1
|
Zhao-Wei R, Xue-Feng J, Gao-Tian X, Jin-Ling C, Jun LI, Shu-Ying LV. Study of the role of transmembrane emp24 domain-containing protein 2 in oral squamous cell carcinoma. J Appl Oral Sci 2025; 33:e20240305. [PMID: 39879476 PMCID: PMC11816950 DOI: 10.1590/1678-7757-2024-0305] [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: 07/24/2024] [Accepted: 11/11/2024] [Indexed: 01/31/2025] Open
Abstract
OBJECTIVE This study aimed to investigate the role of transmembrane emp24 domain-containing protein 2 (TMED2) in oral squamous cell carcinoma (OSCC). METHODOLOGY A bioinformatics analysis was first conducted to explore TMED2 expression in OSCC and its relation with overall survival. The analysis results were further verified by assessing TMED2 expression levels in human normal oral keratinocyte cells and human OSCC cell lines using quantitative real-time polymerase chain reaction and the Western blot. Finally, the effects of TMED2 knockdown and overexpression on the expression levels of TMED2, ADP-ribosylation factor 1, extracellular signal-regulated kinase ½, and phospho-extracellular signal-regulated kinase ½ proteins were examined in cells using the Western blot. RESULTS The GEPIA2 database showed that OSCC tissues expressed more TMED2 than normal tissues. At the cellular level, TMED2 expression significantly increased in SCC-4, HSC-3, and CAL-27 cells than in human normal oral keratinocyte cells. TMED2 knockdown reduced cell proliferation, increased the apoptosis rate in SCC-4 cells, and led to a higher proportion of cells in the G0/G1 phase and a lower proportion in the S phase. CONCLUSION TMED2 may promote OSCC cell proliferation and inhibit apoptosis, potentially by activation of the ADP-ribosylation factor 1/ extracellular signal-regulated kinase ½ signaling pathway.
Collapse
Affiliation(s)
- Ruan Zhao-Wei
- Ningde Hospital Affiliated to Ningde Normal University, Department of Stomatology, Fujian, China
| | - Jiao Xue-Feng
- Ningde Hospital Affiliated to Ningde Normal University, Department of Stomatology, Fujian, China
| | - Xiao Gao-Tian
- Ningde Hospital Affiliated to Ningde Normal University, Department of Stomatology, Fujian, China
| | - Chen Jin-Ling
- Ningde Hospital Affiliated to Ningde Normal University, Department of Stomatology, Fujian, China
| | - L I Jun
- The First Affiliated Hospital of Fujian Medical Universtity, Stomatology Center, Fujian, China
| | - L V Shu-Ying
- Ningde Hospital Affiliated to Ningde Normal University, Department of Stomatology, Fujian, China
| |
Collapse
|
2
|
Jiang K, Xu LZ, Cheng F, Ning JZ. COL10A1 Facilitates Prostate Cancer Progression by Interacting With INHBA to Activate the PI3K/AKT Pathway. J Cell Mol Med 2024; 28:e70249. [PMID: 39656597 DOI: 10.1111/jcmm.70249] [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: 08/21/2024] [Revised: 10/26/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024] Open
Abstract
Prostate cancer (PCa) constitutes a highly common and lethal disease that impacts males globally. However, the specific molecular pathways responsible for its development are still unknown. Therefore, revealing the molecular regulators that contributed to the progression of PCa is pivotal for developing unique management strategies. Through comprehensive bioinformatics analysis of multiple public gene databases, we thoroughly investigated COL10A1 expression level, clinical significance, co-expressed genes and signalling pathways in PCa. COL10A1 and INHBA expression level was assessed in clinical PCa specimens using RT-qPCR, Western blotting and immunohistochemistry. A combination of experimental techniques, including CCK-8 assay, colony formation, flow cytometry, Transwell, wound-healing, immunoprecipitation assays and rescue study, was utilised to examine the fundamental molecular pathways of COL10A1's action across PCa. The COL10A1 expression was significantly elevated in PCa, and its upregulation has been connected with tumour aggressiveness and a weak predictive outcome in subjects. The current investigation revealed that regulation of COL10A1 expression, either by upregulation or downregulation, resulted in sequential augmentation or suppression of PCa cell progression, migration and invasion. Mechanistically, COL10A1 was manifested to directly interact with INHBA and facilitate PI3K and AKT phosphorylation pathways within PCa cells and mouse models. The results of our study offer new perspectives on the tumorigenic role of COL10A1 in PCa and its interactions with INHBA may play important roles in PCa progression.
