1
|
Mi Y, Jiang P, Luan J, Feng L, Zhang D, Gao X. Peptide‑based therapeutic strategies for glioma: Current state and prospects. Peptides 2025; 185:171354. [PMID: 39922284 DOI: 10.1016/j.peptides.2025.171354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Glioma is a prevalent form of primary malignant central nervous system tumor, characterized by its cellular invasiveness, rapid growth, and the presence of the blood-brain barrier (BBB)/blood-brain tumor barrier (BBTB). Current therapeutic approaches, such as chemotherapy and radiotherapy, have shown limited efficacy in achieving significant antitumor effects. Therefore, there is an urgent demand for new treatments. Therapeutic peptides represent an innovative class of pharmaceutical agents with lower immunogenicity and toxicity. They are easily modifiable via chemical means and possess deep tissue penetration capabilities which reduce side effects and drug resistance. These unique pharmacokinetic characteristics make peptides a rapidly growing class of new therapeutics that have demonstrated significant progress in glioma treatment. This review outlines the efforts and accomplishments in peptide-based therapeutic strategies for glioma. These therapeutic peptides can be classified into four types based on their anti-tumor function: tumor-homing peptides, inhibitor/antagonist peptides targeting cell surface receptors, interference peptides, and peptide vaccines. Furthermore, we briefly summarize the results from clinical trials of therapeutic peptides in glioma, which shows that peptide-based therapeutic strategies exhibit great potential as multifunctional players in glioma therapy.
Collapse
Affiliation(s)
- Yajing Mi
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Pengtao Jiang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Jing Luan
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Lin Feng
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Dian Zhang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Xingchun Gao
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China.
| |
Collapse
|
2
|
杜 越, 张 秀, 周 克, 金 星, 原 翔, 高 社. [RgpB contributes to chemoresistance in esophageal squamous cell carcinoma by preventing Cx43 degradation via inhibiting autophagosome-lysosome fusion]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:1670-1676. [PMID: 39505334 PMCID: PMC11744085 DOI: 10.12122/j.issn.1673-4254.2024.09.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Indexed: 11/08/2024]
Abstract
OBJECTIVE To investigate the mechanism through which RgpB, a virulence factor of Porphyromonas gingivalis (Pg), induces chemoresistance in esophageal squamous carcinoma. METHODS The autophagy-regulating factors that interact with RgpB were screened by immunoprecipitation-mass spectrometry. The interaction between RgpB and the autophagy regulator TBC1D5 was investigated using co-immunoprecipitation. The impact of Pg infection on the expression of esophageal cancer cell membrane receptor molecule Cx43 was assessed using Western blotting. Immunofluorescence assay was used to analyze the relationship among Lamp1, Cx43 and TBC1D5. The effect of Pg infection on autophagosome-lysosome fusion was evaluated using autophagy double fluorescence technique. The effects of Pg infection and a Cx43 inhibitor on proliferation of esophageal cancer cells after chemotherapy were examined with plate cloning assay and CCK-8 method. RESULTS Immunoprecipitation-mass spectrometry identified TBC1D5 as an autophagy regulator interacting with RgpB, and coimmunoprecipitation suggested that RgpB could directly bind to TBC1D5. In Pg-infected esophageal cancer cells, the expression of Cx43 on the cell membrane was significantly higher than that in non-infected cells. Immunofluorescence assay showed that the expression of Cx43 on the membrane of esophageal cancer cells increased significantly after Pg infection, which blocked autophagosome-lysosome fusion as shown by stubRFP-sensGFP-LC3 lentivirus study. Plate cloning assay and CCK-8 assay showed that the Cx43 inhibitor significantly attenuated the effect of Pg infection for promoting proliferation of esophageal cancer cells after chemotherapy. CONCLUSION Pg infection in esophageal cancer blocked autophagosome-lysosome fusion in the tumor cells, thereby preventing Cx43 from lysosomal degradation and leading to chemoresistance of esophageal cancer.
