamada A, Yoshizaki K, Ishikawa M, Saito K, Chiba Y, Fukumoto E, Hino R, Hoshikawa S, Chiba M, Nakamura T, Iwamoto T, Fukumoto S. Connexin 43-Mediated Gap Junction Communication Regulates Ameloblast Differentiation via ERK1/2 Phosphorylation. Front Physiol 2021; 12:748574. [PMID: 34630166 PMCID: PMC8500398 DOI: 10.3389/fphys.2021.748574] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 08/25/2021] [Indexed: 11/24/2022] Open
Abstract
Connexin 43 (Cx43) is an integral membrane protein that forms gap junction channels. These channels mediate intercellular transport and intracellular signaling to regulate organogenesis. The human disease oculodentodigital dysplasia (ODDD) is caused by mutations in Cx43 and is characterized by skeletal, ocular, and dental abnormalities including amelogenesis imperfecta. To clarify the role of Cx43 in amelogenesis, we examined the expression and function of Cx43 in tooth development. Single-cell RNA-seq analysis and immunostaining showed that Cx43 is highly expressed in pre-secretory ameloblasts, differentiated ameloblasts, and odontoblasts. Further, we investigated the pathogenic mechanisms of ODDD by analyzing Cx43-null mice. These mice developed abnormal teeth with multiple dental epithelium layers. The expression of enamel matrix proteins such as ameloblastin (Ambn), which is critical for enamel formation, was significantly reduced in Cx43-null mice. TGF-β1 induces Ambn transcription in dental epithelial cells. The induction of Ambn expression by TGF-β1 depends on the density of the cultured cells. Cell culture at low densities reduces cell–cell contact and reduces the effect of TGF-β1 on Ambn induction. When cell density was high, Ambn expression by TGF-β1 was enhanced. This induction was inhibited by the gap junction inhibitors, oleamide, and 18α-grycyrrhizic acid and was also inhibited in cells expressing Cx43 mutations (R76S and R202H). TGF-β1-mediated phosphorylation and nuclear translocation of ERK1/2, but not Smad2/3, were suppressed by gap junction inhibitors. Cx43 gap junction activity is required for TGF-β1-mediated Runx2 phosphorylation through ERK1/2, which forms complexes with Smad2/3. In addition to its gap junction activity, Cx43 may also function as a Ca2+ channel that regulates slow Ca2+ influx and ERK1/2 phosphorylation. TGF-β1 transiently increases intracellular calcium levels, and the increase in intracellular calcium over a short period was not related to the expression level of Cx43. However, long-term intracellular calcium elevation was enhanced in cells overexpressing Cx43. Our results suggest that Cx43 regulates intercellular communication through gap junction activity by modulating TGF-β1-mediated ERK signaling and enamel formation.
Collapse
Affiliation(s)
- Aya Yamada
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Keigo Yoshizaki
- Section of Orthodontics and Dentofacial Orthopedics, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Masaki Ishikawa
- The Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Kan Saito
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Yuta Chiba
- Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Emiko Fukumoto
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Ryoko Hino
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Seira Hoshikawa
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Mitsuki Chiba
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Takashi Nakamura
- Division of Molecular Pharmacology and Cell Biophysics, Department of Oral Biology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Tsutomu Iwamoto
- Division of Oral Health Science, Department of Pediatric Dentistry/Special Needs Dentistry, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Satoshi Fukumoto
- Division of Pediatric Dentistry, Department of Oral Health and Development Sciences, Tohoku University Graduate School of Dentistry, Sendai, Japan.,Section of Oral Medicine for Children, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
8
|
Camal Ruggieri IN, Cícero AM, Issa JPM, Feldman S. Bone fracture healing: perspectives according to molecular basis. J Bone Miner Metab 2021; 39:311-331. [PMID: 33151416 DOI: 10.1007/s00774-020-01168-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022]
Abstract
Fractures have a great impact on health all around the world and with fracture healing optimization; this problem could be resolved partially. To make a practical contribution to this issue, the knowledge of bone tissue, cellularity, and metabolism is essential, especially cytoskeletal architecture and its transformations according to external pressures. Special physical and chemical characteristics of the extracellular matrix (ECM) allow the transmission of mechanical stimuli from outside the cell to the plasmatic membrane. The osteocyte cytoskeleton is conformed by a complex network of actin and microtubules combined with crosslinker proteins like vinculin and fimbrin, connecting and transmitting outside stimuli through EMC to cytoplasm. Herein, critical signaling pathways like Cx43-depending ones, MAPK/ERK, Wnt, YAP/TAZ, Rho-ROCK, and others are activated due to mechanical stimuli, resulting in osteocyte cytoskeletal changes and ECM remodeling, altering the tissue and, therefore, the bone. In recent years, the osteocyte has gained more interest and value in relation to bone homeostasis as a great coordinator of other cell populations, thanks to its unique functions. By integrating the latest advances in relation to intracellular signaling pathways, mechanotransmission system of the osteocyte and bone tissue engineering, there are promising experimental strategies, while some are ready for clinical trials. This work aims to show clearly and precisely the integration between cytoskeleton and main molecular pathways in relation to mechanotransmission mechanism in osteocytes, and the use of this theoretical knowledge in therapeutic tools for bone fracture healing.
Collapse
Affiliation(s)
- Iván Nadir Camal Ruggieri
- School of Medicine, LABOATEM (Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory), Biological Chemistry Cat, School of Medicine, Rosario National University, Rosario, Argentina.
| | - Andrés Mauricio Cícero
- School of Medicine, LABOATEM (Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory), Biological Chemistry Cat, School of Medicine, Rosario National University, Rosario, Argentina
| | | | - Sara Feldman
- School of Medicine, LABOATEM (Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory), Biological Chemistry Cat, School of Medicine, Rosario National University, Rosario, Argentina
- Research Council of the Rosario National University (CIUNR) and CONICET, Rosario, Argentina
| |
Collapse
|
9
|
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
|
10
|
Antagonistic Functions of Connexin 43 during the Development of Primary or Secondary Bone Tumors. Biomolecules 2020; 10:biom10091240. [PMID: 32859065 PMCID: PMC7565206 DOI: 10.3390/biom10091240] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022] Open
Abstract
Despite research and clinical advances during recent decades, bone cancers remain a leading cause of death worldwide. There is a low survival rate for patients with primary bone tumors such as osteosarcoma and Ewing’s sarcoma or secondary bone tumors such as bone metastases from prostate carcinoma. Gap junctions are specialized plasma membrane structures consisting of transmembrane channels that directly link the cytoplasm of adjacent cells, thereby enabling the direct exchange of small signaling molecules between cells. Discoveries of human genetic disorders due to genetic mutations in gap junction proteins (connexins) and experimental data using connexin knockout mice have provided significant evidence that gap-junctional intercellular communication (Gj) is crucial for tissue function. Thus, the dysfunction of Gj may be responsible for the development of some diseases. Gj is thus a main mechanism for tumor cells to communicate with other tumor cells and their surrounding microenvironment to survive and proliferate. If it is well accepted that a low level of connexin expression favors cancer cell proliferation and therefore primary tumor development, more evidence is suggesting that a high level of connexin expression stimulates various cellular process such as intravasation, extravasation, or migration of metastatic cells. If so, connexin expression would facilitate secondary tumor dissemination. This paper discusses evidence that suggests that connexin 43 plays an antagonistic role in the development of primary bone tumors as a tumor suppressor and secondary bone tumors as a tumor promoter.
Collapse
|
11
|
Van Campenhout R, Cooreman A, Leroy K, Rusiecka OM, Van Brantegem P, Annaert P, Muyldermans S, Devoogdt N, Cogliati B, Kwak BR, Vinken M. Non-canonical roles of connexins. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2020; 153:35-41. [PMID: 32220599 DOI: 10.1016/j.pbiomolbio.2020.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/06/2020] [Accepted: 03/16/2020] [Indexed: 12/12/2022]
Abstract
Gap junctions mediate cellular communication and homeostasis by controlling the intercellular exchange of small and hydrophilic molecules and ions. Gap junction channels are formed by the docking of 2 hemichannels of adjacent cells, which in turn are composed of 6 connexin subunits. Connexin proteins as such can also control the cellular life cycle independent of their channel activities. This has been most demonstrated in the context of cell growth and cell death. Different mechanisms are involved mainly related to direct interaction with cell growth or cell death regulators, but also implying effects on the expression of cell growth and cell death regulators. The present paper focuses on these atypical roles of connexin proteins.
Collapse
Affiliation(s)
- Raf Van Campenhout
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Axelle Cooreman
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kaat Leroy
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Olga M Rusiecka
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Pieter Van Brantegem
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Pieter Annaert
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Bruno Cogliati
- Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Brenda R Kwak
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Mathieu Vinken
- Department of In Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
12
|
Hong G. Enoxolone suppresses apoptosis in chondrocytes and progression of osteoarthritis via modulating the ERK1/2 signaling pathway. Arch Med Sci 2020; 20:947-961. [PMID: 39050151 PMCID: PMC11264165 DOI: 10.5114/aoms.2020.93211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 01/04/2020] [Indexed: 07/27/2024] Open
Abstract
Introduction Osteoarthritis (OA) is an inflammatory disorder of synovial joints which is mainly treated with therapeutic agents showing side effects associated with the gastrointestinal (GI) and metabolic system. Consequently, there is urgent need for a potent, safe and novel agent for treating OA and related disorders. Enoxolone is a pentacyclic triterpenoid obtained from the herb liquorice. Based on earlier findings, we postulated that enoxolone may produce chondroprotective activity by exerting anti-inflammatory, anti-catabolic and oxidative stress-decreasing effects. Material and methods The chondrocytes were extracted from the femoral head articular cartilage of healthy rats. Immunofluorescence staining was done for identification of chondrocytes. Cell viability and proliferation studies were done using Cell Counting Kit-8. Apoptotic cells were identified by TUNEL assay and flow cytometry analysis. Autophagy was assessed by monodansylcadaverine assay. Western blot analysis was done for expression of proteins. Results In the present study we investigated the protective effect of enoxolone on interleukin 1β (IL-1β) treated Iry chondrocytes in vitro. Treatment with IL-1β resulted in a significant reduction in cell viability of cells in increasing dose and time. Treatment with enoxolone along with IL-1β caused a significant decrease in growth inhibition. Also, enoxolone inhibited the IL-1β mediated apoptosis and activation of caspase-3 in cells. We also observed that enoxolone elevated the levels of p-ERK1/2, light chain 3 (LC3)-II and Beclin-1 (autophagy markers) in chondrocytes. The expression of (LC3)-II and Beclin-1 was decreased when the cells were treated with U0126 (ERK1/2 inhibitor). Conclusions Our findings demonstrate that enoxolone could suppress inflammatory signaling and apoptosis via the ERK1/2 pathway in chondrocytes.