Collapse
Affiliation(s)
- Kun Jiang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Li-Zhe Xu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Jin-Zhuo Ning
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| |
Collapse
|
3
|
Fang YX, Lu EQ, Xu E, Zhang YY, Zhu M. Arf1 promotes porcine intestinal epithelial cell proliferation via the mTORC1 signaling pathway. In Vitro Cell Dev Biol Anim 2024; 60:1009-1020. [PMID: 39093368 DOI: 10.1007/s11626-024-00942-y] [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: 02/22/2024] [Accepted: 06/20/2024] [Indexed: 08/04/2024]
Abstract
The promotion of gut health, a pervasive problem in modern animal husbandry, positively affects organismal health, productivity, and economics. Porcine intestinal epithelial cells (IPEC-J2) continuously proliferate to maintain intestinal homeostasis, including barrier, immune, and absorptive functions. Gut homeostasis is fundamental to organismal health. ADP-ribosylation factor 1 (Arf1), a small GTPase, plays a crucial role in coordinating mTORC1 in response to nutrients, especially amino acid availability in the gut. mTORC1 is the central hub of proliferation. Thus, it seems likely that Arf1 promotes IPEC-J2 cell proliferation. However, the exact role of Arf1 in the porcine gut remains unclear. Therefore, we evaluated the functional role and possible mechanisms of Arf1 in the porcine intestine through Arf1 overexpression and knockdown in IPEC-J2 cells. Arf1 overexpression and knockdown significantly enhanced and inhibited, respectively, IPEC-J2 cell viability, and PCNA expression varied with Arf1 expression. Moreover, the proportion of Ki67-positive cells was significantly greater in the Arf1-overexpressing group than in the control group. These results suggest that Arf1 improves IPEC-J2 cell proliferation. The underlying mechanism was explored by Western blotting. Arf1 overexpression and knockdown significantly enhanced and suppressed, respectively, the levels of p-S6K1 and p-RPS6, which are key downstream targets of the mTORC1 signaling pathway. Collectively, our findings reveal the role of the Arf1-mTORC1 axis in IPEC-J2 cell proliferation and its potential function in regulating intestinal homeostasis and health.
Collapse
Affiliation(s)
- Yong-Xia Fang
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China
- Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China
| | - En-Qing Lu
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China
- Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China
| | - E Xu
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China
- Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China
| | - Yi-Yu Zhang
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China
- Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China
| | - Min Zhu
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China.
- Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
4
|
Wang Y, Li Q, Ding Y, Luo C, Yang J, Wang N, Jiang N, Yao T, Wang G, Shi G, Hou SX. Novel Arf1 Inhibitors Drive Cancer Stem Cell Aging and Potentiate Anti-Tumor Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404442. [PMID: 39225354 PMCID: PMC11497069 DOI: 10.1002/advs.202404442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/01/2024] [Indexed: 09/04/2024]
Abstract
The small G protein Arf1 has been identified as playing a selective role in supporting cancer stem cells (CSCs), making it an attractive target for cancer therapy. However, the current Arf1 inhibitors have limited translational potential due to their high toxicity and low specificity. In this study, two new potent small-molecule inhibitors of Arf1, identified as DU101 and DU102, for cancer therapy are introduced. Preclinical tumor models demonstrate that these inhibitors triggered a cascade of aging in CSCs and enhance anti-tumor immunity in mouse cancer and PDX models. Through single-cell sequencing, the remodeling of the tumor immune microenvironment induced by these new Arf1 inhibitors is analyzed and an increase in tumor-associated CD8+ CD4+ double-positive T (DPT) cells is identified. These DPT cells exhibit superior features of active CD8 single-positive T cells and a higher percentage of TCF1+PD-1+, characteristic of stem-like T cells. The frequency of tumor-infiltrating stem-like DPT cells correlates with better disease-free survival (DFS) in cancer patients, indicating that these inhibitors may offer a novel cancer immunotherapy strategy by converting the cold tumor immune microenvironment into a hot one, thus expanding the potential for immunotherapy in cancer patients.
