1
|
Bonetto V, Pagano CA, Sabbatini M, Magnelli V, Donadelli M, Marengo E, Masini MA. Microgravity as a Tool to Investigate Cancer Induction in Pleura Mesothelial Cells. Curr Issues Mol Biol 2024; 46:10896-10912. [PMID: 39451527 PMCID: PMC11506709 DOI: 10.3390/cimb46100647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
The present work shows that the exposure of mesothelial cells to simulated microgravity changes their cytoskeleton and adhesion proteins, leading to a cell switch from normal towards tumoral cells. Immunohistochemical and molecular data were obtained from both MeT-5A exposed to simulated microgravity and BR95 mesothelioma cell lines. Simulated microgravity was found to affect the expression of actin, vinculin, and connexin-43, altering their quantitative and spatial distribution pattern inside the cell. The analysis of the tumoral markers p27, CD44, Fibulin-3, and NANOG and the expression of genes related to cancer transformation such as NANOG, CDH-1, and Zeb-1 showed that the simulated microgravity environment led to expression patterns in MeT-5A cells similar to those observed in BR95 cells. The alteration in both quantitative expression and structural organization of the cytoskeleton and adhesion/communication proteins can thus be considered a pivotal mechanism involved in the cellular shift towards tumoral progression.
Collapse
Affiliation(s)
- Valentina Bonetto
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (V.B.); (C.A.P.); (V.M.); (E.M.); (M.A.M.)
| | - Corinna Anais Pagano
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (V.B.); (C.A.P.); (V.M.); (E.M.); (M.A.M.)
| | - Maurizio Sabbatini
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (V.B.); (C.A.P.); (V.M.); (E.M.); (M.A.M.)
| | - Valeria Magnelli
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (V.B.); (C.A.P.); (V.M.); (E.M.); (M.A.M.)
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences (DNBM), University of Verona, 37124 Verona, Italy;
| | - Emilio Marengo
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (V.B.); (C.A.P.); (V.M.); (E.M.); (M.A.M.)
| | - Maria Angela Masini
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (V.B.); (C.A.P.); (V.M.); (E.M.); (M.A.M.)
| |
Collapse
|
2
|
Totland MZ, Knudsen LM, Rasmussen NL, Omori Y, Sørensen V, Elster VCW, Stenersen JM, Larsen M, Jensen CL, Zickfeldt Lade AA, Bruusgaard E, Basing S, Kryeziu K, Brech A, Aasen T, Lothe RA, Leithe E. The E3 ubiquitin ligase ITCH negatively regulates intercellular communication via gap junctions by targeting connexin43 for lysosomal degradation. Cell Mol Life Sci 2024; 81:171. [PMID: 38597989 PMCID: PMC11006747 DOI: 10.1007/s00018-024-05165-8] [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: 01/10/2023] [Revised: 01/27/2024] [Accepted: 02/05/2024] [Indexed: 04/11/2024]
Abstract
Intercellular communication via gap junctions has a fundamental role in regulating cell growth and tissue homeostasis, and its dysregulation may be involved in cancer development and radio- and chemotherapy resistance. Connexin43 (Cx43) is the most ubiquitously expressed gap junction channel protein in human tissues. Emerging evidence indicates that dysregulation of the sorting of Cx43 to lysosomes is important in mediating the loss of Cx43-based gap junctions in cancer cells. However, the molecular basis underlying this process is currently poorly understood. Here, we identified the E3 ubiquitin ligase ITCH as a novel regulator of intercellular communication via gap junctions. We demonstrate that ITCH promotes loss of gap junctions in cervical cancer cells, which is associated with increased degradation of Cx43 in lysosomes. The data further indicate that ITCH interacts with and regulates Cx43 ubiquitination and that the ITCH-induced loss of Cx43-based gap junctions requires its catalytic HECT (homologous to E6-AP C-terminus) domain. The data also suggest that the ability of ITCH to efficiently promote loss of Cx43-based gap junctions and degradation of Cx43 depends on a functional PY (PPXY) motif in the C-terminal tail of Cx43. Together, these data provide new insights into the molecular basis underlying the degradation of Cx43 and have implications for the understanding of how intercellular communication via gap junctions is lost during cancer development.