Collapse
Affiliation(s)
- Gang Hong
- Department of Orthopedics, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Huang Y, Mao Z, Zhang Z, Obata F, Yang X, Zhang X, Huang Y, Mitsui T, Fan J, Takeda M, Yao J. Connexin43 Contributes to Inflammasome Activation and Lipopolysaccharide-Initiated Acute Renal Injury via Modulation of Intracellular Oxidative Status. Antioxid Redox Signal 2019; 31:1194-1212. [PMID: 31319679 DOI: 10.1089/ars.2018.7636] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aims: Inflammasome activation plays a pivotal role in many inflammatory diseases. Given that connexin (Cx) channels regulate numerous cellular events leading to inflammasome activation, we determined whether and how connexin affected inflammasome activation and inflammatory cell injury. Results: Exposure of mouse peritoneal macrophages (PMs) to lipopolysaccharide (LPS) plus ATP caused NLRP3 inflammasome activation, together with an increased connexin43 (Cx43). Inhibition of Cx43 blunted inflammasome activation. Consistently, PMs from the Cx43 heterozygous mouse (Cx43+/-) exhibited weak inflammasome activation, in comparison with those from the Cx43+/+ mouse. Further analysis revealed that inflammasome activation was preceded by an increased reactive oxygen species (ROS) production, nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) oxidase 2 (NOX2), protein carbonylation, and mitogen-activated protein kinase (MAPK) activation. Suppression of ROS with antioxidant, downregulation of NOX2 with small interfering RNA (siRNA), or inhibition of NADPH oxidase or MAPKs with inhibitors blocked Cx43 elevation and inflammasome activation. Intriguingly, suppression of Cx43 also blunted NOX2 expression, protein carbonylation, p38 phosphorylation, and inflammasome activation. In a model of acute renal injury induced by LPS, the Cx43+/- mouse exhibited a significantly lower level of blood interleukin-1β (IL-1β), blood urea nitrogen, and urinary protein, together with milder renal pathological changes and renal expression of NLRP3 and NOX4, as compared with the Cx43+/+ mouse. Moreover, inhibition of gap junctions suppressed IL-1β- and tumor necrosis factor-α-induced expression of NOX4 in glomerular podocytes and tubular epithelial cells. Innovation and Conclusion: Our study indicates that Cx43 contributes to inflammasome activation and the progression of renal inflammatory cell injury through modulation of intracellular redox status. Cx43 could be a novel target for the treatment of certain inflammatory diseases.
Collapse
Affiliation(s)
- Yanru Huang
- Divison of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Zhimin Mao
- Divison of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Zhen Zhang
- Divison of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Fumiko Obata
- Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Xiawen Yang
- Divison of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Xiling Zhang
- Divison of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Yong Huang
- Divison of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Takahiko Mitsui
- Department of Urology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Jianglin Fan
- Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Masayuki Takeda
- Department of Urology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Jian Yao
- Divison of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| |
Collapse
|
14
|
Burgos-Panadero R, Lucantoni F, Gamero-Sandemetrio E, Cruz-Merino LDL, Álvaro T, Noguera R. The tumour microenvironment as an integrated framework to understand cancer biology. Cancer Lett 2019; 461:112-122. [PMID: 31325528 DOI: 10.1016/j.canlet.2019.07.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/11/2019] [Accepted: 07/14/2019] [Indexed: 01/18/2023]
Abstract
Cancer cells all share the feature of being immersed in a complex environment with altered cell-cell/cell-extracellular element communication, physicochemical information, and tissue functions. The so-called tumour microenvironment (TME) is becoming recognised as a key factor in the genesis, progression and treatment of cancer lesions. Beyond genetic mutations, the existence of a malignant microenvironment forms the basis for a new perspective in cancer biology where connections at the system level are fundamental. From this standpoint, different aspects of tumour lesions such as morphology, aggressiveness, prognosis and treatment response can be considered under an integrated vision, giving rise to a new field of study and clinical management. Nowadays, somatic mutation theory is complemented with study of TME components such as the extracellular matrix, immune compartment, stromal cells, metabolism and biophysical forces. In this review we examine recent studies in this area and complement them with our own research data to propose a classification of stromal changes. Exploring these avenues and gaining insight into malignant phenotype remodelling, could reveal better ways to characterize this disease and its potential treatment.
Collapse
Affiliation(s)
- Rebeca Burgos-Panadero
- Departament of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, Valencia, Spain; CIBERONC, Madrid, Spain
| | - Federico Lucantoni
- Departament of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, Valencia, Spain
| | - Esther Gamero-Sandemetrio
- Departament of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, Valencia, Spain; CIBERONC, Madrid, Spain
| | | | - Tomás Álvaro
- CIBERONC, Madrid, Spain; Hospital Verge de la Cinta, Tortosa, Tarragona, Spain.
| | - Rosa Noguera
- Departament of Pathology, Medical School, University of Valencia - INCLIVA Biomedical Health Research Institute, Valencia, Spain; CIBERONC, Madrid, Spain.
| |
Collapse
|
15
|
Connexin43 enhances Wnt and PGE2-dependent activation of β-catenin in osteoblasts. Pflugers Arch 2019; 471:1235-1243. [PMID: 31240382 DOI: 10.1007/s00424-019-02295-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/06/2019] [Accepted: 06/14/2019] [Indexed: 12/13/2022]
Abstract
Connexin43 is an important modulator of many signaling pathways in bone. β-Catenin, a key regulator of the osteoblast differentiation and function, is among the pathways downstream of connexin43-dependent intercellular communication. There are striking overlaps between the functions of these two proteins in bone cells. However, differential effects of connexin43 on β-catenin activity have been reported. Here, we examined how connexin43 influenced both Wnt-dependent and Wnt-independent activation of β-catenin in osteoblasts in vitro. Our data show that loss of connexin43 in primary osteoblasts or connexin43 overexpression in UMR106 cells regulated active β-catenin and phospho-Akt levels, with loss of connexin43 inhibiting and connexin43 overexpression increasing the levels of active β-catenin and phospho-Akt. Increasing connexin43 expression synergistically enhanced Wnt3a-dependent activation of β-catenin protein and β-catenin transcriptional activity, as well as Wnt-independent activation of β-catenin by prostaglandin E2 (PGE2). Finally, we show that the activation of β-catenin by PGE2 required signaling through the phosphatidylinositol 3-kinase (PI3K)/Akt/glycogen synthase kinase 3 beta (GSK3β) pathway, as the PI3K inhibitor, LY-294002, disrupted the synergy between connexin43 and PGE2. These data show that connexin43 regulates Akt and β-catenin activity and synergistically enhances both Wnt-dependent and Wnt-independent β-catenin signaling in osteoblasts.
Collapse
|
16
|
Bregenzer ME, Horst EN, Mehta P, Novak CM, Raghavan S, Snyder CS, Mehta G. Integrated cancer tissue engineering models for precision medicine. PLoS One 2019; 14:e0216564. [PMID: 31075118 PMCID: PMC6510431 DOI: 10.1371/journal.pone.0216564] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tumors are not merely cancerous cells that undergo mindless proliferation. Rather, they are highly organized and interconnected organ systems. Tumor cells reside in complex microenvironments in which they are subjected to a variety of physical and chemical stimuli that influence cell behavior and ultimately the progression and maintenance of the tumor. As cancer bioengineers, it is our responsibility to create physiologic models that enable accurate understanding of the multi-dimensional structure, organization, and complex relationships in diverse tumor microenvironments. Such models can greatly expedite clinical discovery and translation by closely replicating the physiological conditions while maintaining high tunability and control of extrinsic factors. In this review, we discuss the current models that target key aspects of the tumor microenvironment and their role in cancer progression. In order to address sources of experimental variation and model limitations, we also make recommendations for methods to improve overall physiologic reproducibility, experimental repeatability, and rigor within the field. Improvements can be made through an enhanced emphasis on mathematical modeling, standardized in vitro model characterization, transparent reporting of methodologies, and designing experiments with physiological metrics. Taken together these considerations will enhance the relevance of in vitro tumor models, biological understanding, and accelerate treatment exploration ultimately leading to improved clinical outcomes. Moreover, the development of robust, user-friendly models that integrate important stimuli will allow for the in-depth study of tumors as they undergo progression from non-transformed primary cells to metastatic disease and facilitate translation to a wide variety of biological and clinical studies.
Collapse
Affiliation(s)
- Michael E. Bregenzer
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Eric N. Horst
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Pooja Mehta
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Caymen M. Novak
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Shreya Raghavan
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Catherine S. Snyder
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Geeta Mehta
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, School of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Macromolecular Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
17
|
Noguera R, Burgos-Panadero R, Gamero-Sandemetrio E, de la Cruz-Merino L, Álvaro Naranjo T. [An integral view of cancer (I). The study, classification and reprogramming of the tumoral microclimate]. REVISTA ESPAÑOLA DE PATOLOGÍA : PUBLICACIÓN OFICIAL DE LA SOCIEDAD ESPAÑOLA DE ANATOMÍA PATOLÓGICA Y DE LA SOCIEDAD ESPAÑOLA DE CITOLOGÍA 2019; 52:92-102. [PMID: 30902384 DOI: 10.1016/j.patol.2018.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/09/2018] [Accepted: 11/27/2018] [Indexed: 11/16/2022]
Abstract
The group of diseases that we call cancer share a biological structure formed by a complex ecosystem, with altered intercellular communication, information fields, development and tissue function. Beyond the genetic alterations of the tumor cell, the demonstration of an altered ecosystem, with interconnections at systemic levels, opens up a new perspective on cancer biology and behavior. Different tumor facets, such as morphology, classification, clinical aggressiveness, prognosis and response to treatment now appear under a comprehensive vision that offers a new horizon of study, research and clinical management. The Somatic Mutation Theory in cancer, in force for more than one hundred years, is now completed by the study of the tumor microenvironment, the extracellular matrix, the stromal cells, the immune response, the innervation, the nutrition, the mitochondria, the metabolism, the interstitial fluid, the mechanical and electromagnetic properties of the tissue and many other areas of emerging knowledge; thus opening the door to a reprogramming exercise of the tumor phenotype through the modification of the keys offered by this new paradigm. Its recognition makes it possible to go from considering the oncological process as a cellular problem to a supracellular alteration based on the disorganization of tissues, immersed in the relationships of the complex system of the living being.
Collapse
Affiliation(s)
- Rosa Noguera
- Departamento de Patología, Facultad de Medicina, Universidad de Valencia/Instituto de Investigaciones Sanitarias INCLIVA, Valencia, España; CIBERONC, Madrid, España
| | - Rebeca Burgos-Panadero
- Departamento de Patología, Facultad de Medicina, Universidad de Valencia/Instituto de Investigaciones Sanitarias INCLIVA, Valencia, España; CIBERONC, Madrid, España
| | - Esther Gamero-Sandemetrio
- Departamento de Patología, Facultad de Medicina, Universidad de Valencia/Instituto de Investigaciones Sanitarias INCLIVA, Valencia, España; CIBERONC, Madrid, España
| | | | - Tomás Álvaro Naranjo
- CIBERONC, Madrid, España; Hospital Verge de la Cinta, Tortosa, Tarragona, España.
| |
Collapse
|
18
|
Fostok SF, El-Sibai M, El-Sabban M, Talhouk RS. Gap Junctions and Wnt Signaling in the Mammary Gland: a Cross-Talk? J Mammary Gland Biol Neoplasia 2019; 24:17-38. [PMID: 30194659 DOI: 10.1007/s10911-018-9411-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/03/2018] [Indexed: 12/21/2022] Open
Abstract
Connexins (Cxs), the building blocks of gap junctions (GJs), exhibit spatiotemporal patterns of expression and regulate the development and differentiation of the mammary gland, acting via channel-dependent and channel-independent mechanisms. Impaired Cx expression and localization are reported in breast cancer, suggesting a tumor suppressive role for Cxs. The signaling events that mediate the role of GJs in the development and tumorigenesis of the mammary gland remain poorly identified. The Wnt pathways, encompassing the canonical or the Wnt/β-catenin pathway and the noncanonical β-catenin-independent pathway, also play important roles in those processes. Indeed, aberrant Wnt signaling is associated with breast cancer. Despite the coincident roles of Cxs and Wnt pathways, the cross-talk in the breast tissue is poorly defined, although this is reported in a number of other tissues. Our previous studies revealed a channel-independent role for Cx43 in inducing differentiation or suppressing tumorigenesis of mammary epithelial cells by acting as a negative regulator of the Wnt/β-catenin pathway. Here, we provide a brief overview of mammary gland development, with emphasis on the role of Cxs in development and tumorigenesis of this tissue. We also discuss the role of Wnt signaling in similar contexts, and review the literature illustrating interplay between Cxs and Wnt pathways.