Collapse
Affiliation(s)
- 越 杜
- 河南省微生态与食管癌防治重点实验室//河南省肿瘤表观遗传重点实验室,河南 洛阳 471003Henan Provincial Key Laboratory of Microecology and Esophageal Cancer Prevention and Treatment, Henan Provincial Key Laboratory of Tumor Epigenetics, Luoyang 471003, China
- 河南科技大学第一附属医院//河南科技大学临床医学院,河南 洛阳 471003First Affiliated Hospital/School of Clinical Medicine, Henan University of Science and Technology, Luoyang 471003, China
| | - 秀森 张
- 河南省微生态与食管癌防治重点实验室//河南省肿瘤表观遗传重点实验室,河南 洛阳 471003Henan Provincial Key Laboratory of Microecology and Esophageal Cancer Prevention and Treatment, Henan Provincial Key Laboratory of Tumor Epigenetics, Luoyang 471003, China
- 河南科技大学第一附属医院//河南科技大学临床医学院,河南 洛阳 471003First Affiliated Hospital/School of Clinical Medicine, Henan University of Science and Technology, Luoyang 471003, China
| | - 克旭 周
- 河南省微生态与食管癌防治重点实验室//河南省肿瘤表观遗传重点实验室,河南 洛阳 471003Henan Provincial Key Laboratory of Microecology and Esophageal Cancer Prevention and Treatment, Henan Provincial Key Laboratory of Tumor Epigenetics, Luoyang 471003, China
- 河南科技大学第一附属医院//河南科技大学临床医学院,河南 洛阳 471003First Affiliated Hospital/School of Clinical Medicine, Henan University of Science and Technology, Luoyang 471003, China
| | - 星 金
- 河南省微生态与食管癌防治重点实验室//河南省肿瘤表观遗传重点实验室,河南 洛阳 471003Henan Provincial Key Laboratory of Microecology and Esophageal Cancer Prevention and Treatment, Henan Provincial Key Laboratory of Tumor Epigenetics, Luoyang 471003, China
- 河南科技大学第一附属医院//河南科技大学临床医学院,河南 洛阳 471003First Affiliated Hospital/School of Clinical Medicine, Henan University of Science and Technology, Luoyang 471003, China
| | - 翔 原
- 河南省微生态与食管癌防治重点实验室//河南省肿瘤表观遗传重点实验室,河南 洛阳 471003Henan Provincial Key Laboratory of Microecology and Esophageal Cancer Prevention and Treatment, Henan Provincial Key Laboratory of Tumor Epigenetics, Luoyang 471003, China
- 河南科技大学第一附属医院//河南科技大学临床医学院,河南 洛阳 471003First Affiliated Hospital/School of Clinical Medicine, Henan University of Science and Technology, Luoyang 471003, China
| | - 社干 高
- 河南省微生态与食管癌防治重点实验室//河南省肿瘤表观遗传重点实验室,河南 洛阳 471003Henan Provincial Key Laboratory of Microecology and Esophageal Cancer Prevention and Treatment, Henan Provincial Key Laboratory of Tumor Epigenetics, Luoyang 471003, China
- 河南科技大学第一附属医院//河南科技大学临床医学院,河南 洛阳 471003First Affiliated Hospital/School of Clinical Medicine, Henan University of Science and Technology, Luoyang 471003, China
| |
Collapse
|
3
|
Zefferino R, Conese M. A Vaccine against Cancer: Can There Be a Possible Strategy to Face the Challenge? Possible Targets and Paradoxical Effects. Vaccines (Basel) 2023; 11:1701. [PMID: 38006033 PMCID: PMC10674257 DOI: 10.3390/vaccines11111701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/07/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
Is it possible to have an available vaccine that eradicates cancer? Starting from this question, this article tries to verify the state of the art, proposing a different approach to the issue. The variety of cancers and different and often unknown causes of cancer impede, except in some cited cases, the creation of a classical vaccine directed at the causative agent. The efforts of the scientific community are oriented toward stimulating the immune systems of patients, thereby preventing immune evasion, and heightening chemotherapeutic agents effects against cancer. However, the results are not decisive, because without any warning signs, metastasis often occurs. The purpose of this paper is to elaborate on a vaccine that must be administered to a patient in order to prevent metastasis; metastasis is an event that leads to death, and thus, preventing it could transform cancer into a chronic disease. We underline the fact that the field has not been studied in depth, and that the complexity of metastatic processes should not be underestimated. Then, with the aim of identifying the target of a cancer vaccine, we draw attention to the presence of the paradoxical actions of different mechanisms, pathways, molecules, and immune and non-immune cells characteristic of the tumor microenvironment at the primary site and pre-metastatic niche in order to exclude possible vaccine candidates that have opposite effects/behaviors; after a meticulous evaluation, we propose possible targets to develop a metastasis-targeting vaccine. We conclude that a change in the current concept of a cancer vaccine is needed, and the efforts of the scientific community should be redirected toward a metastasis-targeting vaccine, with the increasing hope of eradicating cancer.