Collapse
Affiliation(s)
- Max Zachrisson Totland
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, NO-0424, Norway
| | - Lars Mørland Knudsen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, NO-0424, Norway
| | - Nikoline Lander Rasmussen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, NO-0424, Norway
- Centre for Molecular Medicine Norway, Faculty of Medicine, Oslo, Norway
| | - Yasufumi Omori
- Department of Molecular and Tumour Pathology, Akita University Graduate School of Medicine, Akita, 010-8543, Japan
| | - Vigdis Sørensen
- Department of Core Facilities, Institute for Cancer Research, Oslo University Hospital, Oslo, NO-0424, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, 0379, Norway
| | - Vilde C Wivestad Elster
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, NO-0424, Norway
| | - Jakob Mørkved Stenersen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, NO-0424, Norway
| | - Mathias Larsen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, NO-0424, Norway
| | - Caroline Lunder Jensen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, NO-0424, Norway
| | - Anna A Zickfeldt Lade
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, NO-0424, Norway
| | - Emilie Bruusgaard
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, NO-0424, Norway
| | - Sebastian Basing
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, NO-0424, Norway
| | - Kushtrim Kryeziu
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, NO-0424, Norway
| | - Andreas Brech
- Department of Core Facilities, Institute for Cancer Research, Oslo University Hospital, Oslo, NO-0424, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, 0379, Norway
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, Oslo, 0316, Norway
| | - Trond Aasen
- Patologia Molecular Translacional, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, Barcelona, 08035, Spain
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, NO-0424, Norway
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, Oslo, 0316, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, 0317, Norway
| | - Edward Leithe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, NO-0424, Norway
| |
Collapse
|
3
|
Totland MZ, Omori Y, Sørensen V, Kryeziu K, Aasen T, Brech A, Leithe E. Endocytic trafficking of connexins in cancer pathogenesis. Biochim Biophys Acta Mol Basis Dis 2023:166812. [PMID: 37454772 DOI: 10.1016/j.bbadis.2023.166812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/26/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Gap junctions are specialized regions of the plasma membrane containing clusters of channels that provide for the diffusion of ions and small molecules between adjacent cells. A fundamental role of gap junctions is to coordinate the functions of cells in tissues. Cancer pathogenesis is usually associated with loss of intercellular communication mediated by gap junctions, which may affect tumor growth and the response to radio- and chemotherapy. Gap junction channels consist of integral membrane proteins termed connexins. In addition to their canonical roles in cell-cell communication, connexins modulate a range of signal transduction pathways via interactions with proteins such as β-catenin, c-Src, and PTEN. Consequently, connexins can regulate cellular processes such as cell growth, migration, and differentiation through both channel-dependent and independent mechanisms. Gap junctions are dynamic plasma membrane entities, and by modulating the rate at which connexins undergo endocytosis and sorting to lysosomes for degradation, cells rapidly adjust the level of gap junctions in response to alterations in the intracellular or extracellular milieu. Current experimental evidence indicates that aberrant trafficking of connexins in the endocytic system is intrinsically involved in mediating the loss of gap junctions during carcinogenesis. This review highlights the role played by the endocytic system in controlling connexin degradation, and consequently gap junction levels, and discusses how dysregulation of these processes contributes to the loss of gap junctions during cancer development. We also discuss the therapeutic implications of aberrant endocytic trafficking of connexins in cancer cells.
Collapse
Affiliation(s)
| | - Yasufumi Omori
- Department of Molecular and Tumour Pathology, Akita University Graduate School of Medicine, Akita, Japan
| | | | | | - Trond Aasen
- Patologia Molecular Translacional, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron, Barcelona, Spain
| | - Andreas Brech
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway; Section for Physiology and Cell Biology, Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | | |
Collapse
|
4
|
McCutcheon S, Spray DC. Glioblastoma-Astrocyte Connexin 43 Gap Junctions Promote Tumor Invasion. Mol Cancer Res 2022; 20:319-331. [PMID: 34654721 PMCID: PMC8816813 DOI: 10.1158/1541-7786.mcr-21-0199] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 09/07/2021] [Accepted: 10/11/2021] [Indexed: 11/16/2022]
Abstract
Glioblastoma multiforme (GBM), classified as World Health Organization grade IV astrocytoma, is the deadliest adult cancer of the central nervous system. An important contributing factor to poor survival rates in GBM is extensive invasion, which decreases the efficacy of resection and subsequent adjuvant therapies. These treatments could be markedly improved with increased resolution of the genetic and molecular initiators and effectors of invasion. Connexin 43 (Cx43) is the principal astrocytic gap junction (GJ) protein. Despite the heterogeneity of GBM, a subpopulation of cells in almost all GBM tumors express Cx43. Functional GJs between GBM cells and astrocytes at the tumor edge are of critical interest for understanding invasion. In this study, we find that both in vitro and in ex vivo slice cultures, GBM is substantially less invasive when placed in a Cx43-deficient astrocyte environment. Furthermore, when Cx43 is deleted in GBM, the invasive phenotype is recovered. These data strongly suggest that there are opposing roles for Cx43 in GBM migration. We find that Cx43 is localized to the tumor edge in our ex vivo model, suggesting that GBM-astrocyte GJ communication at the tumor border is a driving force for invasion. Finally, we find that by a Cx43-dependent mechanism, but likely not direct channel-mediated diffusion, miRNAs associated with cell-matrix adhesion are transferred from GBM to astrocytes and miR-19b promotes invasion, revealing a role for post-transcriptional manipulation of astrocytes in fostering an invasion-permissive peritumoral niche. IMPLICATIONS: Cx43-mediated communication, specifically miRNA transfer, profoundly impacts glioblastoma invasion and may enable further therapeutic insight.
Collapse
Affiliation(s)
- Sean McCutcheon
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York.
| | - David C Spray
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
- Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
5
|
Synergistic inhibitory effect of resveratrol and TK/GCV therapy on melanoma cells. J Cancer Res Clin Oncol 2020; 146:1489-1499. [PMID: 32246216 DOI: 10.1007/s00432-020-03203-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/26/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE To investigate the synergistic effect of resveratrol on the bystander effect of TK/GCV suicide gene system in melanoma cells. METHODS The effect of resveratrol on the growth of B16 cells and the synergistic effect of resveratrol with or without GCV were detected by MTT assay and high content screening assay. The effect of resveratrol on GJIC function was detected by flow cytometry combined with fluorescence tracer and fluorescence microscope, and the expression of gap junction protein was detected by western blotting. Synergistic killing effect of resveratrol plus TK/GCV was tested in vivo using transplanted melanoma model. RESULTS In vitro, resveratrol can enhanced GJ function and upregulated Cx32 and Cx43 protein expression in B16 cells. Resveratrol synergized with GCV to kill mixed B16 melanoma cells (20% TK+ cells and 80% TK- cells) and to improve apoptosis rate of TK- cells (the bystander effect of TK system), and the synergistic action was reversed by the GJ inhibitor AGA. In vivo, when B16 cells were mixed with 30% TK+ B16 cells, significantly reduced tumor weight and volume were observed after combinational treatment with resveratrol plus GCV as compared with GCV or resveratrol treatment alone. CONCLUSIONS Resveratrol could synergistically enhance the killing effect of TK/GCV suicide gene system in melanoma B16 cells and transplanted melanoma. It might be a promising adjuvant of TK/GCV therapy.