Collapse
Affiliation(s)
- Sabreen F Fostok
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut (AUB), P.O. Box: 11-0236, Beirut, Lebanon
| | - Mirvat El-Sibai
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University (LAU), Beirut, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut (AUB), Beirut, Lebanon
| | - Rabih S Talhouk
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut (AUB), P.O. Box: 11-0236, Beirut, Lebanon.
| |
Collapse
|
19
|
Plotkin LI, Bruzzaniti A. Molecular signaling in bone cells: Regulation of cell differentiation and survival. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 116:237-281. [PMID: 31036293 PMCID: PMC7416488 DOI: 10.1016/bs.apcsb.2019.01.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The achievement of proper bone mass and architecture, and their maintenance throughout life requires the concerted actions of osteoblasts, the bone forming cells, and osteoclasts, the bone resorbing cells. The differentiation and activity of osteoblasts and osteoclasts are regulated by molecules produced by matrix-embedded osteocytes, as well as by cross talk between osteoblasts and osteoclasts through secreted factors. In addition, it is likely that direct contact between osteoblast and osteoclast precursors, and the contact of these cells with osteocytes and cells in the bone marrow, also modulates bone cell differentiation and function. With the advancement of molecular and genetic tools, our comprehension of the intracellular signals activated in bone cells has evolved significantly, from early suggestions that osteoblasts and osteoclasts have common precursors and that osteocytes are inert cells in the bone matrix, to the very sophisticated understanding of a network of receptors, ligands, intracellular kinases/phosphatases, transcription factors, and cell-specific genes that are known today. These advances have allowed the design and FDA-approval of new therapies to preserve and increase bone mass and strength in a wide variety of pathological conditions, improving bone health from early childhood to the elderly. We have summarized here the current knowledge on selected intracellular signal pathways activated in osteoblasts, osteocytes, and osteoclasts.
Collapse
Affiliation(s)
- Lilian I Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States; Indiana Center for Musculoskeletal Health, Indianapolis, IN, United States; Roudebush Veterans Administration Medical Center, Indianapolis, IN, United States.
| | - Angela Bruzzaniti
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States; Indiana Center for Musculoskeletal Health, Indianapolis, IN, United States; Department of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, IN, United States
| |
Collapse
|
20
|
Gupta A, Leser JM, Gould NR, Buo AM, Moorer MC, Stains JP. Connexin43 regulates osteoprotegerin expression via ERK1/2 -dependent recruitment of Sp1. Biochem Biophys Res Commun 2019; 509:728-733. [PMID: 30626485 DOI: 10.1016/j.bbrc.2018.12.173] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 12/27/2018] [Indexed: 02/08/2023]
Abstract
In bone, connexin43 expression in cells of the osteoblast lineage plays an important role in restraining osteoclastogenesis and bone resorption. While there is a consensus around the notion that the anti-osteoclastogenic factor, osteoprotegerin, is a driver of this effect, how connexin43 regulates osteoprotegerin gene expression is unclear. Here, we show that loss of connexin43 decreased osteoprotegerin gene expression and reduced ERK1/2 activation. Conversely, overexpression of connexin43 increased osteoprotegerin expression and enhanced ERK1/2 activation. This increase in phospho-ERK1/2 is required for connexin43 to induce transcription from the osteoprotegerin proximal promoter. Connexin43 increased promoter activity via a specific 200 base pair region of the osteoprotegerin promoter located at -1486 to -1286 with respect to the transcriptional start site, a region which includes four Sp1 binding elements. Further, activation of this promoter region required an intact functional connexin43, as hypomorphic or dominant negative connexin43 mutant constructs, including one with increased hemichannel activity, were unable to stimulate osteoprotegerin expression as strongly as wild type connexin43. Using chromatin immunoprecipitations, we show that connexin43 expression enhanced the recruitment of Sp1, but not Runx2, to the osteoprotegerin proximal promoter. In total, these data show that connexin43-dependent gap junctional communication among osteoblast cells permits efficient ERK1/2 activation. ERK1/2 signaling promotes the recruitment of the potent transcriptional activator, Sp1, to the osteoprotegerin proximal promoter, resulting in robust transcription of anti-osteoclastogenic factor, osteoprotegerin.
Collapse
Affiliation(s)
- Aditi Gupta
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, United States.
| | - Jenna M Leser
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, United States.
| | - Nicole R Gould
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, United States.
| | - Atum M Buo
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, United States.
| | - Megan C Moorer
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, United States.
| | - Joseph P Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
21
|
Targeting of chondrocyte plasticity via connexin43 modulation attenuates cellular senescence and fosters a pro-regenerative environment in osteoarthritis. Cell Death Dis 2018; 9:1166. [PMID: 30518918 PMCID: PMC6281585 DOI: 10.1038/s41419-018-1225-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 10/26/2018] [Accepted: 11/12/2018] [Indexed: 12/12/2022]
Abstract
Osteoarthritis (OA), a chronic disease characterized by articular cartilage degeneration, is a leading cause of disability and pain worldwide. In OA, chondrocytes in cartilage undergo phenotypic changes and senescence, restricting cartilage regeneration and favouring disease progression. Similar to other wound-healing disorders, chondrocytes from OA patients show a chronic increase in the gap junction channel protein connexin43 (Cx43), which regulates signal transduction through the exchange of elements or recruitment/release of signalling factors. Although immature or stem-like cells are present in cartilage from OA patients, their origin and role in disease progression are unknown. In this study, we found that Cx43 acts as a positive regulator of chondrocyte-mesenchymal transition. Overactive Cx43 largely maintains the immature phenotype by increasing nuclear translocation of Twist-1 and tissue remodelling and proinflammatory agents, such as MMPs and IL-1β, which in turn cause cellular senescence through upregulation of p53, p16INK4a and NF-κB, contributing to the senescence-associated secretory phenotype (SASP). Downregulation of either Cx43 by CRISPR/Cas9 or Cx43-mediated gap junctional intercellular communication (GJIC) by carbenoxolone treatment triggered rediferentiation of osteoarthritic chondrocytes into a more differentiated state, associated with decreased synthesis of MMPs and proinflammatory factors, and reduced senescence. We have identified causal Cx43-sensitive circuit in chondrocytes that regulates dedifferentiation, redifferentiation and senescence. We propose that chondrocytes undergo chondrocyte-mesenchymal transition where increased Cx43-mediated GJIC during OA facilitates Twist-1 nuclear translocation as a novel mechanism involved in OA progression. These findings support the use of Cx43 as an appropriate therapeutic target to halt OA progression and to promote cartilage regeneration.
Collapse
|
22
|
Arshad M, Conzelmann C, Riaz MA, Noll T, Gündüz D. Inhibition of Cx43 attenuates ERK1/2 activation, enhances the expression of Cav‑1 and suppresses cell proliferation. Int J Mol Med 2018; 42:2811-2818. [PMID: 30132504 DOI: 10.3892/ijmm.2018.3828] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 07/31/2018] [Indexed: 11/06/2022] Open
Abstract
In addition to being an important component of the gap junction, connexin 43 (Cx43) has been shown to regulate other cellular functions, including cell proliferation. This regulatory role of Cx43 may be important in therapeutic situations, including wound healing or ischemic injuries. Caveolin‑1 (Cav‑1) has been shown to regulate angiogenesis. The aim of the present study was to analyze whether Cx43 counter‑regulates Cav‑1 in controlling the proliferation and migration of endothelial cells. The inhibition of Cx43 with niflumic acid, flufenamic acid and 18‑α‑glycyrrhetinic acid in cultured human umbilical vein endothelial cells resulted in decreased phosphorylation of extracellular signal‑regulated kinase (ERK)1/2 and increased expression of Cav‑1, as shown by western blot analysis. Furthermore, the inhibition of Cx43 resulted in a 50±7% decrease in cell proliferation, determined using a crystal violet assay, a 48±5% decrease in migration, determined using a migration assay, and a 49±6% decrease in endothelial tube formation, determined using a Matrigel assay, compared with the control. Similar results were obtained following specific inhibition of Cx43 by mimetic peptides (Gap26 and Gap27). Inhibition of the mitogen‑activated protein kinase kinase/ERK pathway with PD‑98059 resulted in an increased expression of Cav‑1 and a reduction in the expression of Cx43. Furthermore, cell proliferation, migration and tube formation in endothelial cells were impaired. By contrast, downregulation of the protein expression of Cav‑1 by small interference RNA resulted in increased expression of Cx43 and phosphorylation of ERK1/2. Accordingly, the number of cells in the Cav‑1 treated‑group increased by 35±5% compared with the controls. The data of the present study showed that Cav‑1 suppressed cell proliferation by inhibiting the activity of Cx43, which is upstream of ERK1/2. The downregulation of Cav‑1 protein resulted in loss of the inhibitory activity of Cav‑1 on cell proliferation and led to increased cell proliferation. This counter‑regulatory effect of Cx43 may be of importance in therapeutic angiogenesis.
