1
|
Paunikar S, Tamagnone L. Connexin-43 in Cancer: Above and Beyond Gap Junctions! Cancers (Basel) 2024; 16:4191. [PMID: 39766090 PMCID: PMC11674308 DOI: 10.3390/cancers16244191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Connexin-43 (Cx43) is the most characterized gap junction protein, primarily involved in the Gap Junctional Intercellular Communication (GJIC) between adjacent cells to facilitate molecule exchange and the formation of a signaling network. It is increasingly evident that the importance of Cx43 is not only limited to its GJIC function, but rather includes its role in connecting the intracellular and extracellular environment by forming membrane hemichannels, as well as its intracellular signaling function mediated by its C-terminal tail (Cx43-CT). Notably, Cx43 has been implicated in a variety of cancers, with earlier notions suggesting a tumor-suppressor function, whereas new studies shed light on its pro-tumorigenic role. Moreover, apart from GJIC-based activities, the relevance of the non-canonical functions of Cx43 in tumor progression is being actively studied. This review provides an analysis of the current research on the pro-tumorigenic roles of Cx43, with a focus on Cx43-CT interactions and the function of hemichannels in cancer progression. A better understanding of the multifaceted functions of Cx43 in cancer biology could foster its recognition as a pivotal target for the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Shishir Paunikar
- School of Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Luca Tamagnone
- School of Medicine, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Fondazione Policlinico Universitario “A.Gemelli” IRCCS, 00168 Rome, Italy
| |
Collapse
|
2
|
Wilczyński B, Dąbrowska A, Kulbacka J, Baczyńska D. Chemoresistance and the tumor microenvironment: the critical role of cell-cell communication. Cell Commun Signal 2024; 22:486. [PMID: 39390572 PMCID: PMC11468187 DOI: 10.1186/s12964-024-01857-7] [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: 07/08/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024] Open
Abstract
Resistance of cancer cells to anticancer drugs remains a major challenge in modern medicine. Understanding the mechanisms behind the development of chemoresistance is key to developing appropriate therapies to counteract it. Nowadays, with advances in technology, we are paying more and more attention to the role of the tumor microenvironment (TME) and intercellular interactions in this process. We also know that important elements of the TME are not only the tumor cells themselves but also other cell types, such as mesenchymal stem cells, cancer-associated fibroblasts, stromal cells, and macrophages. TME elements can communicate with each other indirectly (via cytokines, chemokines, growth factors, and extracellular vesicles [EVs]) and directly (via gap junctions, ligand-receptor pairs, cell adhesion, and tunnel nanotubes). This communication appears to be critical for the development of chemoresistance. EVs seem to be particularly interesting structures in this regard. Within these structures, lipids, proteins, and nucleic acids can be transported, acting as signaling molecules that interact with numerous biochemical pathways, thereby contributing to chemoresistance. Moreover, drug efflux pumps, which are responsible for removing drugs from cancer cells, can also be transported via EVs.
Collapse
Affiliation(s)
- Bartosz Wilczyński
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, Wroclaw, 50-367, Poland
| | - Alicja Dąbrowska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, Wroclaw, 50-367, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, Wroclaw, 50-556, Poland.
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, Santariškių g. 5, Vilnius, LT-08406, Lithuania.
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, Wroclaw, 50-556, Poland
| |
Collapse
|
3
|
Alimohammadi M, Rahimzadeh P, Khorrami R, Bonyadi M, Daneshi S, Nabavi N, Raesi R, Farani MR, Dehkhoda F, Taheriazam A, Hashemi M. A comprehensive review of the PTEN/PI3K/Akt axis in multiple myeloma: From molecular interactions to potential therapeutic targets. Pathol Res Pract 2024; 260:155401. [PMID: 38936094 DOI: 10.1016/j.prp.2024.155401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/02/2024] [Accepted: 06/09/2024] [Indexed: 06/29/2024]
Abstract
Phosphatase and tensin homolog (PTEN), phosphatidylinositol 3-kinase (PI3K), and protein kinase B (Akt) signaling pathways contribute to the development of several cancers, including multiple myeloma (MM). PTEN is a tumor suppressor that influences the PI3K/Akt/mTOR pathway, which in turn impacts vital cellular processes like growth, survival, and treatment resistance. The current study aims to present the role of PTEN and PI3K/Akt/mTOR signaling in the development of MM and its response to treatment. In addition, the molecular interactions in MM that underpin the PI3K/Akt/mTOR pathway and address potential implications for the development of successful treatment plans are also discussed in detail. We investigate their relationship to both upstream and downstream regulators, highlighting new developments in combined therapies that target the PTEN/PI3K/Akt axis to overcome drug resistance, including the use of PI3K and mitogen-activated protein kinase (MAPK) inhibitors. We also emphasize that PTEN/PI3K/Akt pathway elements may be used in MM diagnosis, prognosis, and therapeutic targets.
Collapse
Affiliation(s)
- Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran.