Collapse
|
6
|
Yuan D, Li X, Luo C, Li X, Cheng N, Ji H, Qiu R, Luo G, Chen C, Hei Z. Inhibition of gap junction composed of Cx43 prevents against acute kidney injury following liver transplantation. Cell Death Dis 2019; 10:767. [PMID: 31601792 PMCID: PMC6787008 DOI: 10.1038/s41419-019-1998-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 08/12/2019] [Accepted: 09/23/2019] [Indexed: 12/19/2022]
Abstract
Postoperative acute kidney injury (AKI) is a severe complication after liver transplantation (LT). Its deterioration and magnification lead to the increase in mortality. Connexin43 (Cx43) mediates direct transmission of intracellular signals between neighboring cells, always considered to be the potent biological basis of organ damage deterioration and magnification. Thus, we explored the effects of Cx43 on AKI following LT and its related possible mechanism. In this study, alternations of Cx43 expression were observed in 82 patients, receiving the first-time orthotopic LT. We built autologous orthotopic liver transplantation (AOLT) models with Sprague–Dawley (SD) rats in vivo, and hypoxia-reoxygenation (H/R) or lipopolysaccharide (LPS) pretreatment models with kidney tubular epithelial cells (NRK-52E) in vitro, both of which were the most important independent risk factors of AKI following LT. Then, different methods were used to alter the function of Cx43 channels to determine its protective effects on AKI. The results indicated that patients with AKI suffering from longer time of tracheal intubation or intensive care unit stay, importantly, had significantly lower survival rate at postoperative 30 days and 3 years. In rat AOLT models, as Cx43 was inhibited with heptanol, postoperative AKI was attenuated significantly. In vitro experiments, downregulation of Cx43 with selective inhibitors, or siRNA protected against post-hypoxic NRK-52E cell injuries caused by H/R and/or LPS, while upregulation of Cx43 exacerbated the above-mentioned cell injuries. Of note, alternation of Cx43 function regulated the content of reactive oxygen species (ROS), which not only mediated oxidative stress and inflammation reactions effectively, but also regulated necroptosis. Therefore, we concluded that Cx43 inhibition protected against AKI following LT through attenuating ROS transmission between the neighboring cells. ROS alternation depressed oxidative stress and inflammation reaction, which ultimately reduced necroptosis. This might offer new insights for targeted intervention for organ protection in LT, or even in other major surgeries.
Collapse
Affiliation(s)
- Dongdong Yuan
- Department of Anesthesiology, the third affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, PR China.
| | - Xiaoyun Li
- Department of Anesthesiology, the third affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, PR China
| | - Chenfang Luo
- Department of Anesthesiology, the third affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, PR China
| | - Xianlong Li
- Department of Anesthesiology, the third affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, PR China
| | - Nan Cheng
- Department of Anesthesiology, the third affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, PR China
| | - Haocong Ji
- Department of Anesthesiology, Huizhou first People's Hospital, No. 20, San Xin Nan Road, Jiangbei, Huizhou, PR China
| | - Rongzong Qiu
- Department of Anesthesiology, Huizhou first People's Hospital, No. 20, San Xin Nan Road, Jiangbei, Huizhou, PR China
| | - Gangjian Luo
- Department of Anesthesiology, the third affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, PR China
| | - Chaojin Chen
- Department of Anesthesiology, the third affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, PR China.
| | - Ziqing Hei
- Department of Anesthesiology, the third affiliated Hospital of Sun Yat-sen University, Tianhe Road, Guangzhou, PR China.
| |
Collapse
|
7
|
Cx43 Inhibition Attenuates Sepsis-Induced Intestinal Injury via Downregulating ROS Transfer and the Activation of the JNK1/Sirt1/FoxO3a Signaling Pathway. Mediators Inflamm 2019; 2019:7854389. [PMID: 30948926 PMCID: PMC6425293 DOI: 10.1155/2019/7854389] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/05/2018] [Indexed: 12/17/2022] Open
Abstract
Intestinal injury has long been considered to play a crucial role in the pathophysiology of sepsis and has even been characterized as the “motor” of it. Thus, we explored the effects of connexin43 (Cx43) on sepsis-induced intestinal injury in order to provide potential therapeutic strategies. Rat cecal ligation and puncture (CLP) models in vivo and cell models (IEC-6 cells) pretreated with LPS in vitro were used in the current study. Firstly, different methods, such as Cx43 inhibitors (18-α-GA and oleamide) or siRNA targeting Cx43 and N-acetyl cysteine (NAC) (a kind of ROS scavenger), were used to observe the effects of Cx43 channels mediating ROS transfer on intestinal injury. Secondly, the influence of ROS content on the activity of the JNK1/Sirt1/FoxO3a signaling pathway was explored through the application of NAC, sp600125 (a JNK1 inhibitor), and nicotinamide (a Sirt1 inhibitor). Finally, luciferase assays and ChIP were used to determine the direct regulation of FoxO3a on proapoptotic proteins, Bim and Puma. The results showed that sepsis-induced intestinal injury presented a dynamic change, coincident with the alternation of Cx43 expression. The inhibition of Cx43 attenuated CLP-induced intestinal injury in vivo and LPS-induced IEC-6 injury in vitro. The changes of Cx43 channel function regulated ROS transfer between the neighboring cells, which mediated the activation of the JNK1/Sirt1/FoxO3a signaling pathway. FoxO3a directly affected its downstream target genes, Bim and Puma, which are responsible for cell or tissue apoptosis. In summary, our results suggest that Cx43 inhibition suppresses ROS transfer and inactivates the JNK1/Sirt1/FoxO3a signaling pathway to protect against sepsis-induced intestinal injury.