Collapse
Affiliation(s)
- Yuetong Wang
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyHuman Phenome InstituteDepartment of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan HospitalFudan UniversityShanghai200438China
| | - Qiaoming Li
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyHuman Phenome InstituteDepartment of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan HospitalFudan UniversityShanghai200438China
| | - Yahui Ding
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyHuman Phenome InstituteDepartment of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan HospitalFudan UniversityShanghai200438China
| | - Chenfei Luo
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyHuman Phenome InstituteDepartment of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan HospitalFudan UniversityShanghai200438China
| | - Jun Yang
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyHuman Phenome InstituteDepartment of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan HospitalFudan UniversityShanghai200438China
| | - Na Wang
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyHuman Phenome InstituteDepartment of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan HospitalFudan UniversityShanghai200438China
| | - Ning Jiang
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyHuman Phenome InstituteDepartment of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan HospitalFudan UniversityShanghai200438China
| | - Tiange Yao
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyHuman Phenome InstituteDepartment of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan HospitalFudan UniversityShanghai200438China
| | - Guohao Wang
- The Basic Research LaboratoryCenter for Cancer ResearchNational Cancer Institute at FrederickNational Institutes of HealthFrederickMD21702USA
| | - Guoming Shi
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyHuman Phenome InstituteDepartment of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan HospitalFudan UniversityShanghai200438China
| | - Steven X. Hou
- Department of Cell and Developmental Biology at School of Life SciencesState Key Laboratory of Genetic EngineeringInstitute of Metabolism and Integrative BiologyHuman Phenome InstituteDepartment of Liver Surgery and Transplantation of Liver Cancer Institute at Zhongshan HospitalFudan UniversityShanghai200438China
- Leading Contact
| |
Collapse
|
5
|
Mu Q, Wang X, Huang K, Xia B, Bi S, Kong Y. THUMPD3-AS1 inhibits ovarian cancer cell apoptosis through the miR-320d/ARF1 axis. FASEB J 2024; 38:e23772. [PMID: 38963337 DOI: 10.1096/fj.202302475rrr] [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: 12/13/2023] [Revised: 06/06/2024] [Accepted: 06/17/2024] [Indexed: 07/05/2024]
Abstract
Ovarian cancer is one of the most common gynecologic malignancies that has a poor prognosis. THUMPD3-AS1 is an oncogenic long noncoding RNA (lncRNA) in several cancers. Moreover, miR-320d is downregulated and inhibited proliferation in ovarian cancer cells, whereas ARF1 was upregulated and promoted the malignant progression in epithelial ovarian cancer. Nevertheless, the role of THUMPD3-AS1 in ovarian cancer and the underlying mechanism has yet to be elucidated. Human normal ovarian epithelial cells (IOSE80) and ovarian cancer cell lines (CAVO3, A2780, SKOV3, OVCAR3, and HEY) were adopted for in vitro experiments. The functional roles of THUMPD3-AS1 in cell viability and apoptosis were determined using CCK-8, flow cytometry, and TUNEL assays. Western blot was performed to assess the protein levels of ARF1, Bax, Bcl-2, and caspase 3, whereas RT-qPCR was applied to measure ARF1 mRNA, THUMPD3-AS1, and miR-320d levels. The targeting relationship between miR-320d and THUMPD3-AS1 or ARF1 was validated with dual luciferase assay. THUMPD3-AS1 and ARF1 were highly expressed in ovarian cancer cells, whereas miR-320d level was lowly expressed. THUMPD3-AS1 knockdown was able to repress cell viability and accelerate apoptosis of OVCAR3 and SKOV3 cells. Also, THUMPD3-AS1 acted as a sponge of miR-320d, preventing the degradation of ARF1. MiR-320d downregulation reversed the tumor suppressive function induced by THUMPD3-AS1 depletion. Additionally, miR-320d overexpression inhibited ovarian cancer cell viability and accelerated apoptosis, which was overturned by overexpression of ARF1. THUMPD3-AS1 inhibited ovarian cancer cell apoptosis by modulation of miR-320d/ARF1 axis. The discoveries might provide a prospective target for ovarian cancer treatment.