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Islamic Republic of Iran
| | - Mojtaba Bonyadi
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Islamic Republic of Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Islamic Republic of Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Rasoul Raesi
- Department of Health Services Management, Mashhad University of Medical Sciences, Mashhad, Islamic Republic of Iran; Department of Nursing, Torbat Jam Faculty of Medical Sciences, Torbat Jam, Iran
| | - Marzieh Ramezani Farani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Incheon 22212, Republic of Korea
| | - Farshid Dehkhoda
- Department of Orthopedics, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Islamic Republic of Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Islamic Republic of Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Islamic Republic of Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Islamic Republic of Iran.
| |
Collapse
|
4
|
Wei X, Zhang Y, Wang Z, He Y, Ju S, Fu J. Bone marrow adipocytes is a new player in supporting myeloma cells proliferation and survival in myeloma microenvironment. Transl Oncol 2024; 40:101856. [PMID: 38134840 PMCID: PMC10776777 DOI: 10.1016/j.tranon.2023.101856] [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: 10/11/2023] [Revised: 11/09/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Multiple myeloma (MM) is a lethal B cell neoplasm characterized by clonal expansion of malignant plasma cells in the bone marrow and remains incurable due to disease relapse and drug resistance. Bone marrow adipocytes (BMAs) are emerging as playing active functions that can support myeloma cell growth and survival. The aim of this study is to investigate myeloma-mesenchymal stem cells (MSCs) interaction and the impact of such interactions on the pathogenesis of MM using in vitro co-culture assay. Here we provide evidence that MM cell up-regulated MSCs to express PPAR-γ and pushes MSCs differentiation toward adipocytes at the expense of osteoblasts in co-culture manner. The increased BMAs can effectively enhance MM cell to proliferation, migration, and chemoresistance via cell-cell contact and/or cytokines release regulated by PPAR-γ signal pathway. This effect was partially reversed in medium containing PPAR-γ antagonist G3335 and indicated that G3335 distorts the maturation of MSC-derived adipocytes and cytokines release by adipocytes through inhibition of PPAR-γ, a key transcriptional factor for the activation of adipogenesis, or cell to cell contact, or both. In meantime, we observed higher expression of adipocyte differentiation associated genes DLK1, DGAT1, FABP4, and FASN both in MSCs and MSC derived adipocytes, but the osteoblast differentiation-associated gene ALP was down regulated in MSCs. These finding mean that direct consequence of MM/MSC interaction that play a role in MM pathogenesis. Consistent with those in vitro results, our primary clinical observation also showed that bone marrow samples from MM patients had significantly higher bone adiposity in comparison with controls and the number of adipocytes decreased in those who were response to anti-MM therapy. Our finding suggested that BMAs may have an important contribution to MM progression, particularly in drugs resistant of MM cells, and plays an important contribution in MM bone disease and treatment failure, but more clinical studies are needed to confirm its role.
Collapse
Affiliation(s)
- Xiaoqian Wei
- Hematology Department, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Yangmin Zhang
- Hematology Department, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Ziyan Wang
- Hematology Department, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Yuanning He
- Hematology Department, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China
| | - Songguang Ju
- Institute of Biotechnology, Soochow University, Suzhou 215007, PR China
| | - Jinxiang Fu
- Hematology Department, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, PR China.
| |
Collapse
|
5
|
Staskiewicz A, Wong E, Tucker M, Farhin R, Park J, Saade R, Alkhazali T, Dang T, Wang X. Cytotoxic and Apoptotic Effects of Pinostilbene and Bortezomib Combination Treatment on Human Multiple Myeloma Cells. Int J Mol Sci 2023; 24:12590. [PMID: 37628771 PMCID: PMC10454535 DOI: 10.3390/ijms241612590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 08/01/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
Multiple myeloma (MM) is a cancer of plasma cells in the bone marrow characterized by bone lesions, hypercalcemia, anemia, and renal failure. Bortezomib (BTZ), a common treatment for MM, is a proteasome inhibitor that induces apoptosis in MM cells. However, high doses of BTZ can be very toxic, signifying a need for a synergistic drug combination to improve treatment efficacy. Resveratrol (RES), a phenolic compound found in grapes, has been shown to inhibit MM cell growth. We sought to identify a synergistic combination of BTZ with a RES derivative and analyze the effects on reducing viability and inducing apoptosis in human MM cells. BTZ as well as RES and its derivatives pinostilbene (PIN) and piceatannol (PIC) decreased MM cell viability in a dose- and time-dependent manner and increased expression of cleaved proapoptotic proteins poly(ADP-ribose) polymerase 1 (PARP1) and caspase-3 in a dose-dependent manner. The combination of 5 nM BTZ and 5 μM PIN was identified to have synergistic cytotoxic effects in MM RPMI 8226 cells. MM RPMI 8226 cells treated with this combination for 24 h showed increased cleaved PARP1 and caspase-3 expression and higher percentages of apoptotic cells versus cells treated with the individual compounds alone. The treatment also showed increased apoptosis induction in MM RPMI 8226 cells co-cultured with human bone marrow stromal HS-5 cells in a Transwell model used to mimic the bone marrow microenvironment. Expression of oxidative stress defense proteins (catalase, thioredoxin, and superoxide dismutase) in RPMI 8226 cells were reduced after 24 h treatment, and cytotoxic effects of the treatment were ameliorated by antioxidant N-acetylcysteine (NAC), suggesting the treatment impacts antioxidant levels in RPMI 8226 cells. Our results suggest that this combination of BTZ and PIN decreases MM cell viability synergistically by inducing apoptosis and oxidative stress in MM cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xinyu Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, Philadelphia College of Osteopathic Medicine–Georgia Campus, Suwanee, GA 30024, USA; (A.S.)