Collapse
Affiliation(s)
- Muhammad Arshad
- Department of Cardiology and Angiology, University Hospital Giessen and Marburg, D‑35392 Giessen, Germany
| | - Charlotte Conzelmann
- Department of Cardiology and Angiology, University Hospital Giessen and Marburg, D‑35392 Giessen, Germany
| | - Muhammad Assad Riaz
- Department of Radiotherapy, University Hospital Essen, D‑45147 Essen, Germany
| | - Thomas Noll
- Institute of Physiology, Carl Gustav Carus Technical University of Dresden, D‑01307 Dresden, Germany
| | - Dursun Gündüz
- Department of Cardiology and Angiology, University Hospital Giessen and Marburg, D‑35392 Giessen, Germany
| |
Collapse
|
23
|
Yan Z, Wang P, Wu J, Feng X, Cai J, Zhai M, Li J, Liu X, Jiang M, Luo E, Jing D. Fluid shear stress improves morphology, cytoskeleton architecture, viability, and regulates cytokine expression in a time-dependent manner in MLO-Y4 cells. Cell Biol Int 2018; 42:1410-1422. [DOI: 10.1002/cbin.11032] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/13/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Zedong Yan
- Department of Biomedical Engineering; Fourth Military Medical University; Xi'an China
| | - Pan Wang
- Department of Biomedical Engineering; Fourth Military Medical University; Xi'an China
| | - Junjie Wu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases; Department of Orthodontics; School of Stomatology; Fourth Military Medical University; Xi'an China
| | - Xue Feng
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases; Department of Orthodontics; School of Stomatology; Fourth Military Medical University; Xi'an China
| | - Jing Cai
- College of Basic Medicine; Shaanxi University of Chinese Medicine; Xianyang China
| | - Mingming Zhai
- Department of Biomedical Engineering; Fourth Military Medical University; Xi'an China
| | - Juan Li
- Department of Neurosurgery; Xijing Hospital; Fourth Military Medical University; Xi'an China
| | - Xiyu Liu
- Department of Biomedical Engineering; Fourth Military Medical University; Xi'an China
| | - Maogang Jiang
- Department of Biomedical Engineering; Fourth Military Medical University; Xi'an China
| | - Erping Luo
- Department of Biomedical Engineering; Fourth Military Medical University; Xi'an China
| | - Da Jing
- Department of Biomedical Engineering; Fourth Military Medical University; Xi'an China
| |
Collapse
|
24
|
Fan X, Teng Y, Ye Z, Zhou Y, Tan WS. The effect of gap junction-mediated transfer of miR-200b on osteogenesis and angiogenesis in a co-culture of MSCs and HUVECs. J Cell Sci 2018; 131:jcs.216135. [PMID: 29898921 DOI: 10.1242/jcs.216135] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 06/04/2018] [Indexed: 12/15/2022] Open
Abstract
For successful engineering of pre-vascularized bone tissue in vitro, understanding the interactions between vasculogenic cells and bone-forming cells is a prerequisite. Mounting evidence indicates that microRNAs can serve as intercellular signals that allow cell-cell communication. Here, the role of the transfer of the microRNA miR-200b between vasculogenic and osteogenic cells was explored in a co-culture system. Rat bone-marrow derived mesenchymal stem cells (BMSCs) formed functional gap junctions composed of connexin 43 (Cx43, also known as GJA1) with human umbilical vein endothelial cells (HUVECs), through which miR-200b could transfer from BMSCs to HUVECs to regulate osteogenesis and angiogenesis. As a negative regulator, the decrease in miR-200b level in BMSCs derepressed the expression of VEGF-A, leading to increased osteogenic differentiation. Once inside HUVECs, miR-200b reduced the angiogenic potential of HUVECs through downregulation of ZEB2, ETS1, KDR and GATA2 Additionally, TGF-β was found to trigger the transfer of miR-200b to HUVECs. Upon adding the TGF-β inhibitor SB431542 or TGF-β-neutralizing antibody, the formation of capillary-like structures in co-culture could be partially rescued. These findings may be fundamental to the development of a cell-based bone regeneration strategy.
Collapse
Affiliation(s)
- Xiaoting Fan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Yi Teng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Zhaoyang Ye
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Yan Zhou
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Wen-Song Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, P. R. China
| |
Collapse
|
25
|
Kuang JY, Guo YF, Chen Y, Wang J, Duan JJ, He XL, Li L, Yu SC, Bian XW. Connexin 43 C-terminus directly inhibits the hyperphosphorylation of Akt/ERK through protein-protein interactions in glioblastoma. Cancer Sci 2018; 109:2611-2622. [PMID: 29931708 PMCID: PMC6113504 DOI: 10.1111/cas.13707] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/05/2018] [Accepted: 06/07/2018] [Indexed: 12/16/2022] Open
Abstract
Although the deregulation of epidermal growth factor receptor (EGFR) is one of the most common molecular mechanisms of glioblastoma (GBM) pathogenesis, the efficacy of anti-EGFR therapy is limited. Additionally, response to anti-EGFR therapy is not solely dependent on EGFR expression and is more promising in patients with reduced activity of EGFR downstream signaling pathways. Thus, there is considerable interest in identifying the compensatory regulatory factors of the EGFR signaling pathway to improve the efficacy of anti-EGFR therapies for GBM. In this study, we confirmed the low efficacy of EGFR inhibitors in GBM patients by meta-analysis. We then identified a negative correlation between connexin 43 (Cx43) expression and Akt/ERK activation, which was caused by the direct interactions between Akt/ERK and Cx43. By comparing the interactions between Akt/ERK and Cx43 using a series of truncated and mutated Cx43 variants, we revealed that the residues T286/A305/Q308/Y313 and S272/S273 at the carboxy terminus of Cx43 are critical for its binding with Akt and ERK, respectively. In addition, Kaplan-Meier survival analysis using data from The Cancer Genome Atlas datasets indicated that the expression of Cx43 significantly improved the prognosis of GBM patients who express EGFR. Together, our results suggested that Cx43 acts as an inhibitory regulator of the activation of growth factor receptor downstream signaling pathways, indicating the potential of Cx43 as a marker for predicting the efficacy of EGFR inhibitor treatments for GBM. Targeting the interaction between the carboxy terminus of Cx43 and Akt/ERK could be an effective therapeutic strategy against GBM.
Collapse
Affiliation(s)
- Jing-Ya Kuang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Yu-Feng Guo
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Ying Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Jun Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Jiang-Jie Duan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Xiao-Li He
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Lin Li
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Shi-Cang Yu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.,Key Laboratory of Tumor Immunology and Pathology of Ministry of Education, Chongqing, China
| |
Collapse
|
26
|
Moore D, Walker SI, Levin M. Cancer as a disorder of patterning information: computational and biophysical perspectives on the cancer problem. CONVERGENT SCIENCE PHYSICAL ONCOLOGY 2017. [DOI: 10.1088/2057-1739/aa8548] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
27
|
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: 12] [Impact Index Per Article: 1.5] [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
|
28
|
Zhao Y, Lai Y, Ge H, Guo Y, Feng X, Song J, Wang Q, Fan L, Peng Y, Cao M, Harris AL, Wang X, Tao L. Non-junctional Cx32 mediates anti-apoptotic and pro-tumor effects via epidermal growth factor receptor in human cervical cancer cells. Cell Death Dis 2017; 8:e2773. [PMID: 28492539 PMCID: PMC5520707 DOI: 10.1038/cddis.2017.183] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 03/17/2017] [Accepted: 03/21/2017] [Indexed: 12/13/2022]
Abstract
The role of connexin proteins (Cx), which form gap junctions (GJ), in progression and chemotherapeutic sensitivity of cervical cancer (CaCx), is unclear. Using cervix specimens (313 CaCx, 78 controls) and CaCx cell lines, we explored relationships among Cx expression, prognostic variables and mechanisms that may link them. In CaCx specimens, Cx32 was upregulated and cytoplasmically localized, and three other Cx downregulated, relative to controls. Cx32 expression correlated with advanced FIGO staging, differentiation and increased tumor size. In CaCx cell lines, Cx32 expression suppressed streptonigrin/cisplatin-induced apoptosis in the absence of functional GJ. In CaCx specimens and cell lines, expression of Cx32 upregulated epidermal growth factor receptor (EGFR) expression. Inhibition of EGFR signaling abrogated the anti-apoptotic effect of Cx32 expression. In conclusion, upregulated Cx32 in CaCx cells produces anti-apoptotic, pro-tumorigenic effects in vivo and vitro. Abnormal Cx32 expression/localization in CaCx appears to be both a mechanism and biomarker of chemotherapeutic resistance.
Collapse
Affiliation(s)
- Yifan Zhao
- Department of Pharmacology, Zhongshan
School of Medicine, Sun Yat-Sen University, Guangzhou
510080, China
- Department of Anesthesiology, Sun Yat-Sen
Memorial Hospital, Sun Yat-Sen University, Guangzhou
510120, China
| | - Yongchang Lai
- Department of Pharmacology, Zhongshan
School of Medicine, Sun Yat-Sen University, Guangzhou
510080, China
| | - Hui Ge
- Tumor Research Institute, Xinjiang
Medical University Affiliated Tumor Hospital, Urumqi,
Xinjiang
830000, China
| | - Yunquan Guo
- Department of Pathology, Xinjiang Medical
University Affiliated Tumor Hospital, Urumqi,
Xinjiang
830000, China
| | - Xue Feng
- Tumor Research Institute, Xinjiang
Medical University Affiliated Tumor Hospital, Urumqi,
Xinjiang
830000, China
| | - Jia Song
- Tumor Research Institute, Xinjiang
Medical University Affiliated Tumor Hospital, Urumqi,
Xinjiang
830000, China
| | - Qin Wang
- Department of Pharmacology, Zhongshan
School of Medicine, Sun Yat-Sen University, Guangzhou
510080, China
| | - Lixia Fan
- Department of Pharmacology, Zhongshan
School of Medicine, Sun Yat-Sen University, Guangzhou
510080, China
| | - Yuexia Peng
- Department of Pharmacology, Zhongshan
School of Medicine, Sun Yat-Sen University, Guangzhou
510080, China
| | - Minghui Cao
- Department of Anesthesiology, Sun Yat-Sen
Memorial Hospital, Sun Yat-Sen University, Guangzhou
510120, China
| | - Andrew L Harris
- Department of Pharmacology, Physiology
and Neuroscience, New Jersey Medical School - Rutgers University,
Newark, NJ
07103, USA
| | - Xiyan Wang
- Tumor Research Institute, Xinjiang
Medical University Affiliated Tumor Hospital, Urumqi,
Xinjiang
830000, China
| | - Liang Tao
- Department of Pharmacology, Zhongshan
School of Medicine, Sun Yat-Sen University, Guangzhou
510080, China
| |
Collapse
|
29
|
Mathews J, Levin M. Gap junctional signaling in pattern regulation: Physiological network connectivity instructs growth and form. Dev Neurobiol 2017; 77:643-673. [PMID: 27265625 PMCID: PMC10478170 DOI: 10.1002/dneu.22405] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 12/19/2022]
Abstract
Gap junctions (GJs) are aqueous channels that allow cells to communicate via physiological signals directly. The role of gap junctional connectivity in determining single-cell functions has long been recognized. However, GJs have another important role: the regulation of large-scale anatomical pattern. GJs are not only versatile computational elements that allow cells to control which small molecule signals they receive and emit, but also establish connectivity patterns within large groups of cells. By dynamically regulating the topology of bioelectric networks in vivo, GJs underlie the ability of many tissues to implement complex morphogenesis. Here, a review of recent data on patterning roles of GJs in growth of the zebrafish fin, the establishment of left-right patterning, the developmental dysregulation known as cancer, and the control of large-scale head-tail polarity, and head shape in planarian regeneration has been reported. A perspective in which GJs are not only molecular features functioning in single cells, but also enable global neural-like dynamics in non-neural somatic tissues has been proposed. This view suggests a rich program of future work which capitalizes on the rapid advances in the biophysics of GJs to exploit GJ-mediated global dynamics for applications in birth defects, regenerative medicine, and morphogenetic bioengineering. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 643-673, 2017.