Collapse
|
8
|
Polyamines preserve connexin 43-mediated gap junctional communication during intracellular hypercalcemia and acidosis. Neuroreport 2018; 28:208-213. [PMID: 28134630 DOI: 10.1097/wnr.0000000000000746] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Changes in the regulation, formation, and gating of connexin-based gap junction channels occur in various disorders. It has been shown that H and Ca are involved in the regulation of gap junctional communication. Ischemia-induced intracellular acidification and Ca overload lead to closure of gap junctions and inhibit an exchange by ions and small molecules throughout the network of cells in the heart, brain, and other tissues. In this study, we examined the role of the polyamines in the regulation of connexin 43 (Cx43)-based gap junction channels under elevated intracellular concentrations of hydrogen ([H]i) and calcium ([Ca]i) ions. Experiments, conducted in Novikoff and A172 human glioblastoma cells, which endogenously express Cx43, showed that polyamines prevent downregulation of Cx43-mediated gap junctional communication caused by elevated [Ca]i and [H]i, accompanying ischemic and other pathological conditions. siRNA knockdown of Cx43 significantly reduces gap junctional communication, indicating that Cx43 gap junctions are the targets for spermine regulation.
Collapse
|
9
|
Wu M, Moh MC, Schwarz H. HepaCAM associates with connexin 43 and enhances its localization in cellular junctions. Sci Rep 2016; 6:36218. [PMID: 27819278 PMCID: PMC5098153 DOI: 10.1038/srep36218] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/12/2016] [Indexed: 12/30/2022] Open
Abstract
HepaCAM (GlialCAM) is frequently deleted in carcinomas, and reintroduction of hepaCAM into transformed cell lines reduces cellular growth and induces senescence. Mutations in HEPACAM give rise to the neurodegenerative disease megalencephalic leukoencephalopathy with subcortical cysts (MLC) since mutated hepaCAM prevents shuttling of MLC1 protein to astrocytic junctions in the plasma membrane. Here we identify that hepaCAM associates with connexin 43, a main component of gap junctions, and enhances connexin 43 localization to the plasma membrane at cellular junctions. HepaCAM also increases the levels of connexin 43, not by enhancing its transcription but by stabilizing connexin 43 protein. In the absence of hepaCAM, connexin 43 undergoes a faster degradation via the lysosomal pathway while proteasomal degradation seems not to be involved. Mutations in hepaCAM that cause MLC, or neutralization of hepaCAM by antibodies disrupt its association with connexin 43 at cellular junctions. By discovering the requirement of hepaCAM for localizing connexin 43, a well-established tumor suppressor, to cellular junctions and stabilizing it there, this study suggests a mechanism by which deletion of hepaCAM may support tumor progression.
Collapse
Affiliation(s)
- Meihui Wu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117456
| | - Mei Chung Moh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117456
| | - Herbert Schwarz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597.,Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117456
| |
Collapse
|
10
|
Bell CL, Murray SA. Adrenocortical Gap Junctions and Their Functions. Front Endocrinol (Lausanne) 2016; 7:82. [PMID: 27445985 PMCID: PMC4925680 DOI: 10.3389/fendo.2016.00082] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/20/2016] [Indexed: 12/21/2022] Open
Abstract
Adrenal cortical steroidogenesis and proliferation are thought to be modulated by gap junction-mediated direct cell-cell communication of regulatory molecules between cells. Such communication is regulated by the number of gap junction channels between contacting cells, the rate at which information flows between these channels, and the rate of channel turnover. Knowledge of the factors regulating gap junction-mediated communication and the turnover process are critical to an understanding of adrenal cortical cell functions, including development, hormonal response to adrenocorticotropin, and neoplastic dedifferentiation. Here, we review what is known about gap junctions in the adrenal gland, with particular attention to their role in adrenocortical cell steroidogenesis and proliferation. Information and insight gained from electrophysiological, molecular biological, and imaging (immunocytochemical, freeze fracture, transmission electron microscopic, and live cell) techniques will be provided.
Collapse
Affiliation(s)
- Cheryl L. Bell
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sandra A. Murray
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- *Correspondence: Sandra A. Murray,
| |
Collapse
|
11
|
Suzhi Z, Liang T, Yuexia P, Lucy L, Xiaoting H, Yuan Z, Qin W. Gap Junctions Enhance the Antiproliferative Effect of MicroRNA-124-3p in Glioblastoma Cells. J Cell Physiol 2015; 230:2476-88. [DOI: 10.1002/jcp.24982] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 03/03/2015] [Indexed: 12/31/2022]
Affiliation(s)
- Zhang Suzhi
- Department of Pharmacology; Zhongshan School of Medicine; Sun Yat-Sen University; Guangzhou P.R. China
| | - Tao Liang
- Department of Pharmacology; Zhongshan School of Medicine; Sun Yat-Sen University; Guangzhou P.R. China
| | - Peng Yuexia
- Department of Pharmacology; Zhongshan School of Medicine; Sun Yat-Sen University; Guangzhou P.R. China
| | - Liu Lucy
- Department of Cell and Systems Biology; University of Toronto; Ontario Canada
| | - Hong Xiaoting
- Department of Pharmacology; Zhongshan School of Medicine; Sun Yat-Sen University; Guangzhou P.R. China
| | - Zhang Yuan
- Department of Pharmacology; Zhongshan School of Medicine; Sun Yat-Sen University; Guangzhou P.R. China
| | - Wang Qin
- Department of Pharmacology; Zhongshan School of Medicine; Sun Yat-Sen University; Guangzhou P.R. China
| |
Collapse
|
12
|