Collapse
Affiliation(s)
- Roberto Zefferino
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Massimo Conese
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| |
Collapse
|
4
|
Xu QR, Du XH, Huang TT, Zheng YC, Li YL, Huang DY, Dai HQ, Li EM, Fang WK. Role of Cell-Cell Junctions in Oesophageal Squamous Cell Carcinoma. Biomolecules 2022; 12:biom12101378. [PMID: 36291586 PMCID: PMC9599896 DOI: 10.3390/biom12101378] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 02/05/2023] Open
Abstract
Cell-cell junctions comprise various structures, including adherens junctions, tight junctions, desmosomes, and gap junctions. They link cells to each other in tissues and regulate tissue homeostasis in critical cellular processes. Recent advances in cell-cell junction research have led to critical discoveries. Cell-cell adhesion components are important for the invasion and metastasis of tumour cells, which are not only related to cell-cell adhesion changes, but they are also involved in critical molecular signal pathways. They are of great significance, especially given that relevant molecular mechanisms are being discovered, there are an increasing number of emerging biomarkers, targeted therapies are becoming a future therapeutic concern, and there is an increased number of therapeutic agents undergoing clinical trials. Oesophageal squamous cell carcinoma (ESCC), the most common histological subtype of oesophageal cancer, is one of the most common cancers to affect epithelial tissue. ESCC progression is accompanied by the abnormal expression or localisation of components at cell-cell junctions. This review will discuss the recent scientific developments related to the molecules at cell-cell junctions and their role in ESCC to offer valuable insights for readers, provide a global view of the relationships between position, construction, and function, and give a reference for future mechanistic studies, diagnoses, and therapeutic developments.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - En-Min Li
- Correspondence: (E.-M.L.); (W.-K.F.)
| | | |
Collapse
|
5
|
Jones JC, Bodenstine TM. Connexins and Glucose Metabolism in Cancer. Int J Mol Sci 2022; 23:ijms231710172. [PMID: 36077565 PMCID: PMC9455984 DOI: 10.3390/ijms231710172] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Connexins are a family of transmembrane proteins that regulate diverse cellular functions. Originally characterized for their ability to mediate direct intercellular communication through the formation of highly regulated membrane channels, their functions have been extended to the exchange of molecules with the extracellular environment, and the ability to modulate numerous channel-independent effects on processes such as motility and survival. Notably, connexins have been implicated in cancer biology for their context-dependent roles that can both promote or suppress cancer cell function. Moreover, connexins are able to mediate many aspects of cellular metabolism including the intercellular coupling of nutrients and signaling molecules. During cancer progression, changes to substrate utilization occur to support energy production and biomass accumulation. This results in metabolic plasticity that promotes cell survival and proliferation, and can impact therapeutic resistance. Significant progress has been made in our understanding of connexin and cancer biology, however, delineating the roles these multi-faceted proteins play in metabolic adaptation of cancer cells is just beginning. Glucose represents a major carbon substrate for energy production, nucleotide synthesis, carbohydrate modifications and generation of biosynthetic intermediates. While cancer cells often exhibit a dependence on glycolytic metabolism for survival, cellular reprogramming of metabolic pathways is common when blood perfusion is limited in growing tumors. These metabolic changes drive aggressive phenotypes through the acquisition of functional traits. Connections between glucose metabolism and connexin function in cancer cells and the surrounding stroma are now apparent, however much remains to be discovered regarding these relationships. This review discusses the existing evidence in this area and highlights directions for continued investigation.