Collapse
Affiliation(s)
- Qingling Mu
- Department of Gynecology and Obstetrics, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Xin Wang
- Department of Gynecology and Obstetrics, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Kui Huang
- Department of Obstetrics and Gynecology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Baoguo Xia
- Department of Gynecology and Obstetrics, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Shuna Bi
- Department of Gynecology and Obstetrics, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Yujie Kong
- Department of Gynecology and Obstetrics, Qingdao Municipal Hospital, Qingdao, Shandong, China
| |
Collapse
|
6
|
Wang N, Yao T, Luo C, Sun L, Wang Y, Hou SX. Blockade of Arf1-mediated lipid metabolism in cancers promotes tumor infiltration of cytotoxic T cells via the LPE-PPARγ-NF-κB-CCL5 pathway. LIFE METABOLISM 2023; 2:load036. [PMID: 39872623 PMCID: PMC11749100 DOI: 10.1093/lifemeta/load036] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 01/30/2025]
Abstract
Tumor immunotherapy has achieved breakthroughs in a variety of tumors. However, the systemic absence of T cells in tumors and immunosuppressive tumor microenvironment so far limits the efficacy of immunotherapy to a small population of patients. Therefore, novel agents to increase T-cell tumor infiltration are urgently needed in the clinic. We recently found that inhibition of the ADP-ribosylation factor 1 (Arf1)-mediated lipid metabolism not only kills cancer stem cells (CSCs) but also elicits an anti-tumor immune response. In this study, we revealed a mechanism that targeting Arf1 promotes the infiltration of cytotoxic T lymphocytes (CTLs) into tumors through the C-C chemokine ligand 5 (CCL5)- C-C chemokine receptor type 5 (CCR5) pathway. We found that blockage of Arf1 induces the production of the unsaturated fatty acid (PE 18:1) that binds and sequestrates peroxisome proliferator--activated receptor-γ (PPARγ) from the PPARγ-nuclear factor-κB (NF-κB) cytoplasmic complex. The released NF-κB was then phosphorylated and translocated into the nucleus to regulate the transcription of chemokine CCL5. CCL5 promoted infiltration of CTLs for tumor regression. Furthermore, the combination of the Arf1 inhibitor and programmed cell death protein 1 (PD-1) blockade induced an even stronger anti-tumor immunity. Therefore, targeting Arf1 represents a novel anti-tumor immune approach by provoking T-cell tumor infiltration and may provide a new strategy for tumor immunotherapy.
Collapse
Affiliation(s)
- Na Wang
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Tiange Yao
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Chenfei Luo
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Ling Sun
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Yuetong Wang
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Steven X Hou
- Department of Cell and Developmental Biology at School of Life Sciences, State Key Laboratory of Genetic Engineering, Institute of Metabolism and Integrative Biology, Human Phenome Institute, Fudan University, Shanghai 200438, China
| |
Collapse
|
7
|
Ballari MS, O J Porta E, Zalazar EA, Etichetti CMB, Padrón JM, Girardini JE, Labadie GR. Lipophilic modification of salirasib modulates the antiproliferative and antimigratory activity. Bioorg Med Chem 2023; 92:117417. [PMID: 37531922 DOI: 10.1016/j.bmc.2023.117417] [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: 05/30/2023] [Revised: 07/11/2023] [Accepted: 07/16/2023] [Indexed: 08/04/2023]
Abstract
Salirasib, or farnesylthiosalicylic acid (FTS), is a salicylic acid derivative with demonstrated antineoplastic activity. While designed as a competitor of the substrate S-farnesyl cysteine on Ras, it is a potent competitive inhibitor of isoprenylcysteine carboxymethyl transferase. In this study, the antiproliferative activity on six different solid tumor cell lines was evaluated with a series of lipophilic thioether modified salirasib analogues, including those with or without a 1,2,3-triazole linker. A combination of bioassay, cheminformatics, docking, and in silico ADME-Tox was also performed. SAR analysis that analogues with three or more isoprene units or a long aliphatic chain exhibited the most potent activity. Furthermore, three compounds display superior antiproliferative activity than salirasib and similar potency compared to control anticancer drugs across all tested solid tumor cell lines. In addition, the behavior of the collection on migration and invasion, a key process in tumor metastasis, was also studied. Three analogues with specific antimigratory activity were identified with differential structural features being interesting starting points on the development of new antimetastatic agents. The antiproliferative and antimigratory effects observed suggest that modifying the thiol aliphatic/prenyl substituents can modulate the activity.