Forster T, Rausch V, Zhang Y, Isayev O, Heilmann K, Schoensiegel F, Liu L, Nessling M, Richter K, Labsch S, Nwaeburu CC, Mattern J, Gladkich J, Giese N, Werner J, Schemmer P, Gross W, Gebhard MM, Gerhauser C, Schaefer M, Herr I. Sulforaphane counteracts aggressiveness of pancreatic cancer driven by dysregulated Cx43-mediated gap junctional intercellular communication. Oncotarget 2015; 5:1621-34. [PMID: 24742583 PMCID: PMC4039235 DOI: 10.18632/oncotarget.1764] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The extreme aggressiveness of pancreatic ductal adenocarcinoma (PDA) has been
associated with blocked gap junctional intercellular communication (GJIC) and the
presence of cancer stem cells (CSCs). We examined whether disturbed GJIC is
responsible for a CSC phenotype in established and primary cancer cells and patient
tissue of PDA using interdisciplinary methods based in physiology, cell and molecular
biology, histology and epigenetics. Flux of fluorescent dyes and gemcitabine through
gap junctions (GJs) was intact in less aggressive cells but not in highly malignant
cells with morphological dysfunctional GJs. Among several connexins, only Cx43 was
expressed on the cell surface of less aggressive and GJIC-competent cells, whereas
Cx43 surface expression was absent in highly malignant, E-cadherin-negative and
GJIC-incompetent cells. The levels of total Cx43 protein and Cx43 phosphorylated at
Ser368 and Ser279/282 were high in normal tissue but low to absent in malignant
tissue. si-RNA-mediated inhibition of Cx43 expression in GJIC-competent cells
prevented GJIC and induced colony formation and the expression of stem cell-related
factors. The bioactive substance sulforaphane enhanced Cx43 and E-cadherin levels,
inhibited the CSC markers c-Met and CD133, improved the functional morphology of GJs
and enhanced GJIC. Sulforaphane altered the phosphorylation of several kinases and
their substrates and inhibition of GSK3, JNK and PKC prevented sulforaphane-induced
CX43 expression. The sulforaphane-mediated expression of Cx43 was not correlated with
enhanced Cx43 RNA expression, acetylated histone binding and Cx43 promoter
de-methylation, suggesting that posttranslational phosphorylation is the dominant
regulatory mechanism. Together, the absence of Cx43 prevents GJIC and enhances
aggressiveness, whereas sulforaphane counteracts this process, and our findings
highlight dietary co-treatment as a viable treatment option for PDA.
Collapse
Affiliation(s)
- Tobias Forster
- General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Li G, Liu X, Liu Z, Su Z. Interactions of connexin 43 and aquaporin-4 in the formation of glioma-induced brain edema. Mol Med Rep 2014; 11:1188-94. [PMID: 25373717 DOI: 10.3892/mmr.2014.2867] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 09/18/2014] [Indexed: 11/05/2022] Open
Abstract
Connexin 43 (Cx43) and aquaporin-4 (AQP4) have important roles in the formation of glioma-induced brain edema; however, the association between these two factors in the development of edema has remained to be elucidated. In the present study, immunofluorescence and western blot analysis revealed that in a rat model of intracranial C6 glioma, Cx43 expression levels were low to undetectable and AQP4 expression levels were low in glioma cells. Significantly higher Cx43 and AQP4 levels were detected in the tissue surrounding the glioma. To further investigate the potential interaction between Cx43 and AQP4, normal glial cells and C6 glioma cells were cultured in hypotonic medium. Reverse transcription quantitative polymerase chain reaction indicated that AQP4 and Cx43 mRNA expression levels increased as a function of time in normal glial cells and C6 glioma cells in a hypotonic environment. However, the increase observed in normal glial cells was significantly lower than that observed in C6 glioma cells. Furthermore, AQP4 expression levels changed prior to alterations in Cx43 expression. Following AQP4 silencing in C6 cells, the increase in Cx43 expression was significantly attenuated (P<0.05). In normal cells, Cx43 silencing did not influence AQP4 expression (P>0.05). Therefore, it was hypothesized that AQP4 and Cx43 had two distinct mechanisms underlying brain edema formation within and surrounding the glioma. Cx43 may be a downstream effector of AQP4. The elucidation of this pathway may aid in the development of drugs targeting the interaction between AQP4 and Cx43, providing novel therapeutic possibilities for glioma-induced brain edema.
Collapse
Affiliation(s)
- Gang Li
- Department of Neurosurgery, Tianjin 5th Center Hospital, Tianjin 300450, P.R. China
| | - Xiaozhi Liu
- Department of Neurosurgery, Tianjin 5th Center Hospital, Tianjin 300450, P.R. China
| | - Zhenlin Liu
- Department of Neurosurgery, Tianjin 5th Center Hospital, Tianjin 300450, P.R. China
| | - Zhiguo Su
- Department of Neurosurgery, Tianjin 5th Center Hospital, Tianjin 300450, P.R. China
| |
Collapse
|
14
|
Godlewski J, Krichevsky AM, Johnson MD, Chiocca EA, Bronisz A. Belonging to a network--microRNAs, extracellular vesicles, and the glioblastoma microenvironment. Neuro Oncol 2014; 17:652-62. [PMID: 25301812 DOI: 10.1093/neuonc/nou292] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 09/09/2014] [Indexed: 12/14/2022] Open
Abstract
The complexity of glioblastoma multiforme (GBM) and its distinct pathophysiology belong to a unique brain microenvironment and its cellular interactions. Despite extensive evidence of a role for microRNAs in GBM cells, little is known about microRNA-dependent communication between different cellular compartments of the microenvironment that may contribute to the tumor phenotype. While the majority of microRNAs are found intracellularly, a significant number of microRNAs have been observed outside of cells, often encapsulated in secreted extracellular vesicles (EVs). The function of these circulating/secreted microRNAs has not been explored in the context of the brain tumor microenvironment. Establishing how microRNAs are involved in the regulation of oncogenic signaling networks between tumor cells and stroma is likely to add a needed additional layer of complexity to the tumor network, consisting of intercellular communication. More importantly, microRNA/EV signaling may provide an additional therapeutic target for this deadly disease.