| |
Collapse
|
6
|
The Expression of Serum lncRNA MIR17HG in Patients with Multiple Myeloma and Its Clinical Significance. Eur J Cancer Care (Engl) 2023. [DOI: 10.1155/2023/1728909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Objective. Multiple myeloma (MM) represents a malignant tumor with abnormal proliferation of plasma cells. The current study sought to investigate the changes in serum lncRNA MIR17HG (long noncoding RNA miR-17-92a-1 cluster host gene) levels in MM patients and its values in assessing the accuracy of MM diagnosis and predicting diagnosis. Methods. First, 108MM patients and 85 healthy controls were enrolled as the study subjects. The serum levels of MIR17HG in all subjects were determined by RT-qPCR. MM patients were clinically staged according to the Durie-Salmon (DS) and international staging system (ISS), and the levels of serum MIR17HG were compared among patients at different stages. The correlation of serum MIR17H level with serum creatinine (Scr), lactate dehydrogenase (LDH), and albumin (ALB) was analyzed using the Pearson method. The accuracy of the serum MIR17HG level in identifying MM was evaluated using receiver operating characteristic curves. The progression-free survival (PFS) and overall survival (OS) curves of MM patients were plotted using the Kaplan–Meier method. Results. Serum MIR17HG levels were up-regulated in MM patients and elevated with the development of DS and ISS stages. The serum MIR17HG was positively correlated with Scr and LDH and negatively correlated with ALB in MM patients. Serum MIR17HG level >1.485 could evaluate the accuracy of identifying MM. The PFS and OS were significantly shortened in MM patients with elevated MIR17HG levels. Conclusion. Our findings collectively indicate that the serum MIR17HG can aid the evaluation of accurate MM identification, and a high serum MIR17HG level can predict poor prognosis of patients with MM.
Collapse
|
7
|
Melaccio A, Reale A, Saltarella I, Desantis V, Lamanuzzi A, Cicco S, Frassanito MA, Vacca A, Ria R. Pathways of Angiogenic and Inflammatory Cytokines in Multiple Myeloma: Role in Plasma Cell Clonal Expansion and Drug Resistance. J Clin Med 2022; 11:jcm11216491. [PMID: 36362718 PMCID: PMC9658666 DOI: 10.3390/jcm11216491] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/23/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy, and despite the introduction of innovative therapies, remains an incurable disease. Identifying early and minimally or non-invasive biomarkers for predicting clinical outcomes and therapeutic responses is an active field of investigation. Malignant plasma cells (PCs) reside in the bone marrow (BM) microenvironment (BMME) which comprises cells (e.g., tumour, immune, stromal cells), components of the extracellular matrix (ECM) and vesicular and non-vesicular (soluble) molecules, all factors that support PCs’ survival and proliferation. The interaction between PCs and BM stromal cells (BMSCs), a hallmark of MM progression, is based not only on intercellular interactions but also on autocrine and paracrine circuits mediated by soluble or vesicular components. In fact, PCs and BMSCs secrete various cytokines, including angiogenic cytokines, essential for the formation of specialized niches called “osteoblastic and vascular niches”, thus supporting neovascularization and bone disease, vital processes that modulate the pathophysiological PCs–BMME interactions, and ultimately promoting disease progression. Here, we aim to discuss the roles of cytokines and growth factors in pathogenetic pathways in MM and as prognostic and predictive biomarkers. We also discuss the potential of targeted drugs that simultaneously block PCs’ proliferation and survival, PCs–BMSCs interactions and BMSCs activity, which may represent the future goal of MM therapy.
Collapse
Affiliation(s)
- Assunta Melaccio
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: (A.M.); (R.R.); Tel.: +39-320-55-17-232 (A.M.)
| | - Antonia Reale
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University—Alfred Health, Melbourne 3004, Australia
| | - Ilaria Saltarella
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Vanessa Desantis
- Department of Biomedical Sciences and Human Oncology, Pharmacology Section, University of Bari Aldo Moro Medical School, 70124 Bari, Italy
| | - Aurelia Lamanuzzi
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
| | - Sebastiano Cicco
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
| | - Maria Antonia Frassanito
- General Pathology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70124 Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
| | - Roberto Ria
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine “G. Baccelli”, University of Bari Medical School, 70124 Bari, Italy
- Correspondence: (A.M.); (R.R.); Tel.: +39-320-55-17-232 (A.M.)
| |
Collapse
|
8
|
Goodarzi A, Valikhani M, Amiri F, Safari A. The mechanisms of mutual relationship between malignant hematologic cells and mesenchymal stem cells: Does it contradict the nursing role of mesenchymal stem cells? Cell Commun Signal 2022; 20:21. [PMID: 35236376 PMCID: PMC8889655 DOI: 10.1186/s12964-022-00822-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/18/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are known as the issue in biology because of some unpredictable characteristics in the different microenvironments especially in their bone marrow niche. MSCs are used in the regenerative medicine because of their unique potentials for trans-differentiation, immunomodulation, and paracrine capacity. But, their pathogenic and pro-survival effects in tumors/cancers including hematologic malignancies are indisputable. MSCs and/or their derivatives might be involved in tumor growth, metastasis and drug resistance in the leukemias. One of important relationship is MSCs and hematologic malignancy-derived cells which affects markedly the outcome of disease. The communication between these two cells may be contact-dependent and/or contact-independent. In this review, we studied the crosstalk between MSCs and malignant hematologic cells which results the final feedback either the progression or suppression of blood cell malignancy. Video abstract.
Collapse
Affiliation(s)
- Alireza Goodarzi
- Department of Medical Laboratory Sciences, School of Paramedicine, Hamadan University of Medical Sciences, Shahid Fahmideh Blvd., The Opposite Side of Mardom Park, Hamadan, 6517838741, Iran
| | - Mohsen Valikhani
- Hematology Department, School of Allied Medical Science, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Amiri
- Department of Medical Laboratory Sciences, School of Paramedicine, Hamadan University of Medical Sciences, Shahid Fahmideh Blvd., The Opposite Side of Mardom Park, Hamadan, 6517838741, Iran.