Collapse
Affiliation(s)
- María Sol Ballari
- Instituto de Química Rosario (IQUIR), Universidad Nacional de Rosario-CONICET, Suipacha 531 S2002LRK, Rosario, Argentina
| | - Exequiel O J Porta
- Instituto de Química Rosario (IQUIR), Universidad Nacional de Rosario-CONICET, Suipacha 531 S2002LRK, Rosario, Argentina
| | - Evelyn Arel Zalazar
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Suipacha 590 S2000LRJ, Rosario, Argentina
| | - Carla M Borini Etichetti
- Instituto de Fisiología Experimental de Rosario (IFISE-CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Suipacha 531 2000 Rosario, Argentina
| | - José M Padrón
- BioLab, Instituto Universitario de Bio-Orgánica "Antonio González" (IUBO-AG), Universidad de La Laguna, Apartado 456 E-38071, La Laguna, Spain.
| | - Javier E Girardini
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET). Suipacha 590 S2000LRJ, Rosario, Argentina.
| | - Guillermo R Labadie
- Instituto de Química Rosario (IQUIR), Universidad Nacional de Rosario-CONICET, Suipacha 531 S2002LRK, Rosario, Argentina; Departamento de Química Orgánica, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina.
| |
Collapse
|
8
|
Hu HF, Gao GB, He X, Li YY, Li YJ, Li B, Pan Y, Wang Y, He QY. Targeting ARF1-IQGAP1 interaction to suppress colorectal cancer metastasis and vemurafenib resistance. J Adv Res 2023; 51:135-147. [PMID: 36396045 PMCID: PMC10491971 DOI: 10.1016/j.jare.2022.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/11/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Acquired resistance to BRAF inhibitor vemurafenib is frequently observed in metastatic colorectal cancer (CRC), and it is a thorny issue that results in treatment failure. As adaptive responses for vemurafenib treatment, a series of cellular bypasses are response for the adaptive feedback reactivation of ERK signaling, which warrant further investigation. OBJECTIVES We identified ARF1 (ADP-ribosylation factor 1) as a novel regulator of both vemurafenib resistance and cancer metastasis, its molecular mechanism and potential inhibitor were investigated in this study. METHODS DIA-based quantitative proteomics and RNA-seq were performed to systematic analyze the profiling of vemurafenib-resistant RKO cells (RKO-VR) and highly invasive RKO cells (RKO-I8), respectively. Co‑immunoprecipitation assay was performed to detect the interaction of ARF1 and IQGAP1 (IQ-domain GTPase activating protein 1). An ELISA-based drug screen system on FDA-approved drug library was established to screen the compounds against the interaction of ARF1-IQGAP1.The biological functions of ARF1 and LY2835219 were determined by transwell, western blotting, Annexin V-FITC/PI staining and in vivo experimental metastasis assays. RESULTS We found that ARF1 strongly interacted with IQGAP1 to activate ERK signaling in VR and I8 CRC cells. Deletion of IQGAP1 or inactivation of ARF1 (ARF-T48S) restored the invasive ability induced by ARF1. As ARF1-IQGAP1 interaction is essential for ERK activation, we screened LY2835219 as novel inhibitor of ARF1-IQGAP1 interaction, which inactivated ERK signaling and suppressed CRC metastasis and vemurafenib-resistance in vitro and in vivo with no observed side effect. Furthermore, LY2835219 in combined treatment with vemurafenib exerted significantly inhibitory effect on ARF1-mediated cancer metastasis than used independently. CONCLUSION This study uncovers that ARF1-IQGAP1 interaction-mediated ERK signaling reactivation is critical for vemurafenib resistance and cancer metastasis, and that LY2835219 is a promising therapeutic agent for CRC both as a single agent and in combination with vemurafenib.