Collapse
Affiliation(s)
- Juanita Mathews
- Department of Biology, Tufts Center for Regenerative and Developmental Biology, Tufts University, Medford, MA
| | - Michael Levin
- Department of Biology, Tufts Center for Regenerative and Developmental Biology, Tufts University, Medford, MA
| |
Collapse
|
30
|
Liu Z, Xu D, Wang S, Chen Y, Li Z, Gao X, Jiang L, Tang Y, Peng Y. Astrocytes induce proliferation of oligodendrocyte progenitor cells via connexin 47-mediated activation of the ERK/Id4 pathway. Cell Cycle 2017; 16:714-722. [PMID: 28278052 DOI: 10.1080/15384101.2017.1295183] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
The proliferative ability of oligodendrocyte progenitor cells (OPCs) varied markedly under different culture conditions. Astrocytes (ASTs) have been verified to play a major role in regulating the proliferation of OPCs through direct contact. However, the mechanisms have not been fully clarified. To investigate the effect and mechanism under AST and OPC co-culture conditions, we analyzed all connexins comprehensively in OPCs under OPC mono-culture, AST-secreted cell factor co-culture and AST-OPC direct-contact co-culture, and found that significantly differentially expressed Cx47 was the most significant. To assess whether Cx47 plays a role in proliferation, Cx47 siRNA were conducted. The result indicates that the cell cycle of OPCs was changed, and the cell proliferation was markedly inhibited. Kyoto Encyclopedia of Genes and Genomes (KEGG) predictive analysis suggested that Cx47 regulate cell cycle and proliferation by Ca2+ activation of ERK1/2. To verify the prediction, flow cytometry, confocal microscopy, 5-ethynyl-2'-deoxyuridine (EdU), polymerase chain reaction (RT-PCR) and western blot were used. The results show that interference of Cx47 led to decreased Ca2+ concentrations, lower p-ERK 1/2 levels, reduced transcription factor inhibitor of DNA binding 4 (Id4) expression, arrested cell cycle and reduced OPCs proliferative ability. Additionally, blocking ERK1/2 signaling caused decreased Id4 expression, arrested cell cycle in G1 phase, and reduced OPCs proliferative ability. In conclusion, ASTs can cause Ca2+ signaling activation, ERK1/2 phosphorylation, and Id4 expression stimulation in OPCs, inducing proliferation of these cells, mainly through Cx47.
Collapse
Affiliation(s)
- Zhaoyu Liu
- a Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology , Chongqing Medical University , Chongqing , P.R. China
| | - Dan Xu
- b Department of Neurosurgery , The First Affiliated Hospital of Chongqing Medical University , Chongqing , P.R. China
| | - Shang Wang
- a Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology , Chongqing Medical University , Chongqing , P.R. China
| | - Yi Chen
- a Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology , Chongqing Medical University , Chongqing , P.R. China
| | - Zhen Li
- a Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology , Chongqing Medical University , Chongqing , P.R. China
| | - Xiaoyan Gao
- a Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology , Chongqing Medical University , Chongqing , P.R. China
| | - Lu Jiang
- a Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology , Chongqing Medical University , Chongqing , P.R. China
| | - Yong Tang
- a Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology , Chongqing Medical University , Chongqing , P.R. China
| | - Yan Peng
- a Laboratory of Stem Cell and Tissue Engineering, Department of Histology and Embryology , Chongqing Medical University , Chongqing , P.R. China
| |
Collapse
|
31
|
Abstract
PURPOSE OF THE REVIEW This review highlights recent developments into how intercellular communication through connexin43 facilitates bone modeling and remodeling. RECENT FINDINGS Connexin43 is required for both skeletal development and maintenance, particularly in cortical bone, where it carries out multiple functions, including preventing osteoclastogenesis, restraining osteoprogenitor proliferation, promoting osteoblast differentiation, coordinating organized collagen matrix deposition, and maintaining osteocyte survival. Emerging data shows that connexin43 regulates both the exchange of small molecules among osteoblast lineage cells and the docking of signaling proteins to the gap junction, affecting the efficiency of signal transduction. Understanding how and what connexin43 communicates to coordinate tissue remodeling has therapeutic implications in bone. Altering the information shared by intercellular communication and/or targeting the recruitment of signaling machinery to the gap junction could be used to impact the skeletal homeostatic set point, either driving osteogenesis or inhibiting resorption.
Collapse
Affiliation(s)
- Megan C Moorer
- Department of Orthopaedics, University of Maryland School of Medicine, 100 Penn Street, Allied Health Building, Room 540E, Baltimore, MD, 21201, USA
| | - Joseph P Stains
- Department of Orthopaedics, University of Maryland School of Medicine, 100 Penn Street, Allied Health Building, Room 540E, Baltimore, MD, 21201, USA.
| |
Collapse
|
32
|
Inhibition of connexin 43 prevents trauma-induced heterotopic ossification. Sci Rep 2016; 6:37184. [PMID: 27849058 PMCID: PMC5111117 DOI: 10.1038/srep37184] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 10/26/2016] [Indexed: 12/14/2022] Open
Abstract
Heterotopic ossification (HO) can result from traumatic injury, surgery or genetic diseases. Here, we demonstrate that overexpression of connexin 43 (Cx43) is critical for the development and recurrence of traumatic HO in patients. Inhibition of Cx43 by shRNA substantially suppressed the osteogenic differentiation of MC-3T3 cells and the expression of osteogenic genes. We employed a tenotomy mouse model to explore the hypothesis that Cx43 is vital to the development of HO. Inhibition of Cx43 by a specific shRNA decreased extraskeletal bone formation in vivo. In addition, we demonstrated that ERK signaling activated by Cx43 plays an important role in promoting HO. ERK signaling was highly activated in HO tissue collected from patient and mouse models. Importantly, de novo soft tissue HO was significantly attenuated in mice treated with U0126. Inhibition of Cx43 and ERK led to decreased expressions of Runx2, BSP and Col-1 in vivo and in vitro. Moreover, HO patients with low Cx43 expression or ERK activation had a lower risk of recurrence after the lesions were surgically removed. Our findings indicate that Cx43 promotes trauma-induced HO formation by activating the ERK pathway and enhances the expression of osteogenic markers.
Collapse
|
33
|
Shi C, Zhang H, Louie K, Mishina Y, Sun H. BMP Signaling Mediated by BMPR1A in Osteoclasts Negatively Regulates Osteoblast Mineralization Through Suppression of Cx43. J Cell Biochem 2016; 118:605-614. [PMID: 27649478 DOI: 10.1002/jcb.25746] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/19/2016] [Indexed: 12/23/2022]
Abstract
Osteoblasts and osteoclasts are well orchestrated through different mechanisms of communication during bone remodeling. Previously, we found that osteoclast-specific disruption of one of the BMP receptors, Bmpr1a, results in increased osteoblastic bone formation in mice. We hypothesized that BMPR1A signaling in osteoclasts regulates production of either membrane bound proteins or secreted molecules that regulated osteoblast differentiation. In our current study, we co-cultured wild-type osteoblasts with either control osteoclasts or osteoclasts lacking BMPR1A signaling activity. We found that loss of Bmpr1a in osteoclasts promoted osteoblast mineralization in vitro. Further, we found that the expression of Cx43/Gja1 in the mutant osteoclasts was increased, which encoded for one of the gap junction proteins connexin 43/gap junction alpha 1. Knockdown of Gja1 in the mutant osteoclasts for Bmpr1a reduced osteoblastic mineralization when co-cultured. Our findings suggest that GJA1 may be one of the downstream targets of BMPR1A signaling in osteoclasts that mediates osteoclast-osteoblast communication during bone remodeling. J. Cell. Biochem. 118: 605-614, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ce Shi
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun, Jilin Province, 130021, China.,Department of Biologic and Materials Sciences, University of Michigan, School of Dentistry, Ann Arbor, Michigan, 48109-1078
| | - Honghao Zhang
- Department of Biologic and Materials Sciences, University of Michigan, School of Dentistry, Ann Arbor, Michigan, 48109-1078
| | - Ke'ale Louie
- Department of Biologic and Materials Sciences, University of Michigan, School of Dentistry, Ann Arbor, Michigan, 48109-1078
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, University of Michigan, School of Dentistry, Ann Arbor, Michigan, 48109-1078
| | - Hongchen Sun
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun, Jilin Province, 130021, China
| |
Collapse
|
34
|
Abstract
Shaping of the skeleton (modeling) and its maintenance throughout life (remodeling) require coordinated activity among bone forming (osteoblasts) and resorbing cells (osteoclasts) and osteocytes (bone embedded cells). The gap junction protein connexin43 (Cx43) has emerged as a key modulator of skeletal growth and homeostasis. The skeletal developmental abnormalities present in oculodentodigital and craniometaphyseal dysplasias, both linked to Cx43 gene (GJA1) mutations, demonstrate that the skeleton is a major site of Cx43 action. Via direct action on osteolineage cells, including altering production of pro-osteoclastogenic factors, Cx43 contributes to peak bone mass acquisition, cortical modeling of long bones, and maintenance of bone quality. Cx43 also contributes in diverse ways to bone responsiveness to hormonal and mechanical signals. Skeletal biology research has revealed the complexity of Cx43 function; in addition to forming gap junctions and "hemichannels", Cx43 provides a scaffold for signaling molecules. Hence, Cx43 actively participates in generation and modulation of cellular signals driving skeletal development and homeostasis. Pharmacological interference with Cx43 may in the future help remedy deterioration of bone quality occurring with aging, disuse and hormonal imbalances.
Collapse
Affiliation(s)
- Joseph P Stains
- Department of Orthopaedics, University of Maryland, School of Medicine, Baltimore, MD, United States
| | - Roberto Civitelli
- Division of Bone and Mineral Diseases, Department of Internal Medicine, Washington University in St. Louis, Campus Box 8301, 425 South Euclid, St. Louis, MO 63110, United States.
| |
Collapse
|
35
|
Alqadah A, Hsieh YW, Schumacher JA, Wang X, Merrill SA, Millington G, Bayne B, Jorgensen EM, Chuang CF. SLO BK Potassium Channels Couple Gap Junctions to Inhibition of Calcium Signaling in Olfactory Neuron Diversification. PLoS Genet 2016; 12:e1005654. [PMID: 26771544 PMCID: PMC4714817 DOI: 10.1371/journal.pgen.1005654] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/16/2015] [Indexed: 01/09/2023] Open
Abstract
The C. elegans AWC olfactory neuron pair communicates to specify asymmetric subtypes AWCOFF and AWCON in a stochastic manner. Intercellular communication between AWC and other neurons in a transient NSY-5 gap junction network antagonizes voltage-activated calcium channels, UNC-2 (CaV2) and EGL-19 (CaV1), in the AWCON cell, but how calcium signaling is downregulated by NSY-5 is only partly understood. Here, we show that voltage- and calcium-activated SLO BK potassium channels mediate gap junction signaling to inhibit calcium pathways for asymmetric AWC differentiation. Activation of vertebrate SLO-1 channels causes transient membrane hyperpolarization, which makes it an important negative feedback system for calcium entry through voltage-activated calcium channels. Consistent with the physiological roles of SLO-1, our genetic results suggest that slo-1 BK channels act downstream of NSY-5 gap junctions to inhibit calcium channel-mediated signaling in the specification of AWCON. We also show for the first time that slo-2 BK channels are important for AWC asymmetry and act redundantly with slo-1 to inhibit calcium signaling. In addition, nsy-5-dependent asymmetric expression of slo-1 and slo-2 in the AWCON neuron is necessary and sufficient for AWC asymmetry. SLO-1 and SLO-2 localize close to UNC-2 and EGL-19 in AWC, suggesting a role of possible functional coupling between SLO BK channels and voltage-activated calcium channels in AWC asymmetry. Furthermore, slo-1 and slo-2 regulate the localization of synaptic markers, UNC-2 and RAB-3, in AWC neurons to control AWC asymmetry. We also identify the requirement of bkip-1, which encodes a previously identified auxiliary subunit of SLO-1, for slo-1 and slo-2 function in AWC asymmetry. Together, these results provide an unprecedented molecular link between gap junctions and calcium pathways for terminal differentiation of olfactory neurons.