| | - Armita Safari
- Student Research Committee, Hamadan University of Medical Science, Hamadan, Iran
| |
Collapse
|
9
|
Wu L, Xia L, Chen X, Ruan M, Li L, Xia R. Long non-coding RNA LINC01003 suppresses the development of multiple myeloma by targeting miR-33a-5p/PIM1 axis. Leuk Res 2021; 106:106565. [PMID: 33865032 DOI: 10.1016/j.leukres.2021.106565] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Numerous long non-coding RNAs (lncRNAs) are reported to affect the progression of multiple myeloma (MM). This study is aimed to explore the role and downstream mechanism of lncRNA LINC01003 in MM. MATERIALS AND METHODS Xenograft tumor assay was used to assess the function of LINC01003 in MM in vivo. The mRNA expression levels of LINC01003, miR-33a-5p, and PIM1 were determined by quantitative real-time polymerase chain reaction. Cell viability was examined by MTT assay. Relative protein levels of apoptosis-related factors (Bcl-2 and Bax) and proviral integration site of the Moloney leukemia virus kinase 1 (PIM1) were detected via western blot. Adhesion-related proteins were measured by Enzyme-linked immunosorbent assay was used to determine the levels of adhesion-related proteins. Besides, the target relation among LINC01003, miR-33a-5p and PIM1 was tested via dual-luciferase reporter assay. RESULTS Low expression of LINC01003 was observed in MM cell lines and peripheral blood samples of MM patients. Both LINC01003 up-regulation and miR-33a-5p down-regulation repressed cell viability and adhesion, and promoted apoptosis of MM cells. Moreover, LINC01003 suppressed the growth of xenograft tumor in mice. We then identified miR-33a-5p as a downstream target of LINC01003, and confirmed that PIM1 was a direct target gene of miR-33a-5p. Both high expression of miR-33a-5p and low expression of PIM1 reversed the suppressive effects of LINC01003 overexpression on cell adhesion and viability, and the promoting effect on apoptosis in MM cells. CONCLUSION LINC01003 functioned as a sponge of miR-33a-5p to inhibit the development MM by regulating PIM1 expression.
Collapse
Affiliation(s)
- Linlin Wu
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei City, Anhui Province, 230032, China
| | - Liang Xia
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei City, Anhui Province, 230032, China
| | - Xiaowen Chen
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei City, Anhui Province, 230032, China
| | - Min Ruan
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei City, Anhui Province, 230032, China
| | - Lingling Li
- Department of Hematology of Anhui No.2 Provincial People's Hospital, No. 1868, Dangshan Road, Hefei City, Anhui Province, 230041, China
| | - Ruixiang Xia
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei City, Anhui Province, 230032, China.
| |
Collapse
|
10
|
Mulkearns-Hubert EE, Reizes O, Lathia JD. Connexins in Cancer: Jekyll or Hyde? Biomolecules 2020; 10:E1654. [PMID: 33321749 PMCID: PMC7764653 DOI: 10.3390/biom10121654] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 12/16/2022] Open
Abstract
The expression, localization, and function of connexins, the protein subunits that comprise gap junctions, are often altered in cancer. In addition to cell-cell coupling through gap junction channels, connexins also form hemichannels that allow communication between the cell and the extracellular space and perform non-junctional intracellular activities. Historically, connexins have been considered tumor suppressors; however, they can also serve tumor-promoting functions in some contexts. Here, we review the literature surrounding connexins in cancer cells in terms of specific connexin functions and propose that connexins function upstream of most, if not all, of the hallmarks of cancer. The development of advanced connexin targeting approaches remains an opportunity for the field to further interrogate the role of connexins in cancer phenotypes, particularly through the use of in vivo models. More specific modulators of connexin function will both help elucidate the functions of connexins in cancer and advance connexin-specific therapies in the clinic.
Collapse
Affiliation(s)
- Erin E. Mulkearns-Hubert
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (O.R.); (J.D.L.)
| | - Ofer Reizes
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (O.R.); (J.D.L.)
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College, Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
| | - Justin D. Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (O.R.); (J.D.L.)
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College, Medicine of Case Western Reserve University, Cleveland, OH 44195, USA
- Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, 44195, USA
| |
Collapse
|
11
|
Yang X, Huang H, Wang X, Liu H, Liu H, Lin Z. Knockdown of lncRNA SNHG16 suppresses multiple myeloma cell proliferation by sponging miR-342-3p. Cancer Cell Int 2020; 20:38. [PMID: 32025219 PMCID: PMC6998159 DOI: 10.1186/s12935-020-1118-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 01/20/2020] [Indexed: 12/12/2022] Open
Abstract
Background Aberrant expression of long non-coding RNAs (lncRNAs) is closely associated with development and prognosis of human cancers. LncRNA SNHG16 is reportedly involved in human cancer; however, its roles in multiple myeloma (MM) remain unclear. Methods In this study, we investigated the function and molecular mechanisms of SNHG16 in MM. MM cells were transfected with si-SNHG16 or si-NC. SNHG16 expression levels was measured by qRT-PCR. Cell proliferation was monitored using the MTS. Flow cytometry assay was performed to measure the cell cycle and apoptosis. Luciferase reporter assay were performed to confirm the sponged miRNAs of SNHG16. Results SNHG16 expression was up-regulated in MM tissues. SNHG16 knockdown suppressed cell proliferation, arrested cell cycle transition from G1 to S phase, and promoted the apoptosis of MM cells. Moreover, SNHG16 knockdown promoted cleaved-Caspase-3, cleaved-Caspase-9, Foxa3a, and Bax expression, while markedly inhibiting CCND1, Bcl-2, Cyclin D1, PI3K, and p-AKT expression in MM cells. miR-342-3p was a direct target of SNHG16. SNHG16 knockdown significantly increased miR-342-3p expression in MM cells. Overexpression miR-342-3p markedly suppressed cell proliferation, arrested cell cycle transition from G1 to S phase, and promoted apoptosis of MM cells. Overexpression of miR-342-3p markedly promoted cleaved-Caspase-3/-9, Foxa3a, and Bax expression, and inhibited CCND1, Bcl-2, Cyclin D1, PI3K, and p-AKT expression in MM cells. Additionally, repression of miR-342-3p could rescue the effect of SNHG16 knockdown on MM cell proliferation, cycle arrest, apoptosis, and related protein expression. Conclusion Knockdown of lncRNA SNHG16 suppresses MM cell proliferation by sponging miR-342-3p, implicating SNHG16 as a novel therapeutic target for MM.