Collapse
Affiliation(s)
- Hui-Fang Hu
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China; MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Gui-Bin Gao
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xuan He
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yu-Ying Li
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yang-Jia Li
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Bin Li
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - YunLong Pan
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China.
| | - Yang Wang
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| | - Qing-Yu He
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China; MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
9
|
GTPase Pathways in Health and Diseases. Cells 2022; 11:cells11244055. [PMID: 36552819 PMCID: PMC9777353 DOI: 10.3390/cells11244055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
GTPases, the molecular switches toggling between an inactive GDP-bound state and an active GTP-bound state, play a pivotal role in controlling complex cellular processes (e [...].
Collapse
|
10
|
Lang L, Loveless R, Dou J, Lam T, Chen A, Wang F, Sun L, Juarez J, Qin ZS, Saba NF, Shay C, Teng Y. ATAD3A mediates activation of RAS-independent mitochondrial ERK1/2 signaling, favoring head and neck cancer development. J Exp Clin Cancer Res 2022; 41:43. [PMID: 35093151 PMCID: PMC8800319 DOI: 10.1186/s13046-022-02274-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/25/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Targeting mitochondrial oncoproteins presents a new concept in the development of effective cancer therapeutics. ATAD3A is a nuclear-encoded mitochondrial enzyme contributing to mitochondrial dynamics, cholesterol metabolism, and signal transduction. However, its impact and underlying regulatory mechanisms in cancers remain ill-defined. METHODS We used head and neck squamous cell carcinoma (HNSCC) as a research platform and achieved gene depletion by lentiviral shRNA and CRISPR/Cas9. Molecular alterations were examined by RNA-sequencing, phospho-kinase profiling, Western blotting, RT-qPCR, immunohistochemistry, and immunoprecipitation. Cancer cell growth was assessed by MTT, colony formation, soft agar, and 3D cultures. The therapeutic efficacy in tumor development was evaluated in orthotopic tongue tumor NSG mice. RESULTS ATAD3A is highly expressed in HNSCC tissues and cell lines. Loss of ATAD3A expression suppresses HNSCC cell growth and elicits tumor regression in orthotopic tumor-bearing mice, whereas gain of ATAD3A expression produces the opposite effects. From a mechanistic perspective, the tumor suppression induced by the overexpression of the Walker A dead mutant of ATAD3A (K358) produces a potent dominant-negative effect due to defective ATP-binding. Moreover, ATAD3A binds to ERK1/2 in the mitochondria of HNSCC cells in the presence of VDAC1, and this interaction is essential for the activation of mitochondrial ERK1/2 signaling. Most importantly, the ATAD3A-ERK1/2 signaling axis drives HNSCC development in a RAS-independent fashion and, thus, tumor suppression is more effectively achieved when ATAD3A knockout is combined with RAS inhibitor treatment. CONCLUSIONS These findings highlight the novel function of ATAD3A in regulating mitochondrial ERK1/2 activation that favors HNSCC development. Combined targeting of ATAD3A and RAS signaling may potentiate anticancer activity for HNSCC therapeutics.
Collapse
Affiliation(s)
- Liwei Lang
- Department of Oral Biology and Diagnostic Sciences, Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA
| | - Reid Loveless
- Department of Oral Biology and Diagnostic Sciences, Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA
| | - Juan Dou
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 201 Dowman Dr, Atlanta, GA, 30322, USA
| | - Tiffany Lam
- Department of Oral Biology and Diagnostic Sciences, Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA
| | - Alex Chen
- Department of Oral Biology and Diagnostic Sciences, Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA
| | - Fang Wang
- Department of Oral Biology and Diagnostic Sciences, Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA
| | - Li Sun
- Department of Oral Biology and Diagnostic Sciences, Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA
| | - Jakeline Juarez
- Department of Oral Biology and Diagnostic Sciences, Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA
| | - Zhaohui Steve Qin
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, 30322, USA
| | - Nabil F Saba
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 201 Dowman Dr, Atlanta, GA, 30322, USA
| | - Chloe Shay
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory, University, Atlanta, GA, 30322, USA
| | - Yong Teng
- Department of Oral Biology and Diagnostic Sciences, Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA.