Collapse
|
6
|
Segura IG, Secchi DG, Galíndez MF, Carrica A, Bologna-Molina R, Brunotto M, Centeno VA. Connexin 43, Bcl-2, Bax, Ki67, and E-cadherin patterns in oral squamous cell carcinoma and its relationship with GJA1 rs12197797 C/G. Med Oral Patol Oral Cir Bucal 2022; 27:e366-e374. [PMID: 35717615 PMCID: PMC9271350 DOI: 10.4317/medoral.25298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 06/06/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND To our knowledge, there is no useful and accurate prognostic biomarker or biomarkers for patients with oral squamous cell carcinoma (OSCC), a tumor with uncertain biological behavior, and unpredictable clinical progress. The purposes of this study were: a) to determine the expresión profile of Connexin 43, Bcl-2, Bax, E-cadherin, and Ki67 in patients with OSCC; b) identify the GJCA1 rs12197797 genotypic composition. MATERIAL AND METHODS A cross-sectional study using genomic DNA and biopsy samples extracted from the oral mucosa with/without OSCC, older than 18 years, both genders, attended at Facultad de Odontología, Universidad Nacional Córdoba. Immunostaining for Cx43, Bcl-2, Bax, E-cadherin, and Ki67 and genotyping GJA1 rs12197797 by RFLP were performed. Odds Ratio (95% CI), Spearman Coefficient were estimated. Mann-Whitney test was applied to analyze immunostaining between controls/cases (p <0.05 was set for statistical significance). RESULTS GG (mutant) was the most frequent genotype in patients with OSCC diagnosis (53.2%) in relation to CC "healthy" genotype (p=0.00487; OR=7.33; CI95% [1.1-54.7]). And, the allele G (mutant) had a presence in 75.5% of OSCC patients. However, no significant association was observed between alleles C/G and diagnosis (p=0.0565). The heterozygous genotype was the most frequent in the patients of both groups Cx43 and E-cadherin markers were lower in OSCCs in relation to controls. Ki67 and Bcl-2 immunolabeling were high on OSCC, and Bax immunomarker was diminished in OSCC. CONCLUSIONS We hypothesized that the oral epithelium losses Connexin 43 and E-cadherin in the membrane, which modifies cell differentiation. The Ki67 and Bcl2 overexpression would increase the cell density in the tissue, by promoting proliferation and decreasing apoptosis. And, this study shows evidence that patients who carry on allele G of GJA1rs12197797 could be at risk of developing OSCC.
Collapse
Affiliation(s)
- I-G Segura
- Universidad Nacional de Córdoba, Facultad de Odontología Haya de la Torre S/N, Córdoba, Postal Code: X5000, Argentina
| | | | | | | | | | | | | |
Collapse
|
7
|
Polewko-Klim A, Zhu S, Wu W, Xie Y, Cai N, Zhang K, Zhu Z, Qing T, Yuan Z, Xu K, Zhang T, Lu M, Ye W, Chen X, Suo C, Rudnicki WR. Identification of Candidate Therapeutic Genes for More Precise Treatment of Esophageal Squamous Cell Carcinoma and Adenocarcinoma. Front Genet 2022; 13:844542. [PMID: 35664298 PMCID: PMC9161154 DOI: 10.3389/fgene.2022.844542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/20/2022] [Indexed: 11/23/2022] Open
Abstract
The standard therapy administered to patients with advanced esophageal cancer remains uniform, despite its two main histological subtypes, namely esophageal squamous cell carcinoma (SCC) and esophageal adenocarcinoma (AC), are being increasingly considered to be different. The identification of potential drug target genes between SCC and AC is crucial for more effective treatment of these diseases, given the high toxicity of chemotherapy and resistance to administered medications. Herein we attempted to identify and rank differentially expressed genes (DEGs) in SCC vs. AC using ensemble feature selection methods. RNA-seq data from The Cancer Genome Atlas and the Fudan-Taizhou Institute of Health Sciences (China). Six feature filters algorithms were used to identify DEGs. We built robust predictive models for histological subtypes with the random forest (RF) classification algorithm. Pathway analysis also be performed to investigate the functional role of genes. 294 informative DEGs (87 of them are newly discovered) have been identified. The areas under receiver operator curve (AUC) were higher than 99.5% for all feature selection (FS) methods. Nine genes (i.e., ERBB3, ATP7B, ABCC3, GALNT14, CLDN18, GUCY2C, FGFR4, KCNQ5, and CACNA1B) may play a key role in the development of more directed anticancer therapy for SCC and AC patients. The first four of them are drug targets for chemotherapy and immunotherapy of esophageal cancer and involved in pharmacokinetics and pharmacodynamics pathways. Research identified novel DEGs in SCC and AC, and detected four potential drug targeted genes (ERBB3, ATP7B, ABCC3, and GALNT14) and five drug-related genes.