Collapse
Affiliation(s)
- Xi Yang
- 1Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, 215006 Jiangsu People's Republic of China.,2Department of Hematology, Affiliated Hospital of NanTong University, No.20 Xishi Road, Nantong, 226001 Jiangsu People's Republic of China
| | - Hongming Huang
- 2Department of Hematology, Affiliated Hospital of NanTong University, No.20 Xishi Road, Nantong, 226001 Jiangsu People's Republic of China
| | - Xinfeng Wang
- 2Department of Hematology, Affiliated Hospital of NanTong University, No.20 Xishi Road, Nantong, 226001 Jiangsu People's Republic of China
| | - Haiyan Liu
- 2Department of Hematology, Affiliated Hospital of NanTong University, No.20 Xishi Road, Nantong, 226001 Jiangsu People's Republic of China
| | - Hong Liu
- 2Department of Hematology, Affiliated Hospital of NanTong University, No.20 Xishi Road, Nantong, 226001 Jiangsu People's Republic of China
| | - Zenghua Lin
- 2Department of Hematology, Affiliated Hospital of NanTong University, No.20 Xishi Road, Nantong, 226001 Jiangsu People's Republic of China
| |
Collapse
|
12
|
Singh AK, Cancelas JA. Gap Junctions in the Bone Marrow Lympho-Hematopoietic Stem Cell Niche, Leukemia Progression, and Chemoresistance. Int J Mol Sci 2020; 21:E796. [PMID: 31991829 PMCID: PMC7038046 DOI: 10.3390/ijms21030796] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/19/2020] [Accepted: 01/23/2020] [Indexed: 12/15/2022] Open
Abstract
Abstract: The crosstalk between hematopoietic stem cells (HSC) and bone marrow (BM) microenvironment is critical for homeostasis and hematopoietic regeneration in response to blood formation emergencies after injury, and has been associated with leukemia transformation and progression. Intercellular signals by the BM stromal cells in the form of cell-bound or secreted factors, or by physical interaction, regulate HSC localization, maintenance, and differentiation within increasingly defined BM HSC niches. Gap junctions (GJ) are comprised of arrays of membrane embedded channels formed by connexin proteins, and control crucial signaling functions, including the transfer of ions, small metabolites, and organelles to adjacent cells which affect intracellular mechanisms of signaling and autophagy. This review will discuss the role of GJ in both normal and leukemic hematopoiesis, and highlight some of the most novel approaches that may improve the efficacy of cytotoxic drugs. Connexin GJ channels exert both cell-intrinsic and cell-extrinsic effects on HSC and BM stromal cells, involved in regenerative hematopoiesis after myelosuppression, and represent an alternative system of cell communication through a combination of electrical and metabolic coupling as well as organelle transfer in the HSC niche. GJ intercellular communication (GJIC) in the HSC niche improves cellular bioenergetics, and rejuvenates damaged recipient cells. Unfortunately, they can also support leukemia proliferation and survival by creating leukemic niches that provide GJIC dependent energy sources and facilitate chemoresistance and relapse. The emergence of new strategies to disrupt self-reinforcing malignant niches and intercellular organelle exchange in leukemic niches, while at the same time conserving normal hematopoietic GJIC function, could synergize the effect of chemotherapy drugs in eradicating minimal residual disease. An improved understanding of the molecular basis of connexin regulation in normal and leukemic hematopoiesis is warranted for the re-establishment of normal hematopoiesis after chemotherapy.
Collapse
Affiliation(s)
- Abhishek K. Singh
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH 45229, USA;
- Hoxworth Blood Center, University of Cincinnati Academic Health Center, 3333 Burnet Ave., Cincinnati, OH 45229, USA
| | - Jose A. Cancelas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH 45229, USA;
- Hoxworth Blood Center, University of Cincinnati Academic Health Center, 3333 Burnet Ave., Cincinnati, OH 45229, USA
| |
Collapse
|
13
|
Li CH, Hao ML, Sun Y, Wang ZJ, Li JL. Ultrastructure of gap junction and Cx43 expression in gastric cancer tissues of the patients. Arch Med Sci 2020; 16:352-358. [PMID: 32190146 PMCID: PMC7069450 DOI: 10.5114/aoms.2020.92859] [Citation(s) in RCA: 5] [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: 04/28/2017] [Accepted: 08/21/2017] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION Gap junctions are intercellular channels formed by connexin facilitating communication between cells by allowing transfer of ions and small signaling molecules. Connexin 43 (Cx43) is the most ubiquitous connexin in human tissues. Ample evidence suggests the role of gap junction and its connexins such as connexin 43 in human cancers including gastric cancer, which has an important place in the worldwide incidence of cancer and cancer-related deaths. Due to a number of contradictory studies and insufficient detailed examination in specific cancers, such as gastric cancer, more data on the role of gap junctions and their connexins such as Cx43 involved in gastric cancer remain necessary. MATERIAL AND METHODS Transmission electron microscopy, Western blotting and RT-PCR were used to show the ultrastructure damage of the gap junction in the gastric carcinoma tissue as well as the expression of Cx43 protein and mRNA, respectively. RESULTS Ultrastructure damage of the gap junction in gastric carcinoma tissue was shown while poorly differentiated tissue experienced greater damage. The expression of Cx43 protein and mRNA was higher in healthy gastric tissue than in carcinomatous gastric tissue (p < 0.05). There was higher expression of Cx43 protein and mRNA in high-medium differentiation than in poor differentiation (p < 0.05). Cx43 protein and mRNA expression is not statistically significant for different ages and sex (such as for > 56 and ≤ 56 years) (p > 0.05). CONCLUSIONS Ultrastructural changes of gap junctions with abnormal Cx43 expression are associated with occurrence and development of gastric cancer, which provides a new research direction for gastric cancer pathogenesis and targeted therapy.