Collapse
Affiliation(s)
- Jakub Godlewski
- Department of Neurosurgery (J.G., M.D.J., E.A.C., A.B.); Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (A.M.K)
| | - Anna M Krichevsky
- Department of Neurosurgery (J.G., M.D.J., E.A.C., A.B.); Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (A.M.K)
| | - Mark D Johnson
- Department of Neurosurgery (J.G., M.D.J., E.A.C., A.B.); Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (A.M.K)
| | - E Antonio Chiocca
- Department of Neurosurgery (J.G., M.D.J., E.A.C., A.B.); Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (A.M.K)
| | - Agnieszka Bronisz
- Department of Neurosurgery (J.G., M.D.J., E.A.C., A.B.); Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (A.M.K)
| |
Collapse
|
15
|
Yulyana Y, Endaya BB, Ng WH, Guo CM, Hui KM, Lam PYP, Ho IAW. Carbenoxolone enhances TRAIL-induced apoptosis through the upregulation of death receptor 5 and inhibition of gap junction intercellular communication in human glioma. Stem Cells Dev 2013; 22:1870-82. [PMID: 23428290 DOI: 10.1089/scd.2012.0529] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been used extensively in cancer therapy. However, more than half of glioblastoma multiforme are insensitive to the apoptotic effect of TRAIL. Improvement in therapeutic modalities that enhances the efficacy of TRAIL in glioma is much sought after. In this study, we combined the tumor selectivity of TRAIL and tumor-homing properties of mesenchymal stem cells (MSC) with gap junction (GJ) inhibitory effect of carbenoxolone (CBX) to target orthotopic glioma. MSC were engineered to express TRAIL (MSC-TRAIL) by incorporating the secretable trimeric form of TRAIL into a Herpes Simplex Virus (HSV) type I amplicon vector. Our results showed that combined treatment of MSC-TRAIL and CBX enhanced glioma cell death, especially in three primary human glioma isolates, of which two of those are marginally sensitive to TRAIL. CBX enhanced TRAIL-induced apoptosis through upregulation of death receptor 5, blockade of GJ intercellular communication, and downregulation of connexin 43. Dual arm therapy using TRAIL and CBX prolonged the survival of treated mice by ~27% when compared with the controls in an intracranial glioma model. The enhanced efficacy of TRAIL in combination with CBX coupled with the minimal cytotoxic nature of CBX suggested a favorable clinical usage of this treatment regimen.
Collapse
Affiliation(s)
- Yulyana Yulyana
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division, Humphrey Oei Institute of Cancer Research, National Cancer Centre of Singapore, Singapore, Singapore
| | | | | | | | | | | | | |
Collapse
|
16
|
Ryu CH, Park KY, Kim SM, Jeong CH, Woo JS, Hou Y, Jeun SS. Valproic acid enhances anti-tumor effect of mesenchymal stem cell mediated HSV-TK gene therapy in intracranial glioma. Biochem Biophys Res Commun 2012; 421:585-90. [PMID: 22525671 DOI: 10.1016/j.bbrc.2012.04.050] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 04/10/2012] [Indexed: 11/15/2022]
Abstract
Suicide gene therapy of glioma based on herpes simplex virus type I thymidine kinase (HSV-TK) and prodrug ganciclovir (GCV) suffers from the lack of efficacy in clinical trials, which is mostly due to low transduction efficacy and absence of bystander effect in tumor cells. Recently, stem cells as cellular delivery vehicles of prodrug converting gene has emerged as a new treatment strategy for malignant glioma. In this study, we evaluated the anti-glioma effect of suicide gene therapy using human bone marrow mesenchymal stem cells expressing HSV-TK (MSCs-TK) combined with valproic acid (VPA), which can upregulate the gap junction proteins and may enhance the bystander effect of suicide gene therapy. Expression of HSV-TK in MSCs was confirmed by RT-PCR analysis and the sensitivity of MSCs-TK to GCV was assessed. A bystander effect was observed in co-cultures of MSCs-TK and U87 glioma cells by GCV in a dose-dependent manner. VPA induced the expression of the gap junction proteins connexin (Cx) 43 and 26 in glioma cell and thereby enhanced the bystander effect in co-culture experiment. The enhanced bystander effect was inhibited by the gap junction inhibitor 18-β-glycyrrhetinic acid (18-GA). Moreover, the combined treatment with VPA and MSCs-TK synergistically enhanced apoptosis in glioma cells by caspase activation. In vivo efficacy experiments showed that combination treatment of MSCs-TK and VPA significantly inhibited tumor growth and prolonged the survival of glioma-bearing mice compared with single-treatment groups. In addition, TUNEL staining also demonstrated a significant increase in the number of apoptotic cells in the combination treated group compared with single-treatment groups. Taken together, these results provide the rational for designing novel experimental protocols to increase bystander killing effect against intracranial gliomas using MSCs-TK and VPA.
Collapse
Affiliation(s)
- Chung Heon Ryu
- Department of Biomedical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
17
|
In vitro and in vivo double-enhanced suicide gene therapy mediated by generation 5 polyamidoamine dendrimers for PC-3 cell line. World J Surg Oncol 2012; 10:3. [PMID: 22226139 PMCID: PMC3283475 DOI: 10.1186/1477-7819-10-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2011] [Accepted: 01/08/2012] [Indexed: 11/27/2022] Open
Abstract
Background One of the most frequently used and efficient suicide gene therapies for prostate cancer is HSV-TK/GCV system, but its application has been limited due to lack of favorable gene vector and the reduction of "bystander effect". We investigated the effect of a novel combination of HSV-TK/GCV fused with Cx43 and gemcitabine using non-viral vector generation 5 polyamidoamine dendrimers (G5-PAMAM-D) on PC-3 cells. Methods RT-PCR and Western blot were used to detect TK and Cx43 expression. Cell viability and proliferation were measured by using MTT assay. Cell apoptosis was detected with double-staining of Annexin V-FITC and propidium iodide (PI) by flow cytometry. Nude mice models were established to evaluate the therapeutic effect in vivo. Results G5-PAMAM-D efficiently delivered recombinant plasmids into PC-3 cells and HSV-TK and Cx43 could be expressed successfully. With gemcitabine, G5-PAMAM-D mediated HSV-TK and Cx43 expression effectively inhibited prostate cancer PC-3 cell proliferation, leading to more cellular apoptosis and inhibiting PC-3 tumor growth in nude mice models. Conclusions This study illustrates that this new suicide gene system mediated by G5-PAMAM-D is effective in decreasing PC-3 cell proliferation and inducing cell apoptosis, and inhibiting tumor growth in vivo. In a word, our study could provide a potential approach for gene therapy of prostate cancer.