Collapse
Affiliation(s)
- Aneta Polewko-Klim
- Institute of Computer Science, University in Bialystok, Białystok, Poland
| | - Sibo Zhu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Weicheng Wu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Yijing Xie
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Ning Cai
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Kexun Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Zhen Zhu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Tao Qing
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Ziyu Yuan
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Kelin Xu
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Tiejun Zhang
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
| | - Ming Lu
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
| | - Weimin Ye
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Xingdong Chen
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| | - Chen Suo
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, China
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, China
| | - Witold R. Rudnicki
- Institute of Computer Science, University in Bialystok, Białystok, Poland
- Computational Centre, University of Bialystok, Białystok, Poland
| |
Collapse
|
8
|
Huang Z, Zhang Z, Zhou C, Liu L, Huang C. Epithelial–mesenchymal transition: The history, regulatory mechanism, and cancer therapeutic opportunities. MedComm (Beijing) 2022; 3:e144. [PMID: 35601657 PMCID: PMC9115588 DOI: 10.1002/mco2.144] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 02/05/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT) is a program wherein epithelial cells lose their junctions and polarity while acquiring mesenchymal properties and invasive ability. Originally defined as an embryogenesis event, EMT has been recognized as a crucial process in tumor progression. During EMT, cell–cell junctions and cell–matrix attachments are disrupted, and the cytoskeleton is remodeled to enhance mobility of cells. This transition of phenotype is largely driven by a group of key transcription factors, typically Snail, Twist, and ZEB, through epigenetic repression of epithelial markers, transcriptional activation of matrix metalloproteinases, and reorganization of cytoskeleton. Mechanistically, EMT is orchestrated by multiple pathways, especially those involved in embryogenesis such as TGFβ, Wnt, Hedgehog, and Hippo, suggesting EMT as an intrinsic link between embryonic development and cancer progression. In addition, redox signaling has also emerged as critical EMT modulator. EMT confers cancer cells with increased metastatic potential and drug resistant capacity, which accounts for tumor recurrence in most clinic cases. Thus, targeting EMT can be a therapeutic option providing a chance of cure for cancer patients. Here, we introduce a brief history of EMT and summarize recent advances in understanding EMT mechanisms, as well as highlighting the therapeutic opportunities by targeting EMT in cancer treatment.
Collapse
Affiliation(s)
- Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Chengwei Zhou
- Department of Thoracic Surgery the Affiliated Hospital of Medical School of Ningbo University Ningbo China
| | - Lin Liu
- Department of Thoracic Surgery the Affiliated Hospital of Medical School of Ningbo University Ningbo China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| |
Collapse
|
9
|
Savic I, Milovanovic P, Opric S, Ivanovic N, Oprić D. Expression of connexin-43 in surgical resections of primary tumor and lymph node metastases of squamous cell carcinoma and adenocarcinoma of the lung: a retrospective study. PeerJ 2022; 10:e13055. [PMID: 35287344 PMCID: PMC8917803 DOI: 10.7717/peerj.13055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 02/12/2022] [Indexed: 01/11/2023] Open
Abstract
Background Connexins are transmembrane proteins forming gap junctions between the cells, which allow intercellular communication. Significance of gap junctions and connexins in lung carcinoma is not yet understood. The objective of the study was to investigate immunohistochemical expression and the localization of connexin-43 (Cx43) in primary lung carcinoma and its lymphatic metastases. Methods Surgical specimens of excised tumors from 88 patients (45 men and 43 women, 61.9 ± 7.4 years) with lung carcinoma (52 adenocarcinoma (AC), 36 squamous cell carcinoma (SqCC)) who were operated on at the University Hospital "Bezanijska Kosa" in a five-year period (2012-2016) were used. We conducted immunohistochemical staining for Cx43 and measured the degree of expression (percentage of positive cells and staining intensity) as well as localization of Cx43 in primary tumor and in lymphatic metastases. Results Immunohistochemical analysis of the primary tumors revealed that SqCC showed significantly higher percentage of tumor cells expressing Cx43 as well as higher staining intensity than AC (p < 0.001). Almost 70% of samples with SqCC showed high Cx43 expression, whereas AC showed no expression in more than 50% of cases. Localization of Cx43 expression was most often cytoplasmic (AC and SqCC) and combined membranous and cytoplasmic (SqCC) with very rare instances of nuclear localization (AC). Almost the same pattern in distribution, intensity, and localization of Cx43 expression was observed in the lymph node metastases; however, almost a third of AC cases changed the pattern of Cx43 expression in the metastasis compared to primary tumor. Conclusion The results of this study showed that lung carcinomas express Cx43 in more than 65% of cases and that it was aberrantly localized (not membranous localization). We highlighted that SqCC expressed Cx43 more than did AC, both in primary tumor and lymphatic metastases. Further research is needed to establish whether Cx43 could be used as a prognostic biomarker in lung carcinoma.