Collapse
Affiliation(s)
- Chun-Hui Li
- Department of Pathology, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Mei-Ling Hao
- Department of Pathology, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Yu Sun
- Department of Pathology, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Zhu-Jun Wang
- Department of Pathology, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Jian-Ling Li
- Department of Pathology, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| |
Collapse
|
14
|
Lei J, Fu Y, Zhuang Y, Zhang K, Lu D. miR-382-3p suppressed IL-1β induced inflammatory response of chondrocytes via the TLR4/MyD88/NF-κB signaling pathway by directly targeting CX43. J Cell Physiol 2019; 234:23160-23168. [PMID: 31144313 DOI: 10.1002/jcp.28882] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/18/2022]
Abstract
miR-382-3p has been reported to be upregulated in synovial membrane in knee osteoarthritis (OA). Nevertheless, its role in OA remains largely unknown. The aim of this study was to investigate the specific function and mechanisms of miR-382-3p in the course of OA. In this study, human OA chondrocytes were pretreated with interleukin-1β (IL-1β) at 5 ng/ml for 12 hr to stimulate inflammatory response and matrix metalloproteinases (MMPs) expression in chondrocytes. Meanwhile, miR-382-3p was downregulated in IL-1β-stimulated chondrocytes. In addition, we found that miR-382-3p directly interacts with connexin 43 (CX43) and attenuates the increase of cytochrome c oxidase polypeptide II, inducible nitric oxide synthase, and MMP-1/13 that is induced by IL-1β. Furthermore, our observations indicated that miR-382-3p inhibited the expression of Toll-like receptor 4 (TLR4), Myeloid differentiation primary response 88 (MyD88) and nuclear factor κB (NF-κB) in IL-1β-stimulated chondrocytes, while CX43 overexpression could partly reverse these decreases. In conclusion, miR-382-3p participated in OA may through the TLR4/MyD88/NF-κB signaling pathway by directly targeting CX43.
Collapse
Affiliation(s)
- Jinlai Lei
- Department of Orthopedic Trauma, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaansi, China
| | - Yahui Fu
- Department of Orthopedic Trauma, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaansi, China
| | - Yan Zhuang
- Department of Orthopedic Trauma, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaansi, China
| | - Kun Zhang
- Department of Orthopedic Trauma, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaansi, China
| | - Daigang Lu
- Department of Orthopedic Trauma, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaansi, China
| |
Collapse
|
15
|
Naji A, Favier B, Deschaseaux F, Rouas-Freiss N, Eitoku M, Suganuma N. Mesenchymal stem/stromal cell function in modulating cell death. Stem Cell Res Ther 2019; 10:56. [PMID: 30760307 PMCID: PMC6374902 DOI: 10.1186/s13287-019-1158-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) delivered as cell therapy to individuals with degenerative and/or inflammatory disorders can help improve organ features and resolve inflammation, as demonstrated in preclinical studies and to some extent in clinical studies. MSCs have trophic, homing/migration, and immunosuppression functions, with many benefits in therapeutics. MSC functions are thought to depend on the paracrine action of soluble factors and/or the expression of membrane-bound molecules, mostly belonging to the molecular class of adhesion molecules, chemokines, enzymes, growth factors, and interleukins. Cutting-edge studies underline bioactive exchanges, including that of ions, nucleic acids, proteins, and organelles transferred from MSCs to stressed cells, thereby improving the cells' survival and function. From this aspect, MSC death modulation function appears as a decisive biological function that could carry a significant part of the therapeutic effects of MSCs. Identifying the function and modes of actions of MSCs in modulating cell death may be exploited to enhance consistency and efficiency of cell therapy that is based on MSCs as medical treatment for degenerative and/or inflammatory diseases. Here, we review the essentials of MSC functions in modulating cell death in unfit cells, and its modes of actions based on current advances and outline the clinical implications.
Collapse
Affiliation(s)
- Abderrahim Naji
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School (KMS), Kochi University, Kohasu, Oko-Cho, Nankoku City, Kochi Prefecture, 783-8505, Japan.
| | - Benoit Favier
- CEA-Université Paris Sud INSERM U1184, IDMIT Department, IBFJ, DRF, Fontenay-aux-Roses, France
| | - Frédéric Deschaseaux
- STROMALab, UMR 5273 CNRS, INSERM U1031, Etablissement Français du Sang (EFS) Occitanie, Université de Toulouse, Toulouse, France
| | - Nathalie Rouas-Freiss
- CEA, DRF-Institut Francois Jacob, Division de recherche en hématologie et immunologie (SRHI), Hôpital Saint-Louis, Paris, France
| | - Masamitsu Eitoku
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School (KMS), Kochi University, Kohasu, Oko-Cho, Nankoku City, Kochi Prefecture, 783-8505, Japan
| | - Narufumi Suganuma
- Department of Environmental Medicine, Cooperative Medicine Unit, Research and Education Faculty, Medicine Science Cluster, Kochi Medical School (KMS), Kochi University, Kohasu, Oko-Cho, Nankoku City, Kochi Prefecture, 783-8505, Japan.