Collapse
Affiliation(s)
- Amel Alqadah
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Yi-Wen Hsieh
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Jennifer A. Schumacher
- Division of Developmental Biology, Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Xiaohong Wang
- Division of Developmental Biology, Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Sean A. Merrill
- Department of Biology and Howard Hughes Medical Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Grethel Millington
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Brittany Bayne
- Division of Developmental Biology, Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Erik M. Jorgensen
- Department of Biology and Howard Hughes Medical Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Chiou-Fen Chuang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
36
|
Yamada A, Futagi M, Fukumoto E, Saito K, Yoshizaki K, Ishikawa M, Arakaki M, Hino R, Sugawara Y, Ishikawa M, Naruse M, Miyazaki K, Nakamura T, Fukumoto S. Connexin 43 Is Necessary for Salivary Gland Branching Morphogenesis and FGF10-induced ERK1/2 Phosphorylation. J Biol Chem 2015; 291:904-12. [PMID: 26565022 DOI: 10.1074/jbc.m115.674663] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Indexed: 11/06/2022] Open
Abstract
Cell-cell interaction via the gap junction regulates cell growth and differentiation, leading to formation of organs of appropriate size and quality. To determine the role of connexin43 in salivary gland development, we analyzed its expression in developing submandibular glands (SMGs). Connexin43 (Cx43) was found to be expressed in salivary gland epithelium. In ex vivo organ cultures of SMGs, addition of the gap junctional inhibitors 18α-glycyrrhetinic acid (18α-GA) and oleamide inhibited SMG branching morphogenesis, suggesting that gap junctional communication contributes to salivary gland development. In Cx43(-/-) salivary glands, submandibular and sublingual gland size was reduced as compared with those from heterozygotes. The expression of Pdgfa, Pdgfb, Fgf7, and Fgf10, which induced branching of SMGs in Cx43(-/-) samples, were not changed as compared with those from heterozygotes. Furthermore, the blocking peptide for the hemichannel and gap junction channel showed inhibition of terminal bud branching. FGF10 induced branching morphogenesis, while it did not rescue the Cx43(-/-) phenotype, thus Cx43 may regulate FGF10 signaling during salivary gland development. FGF10 is expressed in salivary gland mesenchyme and regulates epithelial proliferation, and was shown to induce ERK1/2 phosphorylation in salivary epithelial cells, while ERK1/2 phosphorylation in HSY cells was dramatically inhibited by 18α-GA, a Cx43 peptide or siRNA. On the other hand, PDGF-AA and PDGF-BB separately induced ERK1/2 phosphorylation in primary cultured salivary mesenchymal cells regardless of the presence of 18α-GA. Together, our results suggest that Cx43 regulates FGF10-induced ERK1/2 phosphorylation in salivary epithelium but not in mesenchyme during the process of SMG branching morphogenesis.
Collapse
Affiliation(s)
- Aya Yamada
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Masaharu Futagi
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Emiko Fukumoto
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Kan Saito
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Keigo Yoshizaki
- Division of Orthodontics, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Masaki Ishikawa
- Operative Dentistry, Department of Restorative Dentistry Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan and
| | - Makiko Arakaki
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Ryoko Hino
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Yu Sugawara
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Momoko Ishikawa
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Masahiro Naruse
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Kanako Miyazaki
- Division of Orthodontics, Division of Oral Health, Growth and Development, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Takashi Nakamura
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences
| | - Satoshi Fukumoto
- From the Division of Pediatric Dentistry, Department of Oral Health and Development Sciences,
| |
Collapse
|
37
|
Li S, He H, Zhang G, Wang F, Zhang P, Tan Y. Connexin43-containing gap junctions potentiate extracellular Ca2+-induced odontoblastic differentiation of human dental pulp stem cells via Erk1/2. Exp Cell Res 2015; 338:1-9. [DOI: 10.1016/j.yexcr.2015.09.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Revised: 09/08/2015] [Accepted: 09/11/2015] [Indexed: 01/09/2023]
|
38
|
Plotkin LI, Stains JP. Connexins and pannexins in the skeleton: gap junctions, hemichannels and more. Cell Mol Life Sci 2015; 72:2853-67. [PMID: 26091748 PMCID: PMC4503509 DOI: 10.1007/s00018-015-1963-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 06/11/2015] [Indexed: 10/23/2022]
Abstract
Regulation of bone homeostasis depends on the concerted actions of bone-forming osteoblasts and bone-resorbing osteoclasts, controlled by osteocytes, cells derived from osteoblasts surrounded by bone matrix. The control of differentiation, viability and function of bone cells relies on the presence of connexins. Connexin43 regulates the expression of genes required for osteoblast and osteoclast differentiation directly or by changing the levels of osteocytic genes, and connexin45 may oppose connexin43 actions in osteoblastic cells. Connexin37 is required for osteoclast differentiation and its deletion results in increased bone mass. Less is known on the role of connexins in cartilage, ligaments and tendons. Connexin43, connexin45, connexin32, connexin46 and connexin29 are expressed in chondrocytes, while connexin43 and connexin32 are expressed in ligaments and tendons. Similarly, although the expression of pannexin1, pannexin2 and pannexin3 has been demonstrated in bone and cartilage cells, their function in these tissues is not fully understood.
Collapse
Affiliation(s)
- Lilian I Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Dr., MS 5035, Indianapolis, IN, 46202, USA,
| | | |
Collapse
|
39
|
Gindin Y, Jiang Y, Francis P, Walker RL, Abaan OD, Zhu YJ, Meltzer PS. miR-23a impairs bone differentiation in osteosarcoma via down-regulation of GJA1. Front Genet 2015; 6:233. [PMID: 26191074 PMCID: PMC4488756 DOI: 10.3389/fgene.2015.00233] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 06/18/2015] [Indexed: 01/04/2023] Open
Abstract
Osteosarcoma is the most common type of bone cancer in children and adolescents. Impaired differentiation of osteoblast cells is a distinguishing feature of this aggressive disease. As improvements in survival outcomes have largely plateaued, better understanding of the bone differentiation program may provide new treatment approaches. The miRNA cluster miR-23a~27a~24-2, particularly miR-23a, has been shown to interact with genes important for bone development. However, global changes in gene expression associated with functional gain of this cluster have not been fully explored. To better understand the relationship between miR-23a expression and bone cell differentiation, we carried out a large-scale gene expression analysis in HOS cells. Experimental results demonstrate that over-expression of miR-23a delays differentiation in this system. Downstream bioinformatic analysis identified miR-23a target gene connexin-43 (Cx43/GJA1), a mediator of intercellular signaling critical to osteoblast development, as acutely affected by miR-23a levels. Connexin-43 is up-regulated in the course of HOS cell differentiation and is down-regulated in cells transfected with miR-23a. Analysis of gene expression data, housed at Gene Expression Omnibus, reveals that Cx43 is consistently up-regulated during osteoblast differentiation. Suppression of Cx43 mRNA by miR-23a was confirmed in vitro using a luciferase reporter assay. This work demonstrates novel interactions between microRNA expression, intercellular signaling and bone differentiation in osteosarcoma.
Collapse
Affiliation(s)
- Yevgeniy Gindin
- Genetics Branch, Center for Cancer Research, National Institutes of Health Bethesda, MD, USA ; Graduate Program in Bioinformatics, Boston University Boston, MA, USA
| | - Yuan Jiang
- Genetics Branch, Center for Cancer Research, National Institutes of Health Bethesda, MD, USA
| | - Princy Francis
- Genetics Branch, Center for Cancer Research, National Institutes of Health Bethesda, MD, USA
| | - Robert L Walker
- Genetics Branch, Center for Cancer Research, National Institutes of Health Bethesda, MD, USA
| | - Ogan D Abaan
- Genetics Branch, Center for Cancer Research, National Institutes of Health Bethesda, MD, USA
| | - Yuelin J Zhu
- Genetics Branch, Center for Cancer Research, National Institutes of Health Bethesda, MD, USA
| | - Paul S Meltzer
- Genetics Branch, Center for Cancer Research, National Institutes of Health Bethesda, MD, USA
| |
Collapse
|
40
|
Li S, Zhang H, Li S, Yang Y, Huo B, Zhang D. Connexin 43 and ERK regulate tension-induced signal transduction in human periodontal ligament fibroblasts. J Orthop Res 2015; 33:1008-14. [PMID: 25731708 DOI: 10.1002/jor.22830] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 01/15/2015] [Indexed: 02/04/2023]
Abstract
Periodontal ligament (PDL) fibroblasts play an important role in preserving periodontal homeostasis and transmitting mechanical signals to alveolar bone. Connexin 43 (Cx43), a gap junction protein, is essential for bone homeostasis and regulates bone remodeling. However, the function of Cx43 in human PDL fibroblast-regulated bone remodeling has not yet been elucidated. In this study, human PDL fibroblasts were exposed to cyclic mechanical tension with a maximum 5% elongation for different durations. We then examined the expression of signaling molecules related to osteogenesis and osteoclastogenesis at both the mRNA and protein levels as well as the activity of extracellular signal-regulated kinase (ERK) in human PDL fibroblasts after loading. We found that mechanical tension increased Cx43, which further upregulated osteogenic (e.g., RUNX2, Osterix, and OPG) and down-regulated osteoclastogenic (e.g., RANKL) signaling molecules. Suppressing Cx43 gene (Gja1) by siRNA inhibited the increase in osteogenesis-related molecules but enhanced RANKL expression. Similar to Cx43, activated ERK1/2 was also enhanced by mechanical tension and suppressed by Cx43 siRNA. Inhibition of ERK1/2 signaling using PD98059 reduced the tension-regulated increase in osteogenesis-related molecules but enhanced that of osteoclastogenesis-related ones. These findings suggest that cyclic tension may involve into the osteogenic or osteoclastogenetic differentiation potential of human PDL fibroblasts via the Cx43-ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Shengnan Li
- Department of Stomatology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, P.R., China
| | - Huajing Zhang
- Department of Stomatology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, P.R., China
| | - Shuna Li
- Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing, 100081, P.R., China
| | - Yanqi Yang
- Faculty of Dentistry, University of Hong Kong, Hong Kong SAR, China
| | - Bo Huo
- Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing, 100081, P.R., China
| | - Ding Zhang
- Department of Stomatology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, P.R., China
| |
Collapse
|
41
|
Zappitelli T, Aubin JE. The "connexin" between bone cells and skeletal functions. J Cell Biochem 2015; 115:1646-58. [PMID: 24818806 DOI: 10.1002/jcb.24836] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 05/08/2014] [Indexed: 11/11/2022]
Abstract
The processes of bone modeling and remodeling are crucial in the skeleton's functions as a supportive and protective structure, a mineral reservoir, and an endocrine organ. The coordination between bone cell activities (bone formation and bone resorption), necessary to maintain the integrity of the skeleton during these processes, is mediated at least in part by cell-cell and cell-environment interactions across gap junctions and hemichannels. The increasing number of genetically engineered Connexin 43 (Cx43) knockout and missense mouse models have provided insight into the complex and critical roles of Cx43-containing gap junctions and hemichannels in the development and turnover of the skeleton, in differentiation, activity and survival of the bone cell lineages, and in the cellular and molecular mechanisms by which Cx43 functions and assists in mediating cellular responses to stimuli in bone. Cx43 may be an important potential therapeutic target, making it crucial that we continue to gain understanding of the multiple and complex roles of Cx43 in bone.