Collapse
Affiliation(s)
- Ivana Savic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | | | | | | - Dejan Oprić
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
10
|
Yang ZJ, Bi QC, Gan LJ, Zhang LL, Wei MJ, Hong T, Liu R, Qiu CL, Han XJ, Jiang LP. Exosomes Derived from Glioma Cells under Hypoxia Promote Angiogenesis through Up-regulated Exosomal Connexin 43. Int J Med Sci 2022; 19:1205-1215. [PMID: 35919821 PMCID: PMC9339413 DOI: 10.7150/ijms.71912] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 06/18/2022] [Indexed: 12/04/2022] Open
Abstract
Glioblastoma multiform (GBM) is a highly aggressive primary brain tumor. Exosomes derived from glioma cells under a hypoxic microenvironment play an important role in tumor biology including metastasis, angiogenesis and chemoresistance. However, the underlying mechanisms remain to be elucidated. In this study, we aimed to explore the role of connexin 43 on exosomal uptake and angiogenesis in glioma under hypoxia. U251 cells were exposed to 3% oxygen to achieve hypoxia, and the expression levels of HIF-1α and Cx43, involved in the colony formation and proliferation of cells were assessed. Exosomes were isolated by differential velocity centrifugation from U251 cells under normoxia and hypoxia (Nor-Exos and Hypo-Exos), respectively. Immunofluorescence staining, along with assays for CCK-8, tube formation and wound healing along with a transwell assay were conducted to profile exosomal uptake, proliferation, tube formation, migration and invasion of HUVECs, respectively. Our results revealed that Hypoxia significantly up-regulated the expression of HIF-1α in U251 cells as well as promoting proliferation and colony number. Hypoxia also increased the level of Cx43 in U251 cells and in the exosomes secreted. The uptake of Dio-stained Hypo-Exos by HUVECs was greater than that of Nor-Exos, and inhibition of Cx43 by 37,43gap27 or lenti-Cx43-shRNA efficiently prevented the uptake of Hypo-Exos by recipient endothelial cells. In addition, the proliferation and total loops of HUVECs were remarkably increased at 24 h, 48 h, and 10 h after Hypo-Exos, respectively. Notably, 37,43gap27, a specific Cx-mimetic peptide blocker of Cx37 and Cx43, efficiently alleviated Hypo-Exos-induced proliferation and tube formation by HUVECs. Finally, 37,43gap27 also significantly attenuated Hypo-Exos-induced migration and invasion of HUVECs. These findings demonstrate that exosomal Cx43 contributes to glioma angiogenesis mediated by Hypo-Exos, and suggests that exosomal Cx43 might serve as a potential therapeutic target for glioblastoma.
Collapse
Affiliation(s)
- Zhang-Jian Yang
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| | - Qiu-Chen Bi
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| | - Li-Jun Gan
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| | - Le-Ling Zhang
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| | - Min-Jun Wei
- Department of Neurosurgery, First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Tao Hong
- Department of Neurosurgery, First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Rong Liu
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| | - Cheng-Lin Qiu
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| | - Xiao-Jian Han
- Institute of Geriatrics, Jiangxi provincial People's Hospital, First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China.,Department of Neurology, Jiangxi provincial People's Hospital, First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, China
| | - Li-Ping Jiang
- Jiangxi Provincial Key Laboratory of Drug Targets and Drug Screening, School of Pharmaceutical Science, Nanchang University, Nanchang 330006, China
| |
Collapse
|
11
|
Savić I, Ivanović N, Oprić D. Role of gap junctions and connexin 43 in cancerogenesis and tumor metastasis. MEDICINSKI PODMLADAK 2022. [DOI: 10.5937/mp73-38352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Gap junctions are intercellular connections that enable direct communication between neighboring cells. They are important in tissue homeostasis, cell growth, and differentiation. They are composed of connexin proteins, of which the most common and most studied is connexin 43. The role of connexin 43 in the development and progression of tumors is contradictory. The aim of this paper is to summarize the current state of knowledge on the expression of connexin 43 in various primary and secondary tumors, in order to explain its role in the development and progression of malignant tumors. Previous studies have examined the expression of connexin 43 in various primary and secondary tumors, as well as its association with prognosis. The expression of connexin 43 has been shown to be associated with various aspects of tumor behavior. However, it has been shown that the expression of connexin 43 differs between different types and localizations of tumors, as well as between different stages in tumor progression, which indicates the complex role of connexin 43 in tumor evolution. Since gap junctions play a role in carcinogenesis, invasion, and metastasis of malignant cells, further studies should clarify whether connexin 43 can be used as a diagnostic biomarker.