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 201 Dowman Dr, Atlanta, GA, 30322, USA.
| |
Collapse
|
11
|
Endolysosomal ion channel MCOLN2 (Mucolipin-2) promotes prostate cancer progression via IL-1β/NF-κB pathway. Br J Cancer 2021; 125:1420-1431. [PMID: 34548638 PMCID: PMC8575885 DOI: 10.1038/s41416-021-01537-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/13/2021] [Accepted: 08/20/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Prostate cancer (Pca) is the most common cancer type among males worldwide. Dysregulation of Ca2+ signaling plays important roles during Pca progression. However, there is lack of information about the role of endolysosomal Ca2+ -permeable channels in Pca progression. METHODS The expression pattern of MCOLN2 was studied by immunohistochemistry and western blot. Cell viability assay, transwell assay and in vivo tumorigenesis were performed to evaluate the functional role of MCOLN2. Downstream targets of MCOLN2 were investigated by cytokine array, enzyme-linked immunosorbent assay, Ca2+ release experiments and luciferase reporter assays. RESULTS We report that MCOLN2 expression is significantly elevated in Pca tissues, and associated with poor prognosis. Overexpression of MCOLN2 promoted Pca cells proliferation, migration and invasion. Importantly, knockdown of MCOLN2 inhibited Pca xenograft tumor growth and bone lesion development in vivo. In addition, MCOLN2 promoted the production and release of IL-1β. Moreover, luciferase reporter assay and western blot revealed that MCOLN2 promoted Pca development by regulating the IL-1β/NF-κB pathway. CONCLUSION In summary, MCOLN2 is crucially involved in Pca progression. Mechanistically, MCOLN2 regulates Pca progression via IL-1β/NF-κB pathway. Our study highlights an intriguing possibility of targeting MCOLN2 as potential therapeutic strategy in Pca treatment.
Collapse
|
12
|
You KS, Yi YW, Cho J, Park JS, Seong YS. Potentiating Therapeutic Effects of Epidermal Growth Factor Receptor Inhibition in Triple-Negative Breast Cancer. Pharmaceuticals (Basel) 2021; 14:589. [PMID: 34207383 PMCID: PMC8233743 DOI: 10.3390/ph14060589] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a subset of breast cancer with aggressive characteristics and few therapeutic options. The lack of an appropriate therapeutic target is a challenging issue in treating TNBC. Although a high level expression of epidermal growth factor receptor (EGFR) has been associated with a poor prognosis among patients with TNBC, targeted anti-EGFR therapies have demonstrated limited efficacy for TNBC treatment in both clinical and preclinical settings. However, with the advantage of a number of clinically approved EGFR inhibitors (EGFRis), combination strategies have been explored as a promising approach to overcome the intrinsic resistance of TNBC to EGFRis. In this review, we analyzed the literature on the combination of EGFRis with other molecularly targeted therapeutics or conventional chemotherapeutics to understand the current knowledge and to provide potential therapeutic options for TNBC treatment.