| |
Collapse
|
16
|
Drug resistance in multiple myeloma. Cancer Treat Rev 2018; 70:199-208. [DOI: 10.1016/j.ctrv.2018.09.001] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 08/05/2018] [Accepted: 09/01/2018] [Indexed: 02/07/2023]
|
17
|
Sirin DY, Karaarslan N. Evaluation of the effects of pregabalin on chondrocyte proliferation and CHAD, HIF-1α, and COL2A1 gene expression. Arch Med Sci 2018; 14:1340-1347. [PMID: 30393488 PMCID: PMC6209709 DOI: 10.5114/aoms.2018.73134] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 01/03/2018] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION The aim of the present study is to investigate the effects of pregabalin (PGB) on chondrocyte proliferation and collagen type II (COL2A1), hypoxia-inducible factor 1-α (HIF-1α), and chondroadherin (CHAD) gene expression in osteoarthritic chondrocytes. MATERIAL AND METHODS Standard primary chondrocyte cultures were prepared using osteochondral tissues that were surgically obtained from 6 patients with gonarthrosis. Cell morphology was evaluated using an inverted microscope, and cell death and proliferation were determined through MTT analysis, which was confirmed by AO/PI staining and statistically evaluated. The expression levels of CHAD, COL2A1, and HIF-1α genes were assessed using gene-specific TaqMan Gene Expression Assays. RESULTS MTT analyses showed that PGB administration did not have a negative or toxic effect on cell viability and proliferation in cultured chondrocytes (p < 0.001), but in our morphological evaluation extracellular matrix development was observed to be weaker in cultures treated with PGB. After 24 h of treatment, COL2A1, HIF-1α, and CHAD gene expression decreased in the groups to which PGB was applied compared to gene expression before the experiment (at 0 h); at 48 h, CHAD and HIF-1α expression increased to the same level as the control group, but the expression of COL2A1 continued to decrease. CONCLUSIONS Further studies need to be conducted with more participants to prove that there is a negative correlation between extracellular matrix formation and PGB administration. Our preliminary data show that even at low doses and over short-term administration, PGB may affect chondrocyte cells at the gene-expression level.
Collapse
Affiliation(s)
- Duygu Yasar Sirin
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Namik Kemal University, Tekirdag, Turkey
| | - Numan Karaarslan
- Department of Neurosurgery, School of Medicine, Namik Kemal University, Tekirdag, Turkey
| |
Collapse
|
18
|
Yang M, Zhang L, Wang X, Zhou Y, Wu S. Down-regulation of miR-203a by lncRNA PVT1 in multiple myeloma promotes cell proliferation. Arch Med Sci 2018; 14:1333-1339. [PMID: 30393487 PMCID: PMC6209721 DOI: 10.5114/aoms.2018.73975] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/27/2018] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Emerging evidence has indicated that long non-coding RNAs (lncRNAs) play vital roles in multiple myeloma (MM) development and progression. However, the underlying mechanism of PVT1 in MM remains unclear. MATERIAL AND METHODS QRT-PCR was used to detect the expression of PVT1 and miR-203a in MM samples and cell lines. The effects of PVT1 on MM cell proliferation and apoptosis were determined by CCK8 assay and flow cytometer assay, respectively. Bioinformatics methods were used to identify the downstream target miRNAs of PVT1. RESULTS We found that the expression of PVT1 was upregulated in MM samples and cell lines (p < 0.05), while the expression of miR-203a was downregulated in MM samples and cell lines (p < 0.05). There was a negative correlation between PVT1 expression and miR-203a expression in MM samples (p < 0.05). In in vitro function assays, we found that PVT1 inhibition suppressed MM cell proliferation and induced MM cell apoptosis (p < 0.05). The bioinformatics approach predicted that PVT1 sponge miR-203a would modulate MM cells. Rescue experiments confirmed the recovering roles of miR-203a for PVT1 on MM progression. CONCLUSIONS In the present study, we found that lncRNA PVT1 could promote MM cell proliferation and induce cell apoptosis by inhibiting miR-203a expression. Therefore, PVT1 may represent a potential therapeutic target for the treatment of MM patients.
Collapse
Affiliation(s)
- Man Yang
- Department of Hematology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Lingxiu Zhang
- Department of Hematology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Xiufeng Wang
- Department of Hematology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Yanjun Zhou
- Clinical Lab, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Sun Wu
- Department of Hematology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| |
Collapse
|
19
|
Jin J, Wang T, Wang Y, Chen S, Li Z, Li X, Zhang J, Wang J. SRC3 expressed in BMSCs promotes growth and migration of multiple myeloma cells by regulating the expression of Cx43. Int J Oncol 2017; 51:1694-1704. [PMID: 29075794 PMCID: PMC5673026 DOI: 10.3892/ijo.2017.4171] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 09/28/2017] [Indexed: 12/14/2022] Open
Abstract
Interactions between bone marrow stromal cells (BMSCs) and multiple myeloma cells significantly contribute to the progression of multiple myeloma (MM). However, little is known about the molecular mechanisms that regulate these interactions. Connexin-43 (Cx-43) has been implicated in the interplay between BMSCs and MM cells. In this study, we hypothesized that the steroid receptor co-activator-3 (SRC3) expressed in BMSCs regulates the expression of Cx-43 to promote the proliferation and migration of myeloma cells. To address this, we co-cultured a human multiple myeloma cell line, RPMI-8226 transfected with either control BMSCs or sh-SRC3-BMSCs. We found that knocking down SRC3 expression in BMSCs inhibited the proliferation and migration of RPMI-8226 cells. In addition, we found that co-culturing RPMI 8266 cells with BMSCs increased Cx43 expression, while knocking down SRC3 expression in BMSCs decreased Cx43 expression. Moreover, our work revealed that SRC3 in BMSCs regulates Cx43 expression via the mitogen-activated protein kinase (MAPK) pathway. To validate this result in vivo, we knocked down SRC3 expression in BMSCs in nude mice and found that tumor growth and cell apoptosis were significantly decreased. In addition, mice treated with either RPMI 8266 cells overexpressing Cx43 or with a P38 MAPK inhibitor (SB202190) exhibited increased intratumoral leukocyte populations and promoted cell apoptosis in tumor tissue. Our findings demonstrate how SRC3 and Cx43 regulation between BMSCs and myeloma cells mediate cell growth and disease progression, with potential implications for prognosis and therapeutic interventions.