Collapse
|
12
|
The Potential Impact of Connexin 43 Expression on Bcl-2 Protein Level and Taxane Sensitivity in Head and Neck Cancers-In Vitro Studies. Cancers (Basel) 2019; 11:cancers11121848. [PMID: 31766723 PMCID: PMC6966683 DOI: 10.3390/cancers11121848] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 11/08/2019] [Accepted: 11/20/2019] [Indexed: 12/20/2022] Open
Abstract
The poor prognosis of head and neck squamous cell carcinoma (HNSCC) is partly due to the lack of reliable predictive markers. Connexin 43 (Cx43) protein and its cell-communication channels have been assigned tumor suppressor functions while the anti-apoptotic Bcl-2 (B-cell lymphoma-2) protein has been associated with negative prognostic significance in cancer. This study aimed to test the role of Cx43 protein on Bcl-2 expression, tumor progression and response to taxane-based treatment in HNSCC. Human papillomavirus (HPV) negative HNSCC cell lines were tested for paclitaxel sensitivity through measuring apoptosis induction, cell viability and changes in Cx43 and Bcl-2 levels using flow cytometry, cell viability assay, immunocytochemistry and western blot. Inhibition of Cx43 expression using siRNA increased Bcl-2 protein levels in SCC25 (tongue squamous cell carcinoma) cells, while forced Cx43 expression reduced Bcl-2 levels and supported paclitaxel cytotoxicity in FaDu (hypopharynx squamous cell carcinoma) cells. In vitro results were in line with protein expression and clinicopathological features tested in tissue microarray samples of HNSCC patients. Our data demonstrate that elevated Cx43 and reduced Bcl-2 levels may indicate HNSCC sensitivity to taxane-based treatments. On the contrary, silencing of the Cx43 gene GJA1 (gap junction protein alpha-1) can result in increased Bcl-2 expression and reduced paclitaxel efficiency. Clinical tumor-based analysis also confirmed the inverse correlation between Cx43 and Bcl-2 expression.
Collapse
|
13
|
Aasen T, Leithe E, Graham SV, Kameritsch P, Mayán MD, Mesnil M, Pogoda K, Tabernero A. Connexins in cancer: bridging the gap to the clinic. Oncogene 2019; 38:4429-4451. [PMID: 30814684 PMCID: PMC6555763 DOI: 10.1038/s41388-019-0741-6] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/26/2019] [Accepted: 01/26/2019] [Indexed: 02/08/2023]
Abstract
Gap junctions comprise arrays of intercellular channels formed by connexin proteins and provide for the direct communication between adjacent cells. This type of intercellular communication permits the coordination of cellular activities and plays key roles in the control of cell growth and differentiation and in the maintenance of tissue homoeostasis. After more than 50 years, deciphering the links among connexins, gap junctions and cancer, researchers are now beginning to translate this knowledge to the clinic. The emergence of new strategies for connexin targeting, combined with an improved understanding of the molecular bases underlying the dysregulation of connexins during cancer development, offers novel opportunities for clinical applications. However, different connexin isoforms have diverse channel-dependent and -independent functions that are tissue and stage specific. This can elicit both pro- and anti-tumorigenic effects that engender significant challenges in the path towards personalised medicine. Here, we review the current understanding of the role of connexins and gap junctions in cancer, with particular focus on the recent progress made in determining their prognostic and therapeutic potential.
Collapse
Affiliation(s)
- Trond Aasen
- Translational Molecular Pathology, Vall d'Hebron Institute of Research (VHIR), Autonomous University of Barcelona, CIBERONC, Barcelona, Spain.
| | - Edward Leithe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital and K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Sheila V Graham
- MRC-University of Glasgow Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Petra Kameritsch
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, München, Germany
| | - María D Mayán
- CellCOM Research Group, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), University of A Coruña, A Coruña, Spain
| | - Marc Mesnil
- STIM Laboratory, Faculté des Sciences Fondamentales et Appliquées, Université de Poitiers, Poitiers, France
| | - Kristin Pogoda
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität München and Munich University Hospital, München, Germany
| | - Arantxa Tabernero
- Departamento de Bioquímica y Biología Molecular, Instituto de Neurociencias de Castilla y León (INCYL), Universidad de Salamanca, Salamanca, Spain.