Collapse
|
18
|
Chen Y, Wang G, Kong D, Zhang Z, Yang K, Liu R, Zhao W, Xu Y. Double-targeted and double-enhanced suicide gene therapy mediated by generation 5 polyamidoamine dendrimers for prostate cancer. Mol Carcinog 2011; 52:237-46. [DOI: 10.1002/mc.21850] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 11/02/2011] [Accepted: 11/08/2011] [Indexed: 01/09/2023]
|
19
|
Abstract
Glioblastoma is a very aggressive astrocytic tumor and most patients have 1-year survival time after diagnosis. A promising therapeutic strategy is the local delivery of the herpes simplex virus thymidine kinase gene in the tumor bed followed by ganciclovir treatment. The presence of functional gap junctions is highly relevant for the success of suicide gene therapy. Connexins are expressed in practically all tissues and form gap junctions that allow intercellular communication. Connexin 43 (Cx43) is the major connexin member being expressed in astrocytes but its status in glioblastoma is not well defined. We have investigated by immunofluorescence the presence of Cx43 in 74 human glioblastoma samples; its expression was detected in 77% of the samples analyzed. We report here that glioblastoma is a heterogenous disease as regards Cx43 expression with presentations, in which Cx43 expression is unaltered, reduced or totally lost. A predominant Cx43 cytoplasmic localization was observed in four out of eight primary glioblastoma cultures that we have established. This aberrant localization reduced gap junctionnal intercellular communication by 50 to 75% as compared with primary cell cultures displaying gap junctional plaques. However, the bystander effect evaluated after lentiviral delivery of the herpes simplex virus thymidine kinase gene and ganciclovir treatment was detected in all Cx43-positive primary cell cultures, and it was independant of the Cx43 localization. These findings may have important clinical implications for the design of anticancer cytotoxic therapies that rely on the gap junction-mediated bystander effect for their success.
Collapse
|
20
|
Garcia-Rodríguez L, Pérez-Torras S, Carrió M, Cascante A, García-Ribas I, Mazo A, Fillat C. Connexin-26 is a key factor mediating gemcitabine bystander effect. Mol Cancer Ther 2011; 10:505-17. [PMID: 21388975 DOI: 10.1158/1535-7163.mct-10-0693] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Gemcitabine is a nucleoside analogue with anticancer activity. Inside the cell, it is sequentially phosphorylated to generate the active drug. Phosphorylated nucleoside analogues have been shown to traffic through gap junctions. We investigated the participation of gap junctional intercellular communication (GJIC) as a possible mechanism spreading gemcitabine cytotoxicity in pancreatic tumors. Immunohistochemical analysis of pancreatic cancer biopsies revealed increased connexin 26 (Cx26) content but loss of connexins 32 (Cx32) and 43 (Cx43) expression. Cx26 abundance in neoplastic areas was confirmed by Cx26 mRNA in situ hybridization. Heterogeneity on the expression levels and the localization of Cx26, Cx32, and Cx43 were identified in pancreatic cancer cells and found to be associated with the extent of GJIC, and correlated with gemcitabine bystander cytotoxic effect. The abundance of Cx26 at the contact points in tumoral regions prompted us to study the involvement of Cx26 in the GJIC of gemcitabine toxic metabolites and their influence on the antitumoral effects of gemcitabine. Knockdown of Cx26 led to decreased GJIC and reduced gemcitabine bystander killing whereas overexpression of Cx26 triggered increased GJIC and enhanced the gemcitabine cytotoxic bystander effect. Gemcitabine treatment of mice bearing tumors, with a high GJIC capacity, resulted in a significant delay in tumor progression. Interestingly, gemcitabine administration in mice bearing tumors that overexpress Cx26 triggered a dramatic tumor regression of 50% from the initial volume. This study shows that Cx26 participates in the gap junction-mediated bystander cytoxic effect of gemcitabine and provides evidence that upregulation of Cx26 improves gemcitabine anticancer efficacy.
Collapse
Affiliation(s)
- Laura Garcia-Rodríguez
- Programa Gens i Malaltia, Centre de Regulació Genòmica. Dr. Aiguader, 88. 08003-Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
21
|
Paíno T, Gangoso E, Medina JM, Tabernero A. Inhibition of ATP-sensitive potassium channels increases HSV-tk/GCV bystander effect in U373 human glioma cells by enhancing gap junctional intercellular communication. Neuropharmacology 2010; 59:480-91. [PMID: 20603136 DOI: 10.1016/j.neuropharm.2010.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Revised: 06/15/2010] [Accepted: 06/25/2010] [Indexed: 01/16/2023]
Abstract
It is well known that the efficiency of Herpes simplex virus thymidine kinase gene/ganciclovir (HSV-tk/GCV) therapy is improved by the bystander effect, which mainly relies on gap junctional intercellular communication (GJIC). Malignant gliomas communicate poorly through gap junctions, consequently, agents with the ability to increase GJIC are good candidates to improve the efficiency of this therapy. Since we previously showed that the inhibition of ATP-sensitive potassium (KATP) channels promoted by tolbutamide increased GJIC in rat C6 glioma cells, we have investigated whether tolbutamide could increase the bystander effect in HSV-tk/GCV therapy against human glioma cells. We found that tolbutamide increased GJIC in U373 human glioma cells, an effect that was due to the up-regulation of connexin43, a protein that forms gap junctions channels. More interestingly, our results show that tolbutamide increased the efficiency of HSV-tk/GCV in co-cultures containing U373 cells and U373 cells transfected with HSV-tk. This effect was impaired in the presence of carbenoxolone, an inhibitor of GJIC. Furthermore, tolbutamide did not enhance the bystander effect in connexin43-silenced co-cultures. Together our results reveal that the inhibition of KATP channels promoted by tolbutamide enhances the bystander effect in HSV-tk/GCV therapy by increasing connexin43-mediated gap junctional intercellular communication in U373 human glioma cells.