Collapse
Affiliation(s)
- Jie Jin
- Department of Hematology, The Third Affiliated Daping Hospital of Army Medical University, Chongqing 400042, P.R. China
| | - Tao Wang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Army Medical University, Chongqing 400038, P.R. China
| | - Yu Wang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Army Medical University, Chongqing 400038, P.R. China
| | - Shidi Chen
- Department of Hematology, The Third Affiliated Daping Hospital of Army Medical University, Chongqing 400042, P.R. China
| | - Zheng Li
- Department of Hematology, The Third Affiliated Daping Hospital of Army Medical University, Chongqing 400042, P.R. China
| | - Xiang Li
- Department of Hematology, The Third Affiliated Daping Hospital of Army Medical University, Chongqing 400042, P.R. China
| | - Jiazhen Zhang
- Department of Hematology, The Third Affiliated Daping Hospital of Army Medical University, Chongqing 400042, P.R. China
| | - Jin Wang
- Department of Hematology, The Third Affiliated Daping Hospital of Army Medical University, Chongqing 400042, P.R. China
| |
Collapse
|
20
|
Ribeiro-Rodrigues TM, Martins-Marques T, Morel S, Kwak BR, Girão H. Role of connexin 43 in different forms of intercellular communication - gap junctions, extracellular vesicles and tunnelling nanotubes. J Cell Sci 2017; 130:3619-3630. [PMID: 29025971 DOI: 10.1242/jcs.200667] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Communication is important to ensure the correct and efficient flow of information, which is required to sustain active social networks. A fine-tuned communication between cells is vital to maintain the homeostasis and function of multicellular or unicellular organisms in a community environment. Although there are different levels of complexity, intercellular communication, in prokaryotes to mammalians, can occur through secreted molecules (either soluble or encapsulated in vesicles), tubular structures connecting close cells or intercellular channels that link the cytoplasm of adjacent cells. In mammals, these different types of communication serve different purposes, may involve distinct factors and are mediated by extracellular vesicles, tunnelling nanotubes or gap junctions. Recent studies have shown that connexin 43 (Cx43, also known as GJA1), a transmembrane protein initially described as a gap junction protein, participates in all these forms of communication; this emphasizes the concept of adopting strategies to maximize the potential of available resources by reutilizing the same factor in different scenarios. In this Review, we provide an overview of the most recent advances regarding the role of Cx43 in intercellular communication mediated by extracellular vesicles, tunnelling nanotubes and gap junctions.
Collapse
Affiliation(s)
- Teresa M Ribeiro-Rodrigues
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga de Sta Comba, 3000-548 Coimbra, Portugal.,CNC.IBILI, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Tânia Martins-Marques
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga de Sta Comba, 3000-548 Coimbra, Portugal.,CNC.IBILI, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Sandrine Morel
- Dept. of Pathology and Immunology, and Dept. of Medical Specialties - Cardiology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Brenda R Kwak
- Dept. of Pathology and Immunology, and Dept. of Medical Specialties - Cardiology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Henrique Girão
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Azinhaga de Sta Comba, 3000-548 Coimbra, Portugal .,CNC.IBILI, University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
21
|
Zhang Y, Wang Z, Zhang L, Zhou D, Sun Y, Wang P, Ju S, Chen P, Li J, Fu J. Impact of connexin 43 coupling on survival and migration of multiple myeloma cells. Arch Med Sci 2017; 13:1335-1346. [PMID: 29181063 PMCID: PMC5701698 DOI: 10.5114/aoms.2017.71065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 11/01/2016] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Gap junctions (GJs) represent the best known intercellular communication (IC) system and are membrane-spanning channels that facilitate intercellular communication by allowing small signaling molecules to pass from cell to cell. In this study, we constructed an amino terminus of human Cx43 (Cx43NT-GFP), verified the overexpression of Cx43-NT in HUVEC cells and explored the impact of gap junctions (GJs) on multiple myeloma (MM). MATERIAL AND METHODS The levels of phosphorylated Cx43(s368) and the change of MAPK pathway associated molecules (ERK1/2, JNK, p38, NFκB) were also investigated in our cell models. Cx43 mRNA and proteins were detected in both MM cell lines and mesenchymal stem cells (MSCs). Dye transfer assays demonstrated that gap junction intercellular communication (GJIC) occurring via Cx43 situated between MM and MSCs or MM and HUVECCx43NT is functional. RESULTS Our results present evidence for a channel-dependent modulator action of connexin 43 on the migratory activity of MM cells toward MSCs or HUVECCx43-N was higher than those of spontaneous migration (p < 0.05) and protection them from apoptosis in the presence of dexamethasone via cytokines secretion. In the meantime, the migration of MM cells involves an augmented response of p38 and JNK signaling pathway of carboxyl tail of the protein. CONCLUSIONS Our data suggest that GJIC between MM and MSCs is one of the essential factors in tumor cell proliferation and drug sensitivity, and is implicated in MM pathogenesis.
Collapse
Affiliation(s)
- Yangmin Zhang
- Department of Hematology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ziyan Wang
- Department of Hematology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liying Zhang
- Department of Hematology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Dongming Zhou
- Department of Hematology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Sun
- Department of Hematology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Panjun Wang
- Department of Hematology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Songguang Ju
- Department of Immunology, Medical College of Soochow University, Suzhou, China
| | - Ping Chen
- Department of Hematology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Li
- Department of Hematology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinxiang Fu
- Department of Hematology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|