| |
Collapse
|
14
|
Connexin 43 (Cx43) in cancer: Implications for therapeutic approaches via gap junctions. Cancer Lett 2018; 442:439-444. [PMID: 30472182 DOI: 10.1016/j.canlet.2018.10.043] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 09/30/2018] [Accepted: 10/25/2018] [Indexed: 01/11/2023]
Abstract
Gap junctions are membrane channels found in all cells of the human body that are essential to cellular physiology. Gap junctions are formed from connexin proteins and are responsible for transfer of biologically active molecules, metabolites, and salts between neighboring cells or cells and their extracellular environment. Over the last few years, aberrant connexin 43 (Cx43) expression has been associated with cancer recurrence, metastatic spread, and poor survival. Here we provide an overview of the general structure and function of gap junctions and review their roles in different cancer types. We discuss new therapeutic approaches targeting Cx43 and potential new ways of exploiting gap junction transfer for drug delivery and anti-cancer treatment. The permeability of Cx43 channels to small molecules and macromolecules makes them highly attractive targets for delivering drugs directly into the cytoplasm. Cancer cells overexpressing Cx43 may be more permeable and sensitive to chemotherapeutics. Because Cx43 can either act as a tumor suppressor or oncogene, biomarker analysis and a better understanding of how Cx43 contextually mediates cancer phenotypes will be required to develop clinically viable Cx43-based therapies.
Collapse
|
15
|
Gleisner MA, Navarrete M, Hofmann F, Salazar-Onfray F, Tittarelli A. Mind the Gaps in Tumor Immunity: Impact of Connexin-Mediated Intercellular Connections. Front Immunol 2017; 8:1067. [PMID: 28919895 PMCID: PMC5585150 DOI: 10.3389/fimmu.2017.01067] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 08/16/2017] [Indexed: 12/22/2022] Open
Abstract
Gap junctions (GJs)-mediated intercellular communications (GJICs) are connexin (Cx)-formed plasma membrane channels that allow for the passage of small molecules between adjacent cells, and are involved in several physiopathological processes, including immune responses against cancer. In general, tumor cells are poorly coupled through GJs, mainly due to low Cx expression or reduced channel activity, suggesting that Cxs may have tumor suppressor roles. However, more recent data indicate that Cxs and/or GJICs may also in some cases promote tumor progression. This dual role of Cx channels in tumor outcome may be due, at least partially, to the fact that GJs not only interconnect cells from the same type, such as cancer cells, but also promote the intercellular communication of tumor cells with different types of cells from their microenvironment, and such diverse intercellular interactions have distinctive impact on tumor development. For example, whereas GJ-mediated interactions among tumor cells and microglia have been implicated in promotion of tumor growth, tumor cells delivery to dendritic cells of antigenic peptides through GJs have been associated with enhanced immune-mediated tumor elimination. In this review, we provide an updated overview on the role of GJICs in tumor immunity, focusing on the pro-tumor and antitumor effect of GJs occurring among tumor and immune cells. Accumulated data suggest that GJICs may act as tumor suppressors or enhancers depending on whether tumor cells interact predominantly with antitumor immune cells or with stromal cells. The complex modulation of immune-tumor cell GJICs should be taken into consideration in order to potentiate current cancer immunotherapies.
Collapse
Affiliation(s)
- María Alejandra Gleisner
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Faculty of Medicine, Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Mariela Navarrete
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Faculty of Medicine, Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Francisca Hofmann
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Faculty of Medicine, Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Faculty of Medicine, Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Andrés Tittarelli
- Disciplinary Program of Immunology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Faculty of Medicine, Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| |
Collapse
|
16
|
Abstract
Fifty years ago, tumour cells were found to lack electrical coupling, leading to the hypothesis that loss of direct intercellular communication is commonly associated with cancer onset and progression. Subsequent studies linked this phenomenon to gap junctions composed of connexin proteins. Although many studies support the notion that connexins are tumour suppressors, recent evidence suggests that, in some tumour types, they may facilitate specific stages of tumour progression through both junctional and non-junctional signalling pathways. This Timeline article highlights the milestones connecting gap junctions to cancer, and underscores important unanswered questions, controversies and therapeutic opportunities in the field.
Collapse
Affiliation(s)
- Trond Aasen
- (Co-corresponding authors) Correspondence to
T.A. () and D.W.L.
()
| | - Marc Mesnil
- STIM Laboratory ERL 7368 CNRS - Faculté des Sciences
Fondamentales et Appliquées, Université de Poitiers, Poitiers,
France
| | - Christian C. Naus
- Department of Cellular and Physiological Sciences, The Life
Sciences Institute, University of British Columbia, Vancouver, British
Columbia, Canada
| | - Paul D. Lampe
- Translational Research Program, Fred Hutchinson Cancer Research
Center, Seattle, United States
| | - Dale W. Laird
- (Co-corresponding authors) Correspondence to
T.A. () and D.W.L.
()
| |
Collapse
|