Collapse
Affiliation(s)
- Tanya Zappitelli
- Department of Medical Biophysics, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada, M5S 1A8
| | | |
Collapse
|
42
|
Gago-Fuentes R, Fernández-Puente P, Megias D, Carpintero-Fernández P, Mateos J, Acea B, Fonseca E, Blanco FJ, Mayan MD. Proteomic Analysis of Connexin 43 Reveals Novel Interactors Related to Osteoarthritis. Mol Cell Proteomics 2015; 14:1831-45. [PMID: 25903580 DOI: 10.1074/mcp.m115.050211] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Indexed: 11/06/2022] Open
Abstract
We have previously reported that articular chondrocytes in tissue contain long cytoplasmic arms that physically connect two distant cells. Cell-to-cell communication occurs through connexin channels termed Gap Junction (GJ) channels, which achieve direct cellular communication by allowing the intercellular exchange of ions, small RNAs, nutrients, and second messengers. The Cx43 protein is overexpressed in several human diseases and inflammation processes and in articular cartilage from patients with osteoarthritis (OA). An increase in the level of Cx43 is known to alter gene expression, cell signaling, growth, and cell proliferation. The interaction of proteins with the C-terminal tail of connexin 43 (Cx43) directly modulates GJ-dependent and -independent functions. Here, we describe the isolation of Cx43 complexes using mild extraction conditions and immunoaffinity purification. Cx43 complexes were extracted from human primary articular chondrocytes isolated from healthy donors and patients with OA. The proteomic content of the native complexes was determined using LC-MS/MS, and protein associations with Cx43 were validated using Western blot and immunolocalization experiments. We identified >100 Cx43-associated proteins including previously uncharacterized proteins related to nucleolar functions, RNA transport, and translation. We also identified several proteins involved in human diseases, cartilage structure, and OA as novel functional Cx43 interactors, which emphasized the importance of Cx43 in the normal physiology and structural and functional integrity of chondrocytes and articular cartilage. Gene Ontology (GO) terms of the proteins identified in the OA samples showed an enrichment of Cx43-interactors related to cell adhesion, calmodulin binding, the nucleolus, and the cytoskeleton in OA samples compared with healthy samples. However, the mitochondrial proteins SOD2 and ATP5J2 were identified only in samples from healthy donors. The identification of Cx43 interactors will provide clues to the functions of Cx43 in human cells and its roles in the development of several diseases, including OA.
Collapse
Affiliation(s)
- Raquel Gago-Fuentes
- From the ‡CellCOM Research Group. Instituto de Investigación Biomédica A Coruña (INIBIC), XXIAC, University of A Coruña. Xubias de Arriba 84, 15006 A Coruña, Spain
| | - Patricia Fernández-Puente
- §Rheumatology Division, ProteoRed/ISCIII, Proteomics Group, Instituto de Investigación Biomédica A Coruña (INIBIC), XXIAC. University of A Coruña. Xubias de Arriba 84, 15006 A Coruña, Spain; ¶Rheumatology Division, CIBER-BBN/ISCIII, Instituto de Investigación Biomédica A Coruña (INIBIC), XXIAC. University of A Coruña. Xubias de Arriba 84, 15006 A Coruña, Spain
| | - Diego Megias
- ‖Confocal Microscopy Core Unit. Centro Nacional de Investigaciones Oncológicas (CNIO), 28029, Madrid, Spain
| | - Paula Carpintero-Fernández
- From the ‡CellCOM Research Group. Instituto de Investigación Biomédica A Coruña (INIBIC), XXIAC, University of A Coruña. Xubias de Arriba 84, 15006 A Coruña, Spain
| | - Jesus Mateos
- §Rheumatology Division, ProteoRed/ISCIII, Proteomics Group, Instituto de Investigación Biomédica A Coruña (INIBIC), XXIAC. University of A Coruña. Xubias de Arriba 84, 15006 A Coruña, Spain; ¶Rheumatology Division, CIBER-BBN/ISCIII, Instituto de Investigación Biomédica A Coruña (INIBIC), XXIAC. University of A Coruña. Xubias de Arriba 84, 15006 A Coruña, Spain
| | - Benigno Acea
- From the ‡CellCOM Research Group. Instituto de Investigación Biomédica A Coruña (INIBIC), XXIAC, University of A Coruña. Xubias de Arriba 84, 15006 A Coruña, Spain
| | - Eduardo Fonseca
- From the ‡CellCOM Research Group. Instituto de Investigación Biomédica A Coruña (INIBIC), XXIAC, University of A Coruña. Xubias de Arriba 84, 15006 A Coruña, Spain
| | - Francisco Javier Blanco
- §Rheumatology Division, ProteoRed/ISCIII, Proteomics Group, Instituto de Investigación Biomédica A Coruña (INIBIC), XXIAC. University of A Coruña. Xubias de Arriba 84, 15006 A Coruña, Spain; ¶Rheumatology Division, CIBER-BBN/ISCIII, Instituto de Investigación Biomédica A Coruña (INIBIC), XXIAC. University of A Coruña. Xubias de Arriba 84, 15006 A Coruña, Spain
| | - Maria Dolores Mayan
- From the ‡CellCOM Research Group. Instituto de Investigación Biomédica A Coruña (INIBIC), XXIAC, University of A Coruña. Xubias de Arriba 84, 15006 A Coruña, Spain;
| |
Collapse
|
43
|
Uzu M, Sato H, Yamada R, Kashiba T, Shibata Y, Yamaura K, Ueno K. Effect of enhanced expression of connexin 43 on sunitinib-induced cytotoxicity in mesothelioma cells. J Pharmacol Sci 2015; 128:17-26. [PMID: 26003083 DOI: 10.1016/j.jphs.2015.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 03/31/2015] [Accepted: 04/02/2015] [Indexed: 12/30/2022] Open
Abstract
Connexin (Cx) makes up a type of intercellular channel called gap junction (GJ). GJ plays a regulatory role in cellular physiology. The Cx expression level is often decreased in cancer cells compared to that in healthy ones, and the restoration of its expression has been shown to exert antiproliferative effects. This work aims to evaluate the effect of the restoration of connexin 43 (Cx43) (the most ubiquitous Cx subtype) expression on sunitinib (SU)-induced cytotoxicity in malignant mesothelioma (MM) cells. Increased Cx43 expression in an MM cell line (H28) improved the ability of SU to inhibit receptor tyrosine kinase (RTK) signaling. Moreover, higher Cx43 expression promoted SU-induced apoptosis. The cell viability test revealed that Cx43 enhanced the cytotoxic effect of SU in a GJ-independent manner. The effect of Cx43 on a proapoptotic factor, Bax, was then investigated. The interaction between Cx43 and Bax was confirmed by immunoprecipitation. Furthermore, higher Cx43 expression increased the production of a cleaved (active) form of Bax during SU-induced apoptosis with no alteration in total Bax expression. These findings indicate that Cx43 most likely increases sensitivity to SU in H28 through direct interaction with Bax. In conclusion, we found that Cx43 overcame the chemoresistance of MM cells.
Collapse
Affiliation(s)
- Miaki Uzu
- Department of Geriatric Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ward, Chiba-city, Chiba 260-8675, Japan
| | - Hiromi Sato
- Department of Geriatric Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ward, Chiba-city, Chiba 260-8675, Japan.
| | - Ryota Yamada
- Department of Geriatric Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ward, Chiba-city, Chiba 260-8675, Japan
| | - Tatsuro Kashiba
- Department of Geriatric Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ward, Chiba-city, Chiba 260-8675, Japan
| | - Yukihiro Shibata
- Department of Geriatric Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ward, Chiba-city, Chiba 260-8675, Japan
| | - Katsunori Yamaura
- Department of Geriatric Pharmacology and Therapeutics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ward, Chiba-city, Chiba 260-8675, Japan
| | - Koichi Ueno
- Center for Preventive Medical Science, Chiba University, 1-8-1 Inohana, Chuo-ward, Chiba-city, Chiba 260-8675, Japan
| |
Collapse
|
44
|
Casagrande D, Stains JP, Murthi AM. Identification of shoulder osteoarthritis biomarkers: comparison between shoulders with and without osteoarthritis. J Shoulder Elbow Surg 2015; 24:382-90. [PMID: 25595362 PMCID: PMC4331258 DOI: 10.1016/j.jse.2014.11.039] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 11/12/2014] [Accepted: 11/15/2014] [Indexed: 02/01/2023]
Abstract
BACKGROUND The biologic factors associated with shoulder osteoarthritis (OA) have not been elucidated. The purpose of this study was to investigate osteoarthritic biomarkers of the shoulder. To our knowledge, this is the first study to analyze shoulder cartilage for OA-associated genes and to examine human shoulder cartilage for a possible biomarker, connexin 43 (Cx43). MATERIALS AND METHODS Cartilage from 16 osteoarthritic and 10 nonosteoarthritic humeral heads was assessed for expression of the following genes by real-time polymerase chain reaction: types I, II, and X collagen; matrix metalloproteinases (MMPs); tissue inhibitors of MMP (TIMPs); interleukins; versican; cyclooxygenase 2 (Cox-2); inducible nitric oxide synthase (iNOS); tumor necrosis factor α (TNF-α); aggrecanase 2 (ADAMTS5); and Cx43. RESULTS In osteoarthritic shoulders, Cx43, Cox-2, versican, collagen type I, ADAMTS5, MMP-3, and TNF-α expressions were significantly increased compared with controls. TIMP-3 and iNOS trended toward significance, with robust expression in osteoarthritic shoulders and low expression in nonosteoarthritic shoulders. In osteoarthritic shoulders, gene expression of Cx43, ADAMTS5, collagen type I, Cox-2, versican, and TIMP-3 showed predominance (85-, 33-, 13-, 12-, 11.5-, and 3-fold increases, respectively) relative to nonosteoarthritic controls. Spearman correlation analysis showed significant correlations between Cx43 and collagen (types I, II, and X), MMP-9, TIMP-2 and TIMP-3, versican, Cox-2, iNOS, and ADAMTS5. CONCLUSIONS Certain genes are markedly upregulated in osteoarthritic shoulders compared with nonosteoarthritic shoulders, with Cx43, Cox-2, versican, collagen type I, ADAMTS5, MMP-3, and TNF-α expression being significantly increased. These genes might be useful biomarkers for examining shoulder OA. CLINICAL RELEVANCE Identification of osteoarthritic biomarkers can help us better understand shoulder OA and build the foundation for future research on disease progression and treatments.