Collapse
Affiliation(s)
- Teresa Paíno
- Departamento de Bioquímica y Biología Molecular, Instituto de Neurociencias de Castilla y León, INCYL, Universidad de Salamanca, Spain
| | | | | | | |
Collapse
|
22
|
Cottin S, Ghani K, de Campos-Lima PO, Caruso M. Gemcitabine intercellular diffusion mediated by gap junctions: new implications for cancer therapy. Mol Cancer 2010; 9:141. [PMID: 20537146 PMCID: PMC2898703 DOI: 10.1186/1476-4598-9-141] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 06/10/2010] [Indexed: 02/06/2023] Open
Abstract
Background Solid tumors are often poorly vascularized, with cells that can be 100 μm away from blood vessels. These distant cells get less oxygen and nutrients and are exposed to lower doses of chemotherapeutic agents. As gap junctions allow the passage of small molecules between cells, we tested the possibility that the chemotherapeutic agent gemcitabine can diffuse through gap junctions in solid tumors. Results We first showed with a dye transfer assay that the glioblastoma and the osteosarcoma cells used in this study have functional gap junctions. These cells were genetically engineered to express the herpes simplex virus thymidine kinase (TK), and induced a "bystander effect" as demonstrated by the killing of TK-negative cells in presence of the nucleoside analogue ganciclovir (GCV). The ability of gemcitabine to induce a similar bystander effect was then tested by mixing cells treated with 3 μM gemcitabine for 24 hours with untreated cells at different ratios. In all cell lines tested, bystander cells were killed with ratios containing as low as 5% treated cells, and this toxic effect was reduced in presence of α-glycyrrhetinic acid (AGA), a specific gap junction inhibitor. We also showed that a 2- or a 24-hour gemcitabine treatment was more efficient to inhibit the growth of spheroids with functional gap junctions as compared to the same treatment made in presence of AGA. Finally, after a 24-hour gemcitabine treatment, the cell viability in spheroids was reduced by 92% as opposed to 51% in presence of AGA. Conclusion These results indicate that gemcitabine-mediated toxicity can diffuse through gap junctions, and they suggest that gemcitabine treatment could be more efficient for treating solid tumors that display gap junctions. The presence of these cellular channels could be used to predict the responsiveness to this nucleoside analogue therapy.
Collapse
Affiliation(s)
- Sylvine Cottin
- Centre de Recherche en Cancérologie de l'Université Laval, L'Hôtel-Dieu de Québec, Centre Hospitalier Universitaire de Québec, Qc G1R 2J6, Canada
| | | | | | | |
Collapse
|
23
|
Zhang B, Feng X, Wang J, Xu X, Liu H, Lin N. Adenovirus-mediated delivery of bFGF small interfering RNA increases levels of connexin 43 in the glioma cell line, U251. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2010; 29:3. [PMID: 20074329 PMCID: PMC2830951 DOI: 10.1186/1756-9966-29-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Accepted: 01/14/2010] [Indexed: 11/10/2022]
Abstract
BACKGROUND bFGF is an important growth factor for glioma cell proliferation and invasion, while connexin 43 is implicated in the suppression of glioma growth. Correspondingly, gliomas have been shown to have reduced, or compromised, connexin 43 expression. METHODS In this study, a bFGF-targeted siRNA was delivered to the glioma cell line, U251, using adenovirus (Ad-bFGF-siRNA) and the expression of connexin 43 and its phosphorylation state were evaluated. U251 cells were infected with Ad-bFGF-siRNA (100, 50, or 25 MOI), and infection with adenovirus expressing green fluorescent protein (Ad-GFP) at 100 MOI served as a control. Western blotting and immunofluorescence were used to detect the expression levels, phosphorylation, and localization of connexin 43 in U251 cells infected, and not infected, with Ad-bFGF-siRNA. RESULTS Significantly higher levels of connexin 43 were detected in U251 cells infected with Ad-bFGF-siRNA at 100 and 50 MOI than in cells infected with Ad-GFP, and the same amount of connexin 43 was detected in Ad-GFP-infected and uninfected U251 cells. Connexin 43 phosphorylation did not differ between Ad-bFGF-siRNA-infected and uninfected U251 cells. However, the ratio of phosphorylated to unphosphorylated connexin 43 in Ad-bFGF-siRNA cells was lower, and connexin 43 was predominantly localized to the cytoplasm. Using a scrape loading dye transfer assay, more Lucifer Yellow was transferred to neighboring cells in the Ad-bFGF-siRNA treated group than in the control group. CONCLUSION To our knowledge, this is the first description of a role for connexin 43 in the inhibition of U251 growth using Ad-bFGF-siRNA.
Collapse
Affiliation(s)
- Biao Zhang
- Key Lab for Critical Care Medicine of the Ministry of Health, Affiliated Tianjin First Center Hospital, Tianjin Medical University, Tianjin, 300192, China
| | | | | | | | | | | |
Collapse
|