Collapse
|
45
|
Artesi M, Kroonen J, Bredel M, Nguyen-Khac M, Deprez M, Schoysman L, Poulet C, Chakravarti A, Kim H, Scholtens D, Seute T, Rogister B, Bours V, Robe PA. Connexin 30 expression inhibits growth of human malignant gliomas but protects them against radiation therapy. Neuro Oncol 2014; 17:392-406. [PMID: 25155356 DOI: 10.1093/neuonc/nou215] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 07/28/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Glioblastomas remain ominous tumors that almost invariably escape treatment. Connexins are a family of transmembrane, gap junction-forming proteins, some members of which were reported to act as tumor suppressors and to modulate cellular metabolism in response to cytotoxic stress. METHODS We analyzed the copy number and expression of the connexin (Cx)30 gene gap junction beta-6 (GJB6), as well as of its protein immunoreactivity in several public and proprietary repositories of glioblastomas, and their influence on patient survival. We evaluated the effect of the expression of this gap junction protein on the growth, DNA repair and energy metabolism, and treatment resistance of these tumors. RESULTS The GJB6 gene was deleted in 25.8% of 751 analyzed tumors and mutated in 15.8% of 158 tumors. Cx30 immunoreactivity was absent in 28.9% of 145 tumors. Restoration of Cx30 expression in human glioblastoma cells reduced their growth in vitro and as xenografts in the striatum of immunodeficient mice. Cx30 immunoreactivity was, however, found to adversely affect survival in 2 independent retrospective cohorts of glioblastoma patients. Cx30 was found in clonogenic assays to protect glioblastoma cells against radiation-induced mortality and to decrease radiation-induced DNA damage. This radioprotection correlated with a heat shock protein 90-dependent mitochondrial translocation of Cx30 following radiation and an improved ATP production following this genotoxic stress. CONCLUSION These results underline the complex relationship between potential tumor suppressors and treatment resistance in glioblastomas and single out GJB6/Cx30 as a potential biomarker and target for therapeutic intervention in these tumors.
Collapse
Affiliation(s)
- Maria Artesi
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Jerome Kroonen
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Markus Bredel
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Minh Nguyen-Khac
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Manuel Deprez
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Laurent Schoysman
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Christophe Poulet
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Arnab Chakravarti
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Hyunsoo Kim
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Denise Scholtens
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Tatjana Seute
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Bernard Rogister
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Vincent Bours
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| | - Pierre A Robe
- Department of Human Genetics, CBIG/GIGA Research Center, University of Liège, Liège, Belgium (M.A., J.K., M.N.-K., L.S., C.P., V.B., P.A.R.); Department of Neurology and Neurosurgery and T. and P. Bonhenn Neuro-Oncology Laboratory, University Hospital of Utrecht, Utrecht, Netherlands (J.K., L.S., T.S., P.A.R.); Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, Illinois (D.S.); Center for Population Health Sciences, Institute for Public Health and Medicine, Northwestern University, Chicago (D.S.); Division of Neuropathology, University Hospital of Liège, Liège, Belgium (M.D.); Division of Neurobiology, CBIG/GIGA Research Center, University Hospital of Liège, Liège, Belgium (B.R.); Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University Medical Center, Columbus, Ohio (A.C., P.A.R.); Department of Radiation Oncology, Hazelrig-Salter Radiation Oncology Center and UAB Comprehensive Cancer Center, Birmingham, Alabama (M.B., H.K.)
| |
Collapse
|
46
|
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]
|
47
|
Abed A, Toubas J, Kavvadas P, Authier F, Cathelin D, Alfieri C, Boffa JJ, Dussaule JC, Chatziantoniou C, Chadjichristos CE. Targeting connexin 43 protects against the progression of experimental chronic kidney disease in mice. Kidney Int 2014; 86:768-79. [PMID: 24850151 DOI: 10.1038/ki.2014.108] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 02/17/2014] [Accepted: 02/20/2014] [Indexed: 11/09/2022]
Abstract
Excessive recruitment of monocytes and progression of fibrosis are hallmarks of chronic kidney disease (CKD). Recently we reported that the expression of connexin 43 (Cx43) was upregulated in the kidney during experimental nephropathy. To investigate the role of Cx43 in the progression of CKD, we interbred RenTg mice, a genetic model of hypertension-induced CKD, with Cx43+/- mice. The renal cortex of 5-month-old RenTgCx43+/- mice showed a marked decrease of cell adhesion markers leading to reduced monocyte infiltration and interstitial renal fibrosis compared with their littermates. In addition, functional and histological parameters such as albuminuria and glomerulosclerosis were ameliorated in RenTgCx43+/- mice. Interestingly, treatment with Cx43 antisense produced remarkable improvement of renal function and structure in 1-year-old RenTg mice. Similar results were found in Cx43+/- or wild-type mice treated with Cx43 antisense after obstructive nephropathy. Furthermore, in these mice, Cx43 antisense attenuated E-cadherin downregulation and phosphorylation of the transcription factor Sp1 by the ERK pathway resulting in decreased transcription of type I collagen gene. Interestingly, Cx43-specific blocking peptide inhibited monocyte adhesion in activated endothelium and profibrotic pathways in tubular cells. Cx43 was highly increased in biopsies of patients with CKD. Thus, Cx43 may represent a new therapeutic target against the progression of CKD.
Collapse
Affiliation(s)
- Ahmed Abed
- 1] INSERM UMR-S1155, Tenon Hospital, Paris, France [2] Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | - Julie Toubas
- 1] INSERM UMR-S1155, Tenon Hospital, Paris, France [2] Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | | | | | | | | | - Jean-Jacques Boffa
- 1] INSERM UMR-S1155, Tenon Hospital, Paris, France [2] Sorbonne Universités, UPMC Univ Paris 6, Paris, France [3] Department of Nephrology, Tenon Hospital, Paris, France
| | - Jean-Claude Dussaule
- 1] INSERM UMR-S1155, Tenon Hospital, Paris, France [2] Sorbonne Universités, UPMC Univ Paris 6, Paris, France [3] Department of Physiology, Saint-Antoine Hospital, Paris, France
| | - Christos Chatziantoniou
- 1] INSERM UMR-S1155, Tenon Hospital, Paris, France [2] Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | - Christos E Chadjichristos
- 1] INSERM UMR-S1155, Tenon Hospital, Paris, France [2] Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| |
Collapse
|
48
|
Buo AM, Stains JP. Gap junctional regulation of signal transduction in bone cells. FEBS Lett 2014; 588:1315-21. [PMID: 24486014 DOI: 10.1016/j.febslet.2014.01.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 01/17/2014] [Accepted: 01/20/2014] [Indexed: 11/30/2022]
Abstract
The role of gap junctions, particularly that of connexin43 (Cx43), has become an area of increasing interest in bone physiology. An abundance of studies have shown that Cx43 influences the function of osteoblasts and osteocytes, which ultimately impacts bone mass acquisition and skeletal homeostasis. However, the molecular details underlying how Cx43 regulates bone are only coming into focus and have proven to be more complex than originally thought. In this review, we focus on the diverse molecular mechanisms by which Cx43 gap junctions and hemichannels regulate cell signaling pathways, gene expression, mechanotransduction and cell survival in bone cells. This review will highlight key signaling factors that have been identified as downstream effectors of Cx43 and the impact of these pathways on distinct osteoblast and osteocyte functions.
Collapse
Affiliation(s)
- Atum M Buo
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joseph P Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
49
|
Stains JP, Watkins MP, Grimston SK, Hebert C, Civitelli R. Molecular mechanisms of osteoblast/osteocyte regulation by connexin43. Calcif Tissue Int 2014; 94:55-67. [PMID: 23754488 PMCID: PMC3815501 DOI: 10.1007/s00223-013-9742-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 04/23/2013] [Indexed: 01/23/2023]
Abstract
Osteoblasts, osteocytes, and osteoprogenitor cells are interconnected into a functional network by gap junctions formed primarily by connexin43 (Cx43). Over the past two decades, it has become clear that Cx43 is important for the function of osteoblasts and osteocytes. This connexin contributes to the acquisition of peak bone mass and is a major modulator of cortical modeling. We review key data from human and mouse genetics on the skeletal consequences of ablation or mutation of the Cx43 gene (Gja1) and the molecular mechanisms by which Cx43 regulates the differentiation, function, and survival of osteogenic lineage cells. We also discuss putative second messengers that are communicated by Cx43 gap junctions, the role of hemichannels, and the function of Cx43 as a scaffold for signaling molecules. Current knowledge demonstrates that Cx43 is more than a passive channel; rather, it actively participates in the generation and modulation of cellular signals that drive skeletal development and homeostasis.
Collapse
Affiliation(s)
- Joseph P Stains
- Department of Orthopaedics, University of Maryland, School of Medicine, 100 Penn Street, Allied Health Building, Room 540E, Baltimore, MD, 21201, USA,
| | | | | | | | | |
Collapse
|
50
|
Li X, Liu C, Li P, Li S, Zhao Z, Chen Y, Huo B, Zhang D. Connexin 43 is a potential regulator in fluid shear stress-induced signal transduction in osteocytes. J Orthop Res 2013; 31:1959-65. [PMID: 23878018 DOI: 10.1002/jor.22448] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 06/26/2013] [Indexed: 02/05/2023]
Abstract
Connexin 43 (Cx43), a gap junctional protein, regulates osteocyte viability, and modulates mechanical stimulation-induced bone remodeling. However, the underlying mechanisms of its action remain unclear. In the current study, osteocyte-like MLO-Y4 cells were exposed to fluid shear stress (FSS) of 16 (physiological) or 30 (high) dyne/cm(2) for the indicated time points. Cx43 gene (Gja1) was silenced using siRNA or the protein was blocked chemically. The signaling molecules related to osteocyte apoptosis, osteogenesis, or osteoclastogenesis were detected at mRNA or protein levels. The results showed that physiological FSS significantly upregulated Cx43, which further inhibited apoptosis pathways (e.g., caspase-3) and osteoclastogenesis signaling (e.g., RANKL), but activated osteogenesis signaling (Sost/sclerostin). Suppressing Cx43 gene (Gja1) by siRNA or chemically blocking gap junction communication enhanced caspase-3, RANKL, and Sost/sclerostin, which could be restored with physiological FSS over 8 h. In addition, high FSS decreased Cx43 expression and adversely affected signaling molecules compared with physiological FSS. The findings indicate the involvement of Cx43 in mechanotransduction of FSS and in the modulation of mechanical loading-related apoptosis, osteogenesis, and osteoclastogenesis of osteocytes. This may provide a cellular and molecular basis for interpreting the biomechanical mechanism of bone absorption and remodeling.
Collapse
Affiliation(s)
- Xiaoting Li
- State Key Laboratory of Oral Disease and Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan Province, PR China; Department of Stomatology, Chengdu Women's & Children's Central Hospital, Chengdu, 610041, Sichuan Province, PR China
| | | | | | | | | | | | | | | |
Collapse
|