Collapse
Affiliation(s)
- Kyu Sic You
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
| | - Yong Weon Yi
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeonghee Cho
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeong-Soo Park
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
| | - Yeon-Sun Seong
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| |
Collapse
|
13
|
Hu HF, Xu WW, Li YJ, He Y, Zhang WX, Liao L, Zhang QH, Han L, Yin XF, Zhao XX, Pan YL, Li B, He QY. Anti-allergic drug azelastine suppresses colon tumorigenesis by directly targeting ARF1 to inhibit IQGAP1-ERK-Drp1-mediated mitochondrial fission. Am J Cancer Res 2021; 11:1828-1844. [PMID: 33408784 PMCID: PMC7778598 DOI: 10.7150/thno.48698] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
This study aimed to screen novel anticancer strategies from FDA-approved non-cancer drugs and identify potential biomarkers and therapeutic targets for colorectal cancer (CRC). Methods: A library consisting of 1056 FDA-approved drugs was screened for anticancer agents. WST-1, colony-formation, flow cytometry, and tumor xenograft assays were used to determine the anticancer effect of azelastine. Quantitative proteomics, confocal imaging, Western blotting and JC-1 assays were performed to examine the effects on mitochondrial pathways. The target protein of azelastine was analyzed and confirmed by DARTS, WST-1, Biacore and tumor xenograft assays. Immunohistochemistry, gain- and loss-of-function experiments, WST-1, colony-formation, immunoprecipitation, and tumor xenograft assays were used to examine the functional and clinical significance of ARF1 in colon tumorigenesis. Results: Azelastine, a current anti-allergic drug, was found to exert a significant inhibitory effect on CRC cell proliferation in vitro and in vivo, but not on ARF1-deficient or ARF1-T48S mutant cells. ARF1 was identified as a direct target of azelastine. High ARF1 expression was associated with advanced stages and poor survival of CRC. ARF1 promoted colon tumorigenesis through its interaction with IQGAP1 and subsequent activation of ERK signaling and mitochondrial fission by enhancing the interaction of IQGAP1 with MEK and ERK. Mechanistically, azelastine bound to Thr-48 in ARF1 and repressed its activity, decreasing Drp1 phosphorylation. This, in turn, inhibited mitochondrial fission and suppressed colon tumorigenesis by blocking IQGAP1-ERK signaling. Conclusions: This study provides the first evidence that azelastine may be novel therapeutics for CRC treatment. ARF1 promotes colon tumorigenesis, representing a promising biomarker and therapeutic target in CRC.
Collapse
|
14
|
Cheng J, Lou Y, Jiang K. Downregulation of long non-coding RNA LINC00460 inhibits the proliferation, migration and invasion, and promotes apoptosis of pancreatic cancer cells via modulation of the miR-320b/ARF1 axis. Bioengineered 2020; 12:96-107. [PMID: 33345740 PMCID: PMC8806231 DOI: 10.1080/21655979.2020.1863035] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Pancreatic adenocarcinoma (PAAD) ranks among the most lethal cancers worldwide with high mortality. A marked increase in the level of long non-coding RNA LINC00460 was reported in PAAD patients, in comparison with the healthy controls. However, the underlying mechanisms of the above phenomenon are not yet well understood. Hence, the present study was designed to investigate the molecular mechanism underlying the role of LINC00460 in proliferation, migration and invasion of pancreatic cancer (PC) cells. It was found in our study that LINC00460 knockdown inhibited SW1990 cell proliferation, migration and invasion and promoted its apoptosis. Moreover, miR-320b was targeted straight and its expression was downregulated by LINC00460, whose knockdown led to a reduction in ARF1 expression. Interestingly, miR-320b downregulation partly reversed the effect of LINC00460 knockdown on the proliferation, migration, invasion and apoptosis of SW1990 cells, as well as ARF1expression. In conclusion, LINC00460 knockdown inhibited the proliferation, migration and invasion, and promotes the apoptosis of SW1990 cells via modulation of the miR-320b/ARF1 axis. Thus, LINC00460 can be perceived as a promising target in the treatment of PAAD.
Collapse
Affiliation(s)
- Jian Cheng
- Department of Hepatobiliary, Pancreatic and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College , Hangzhou City, Zhejiang Province, PR China
| | - Yanghui Lou
- Department of Anesthesiology, Yiwu Maternity and Children Hospital , Yiwu City, Zhejiang Province, PR China
| | - Kai Jiang
- Department of Hepatobiliary, Pancreatic and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College , Hangzhou City, Zhejiang Province, PR China
| |
Collapse
|
15
|
Casalou C, Ferreira A, Barral DC. The Role of ARF Family Proteins and Their Regulators and Effectors in Cancer Progression: A Therapeutic Perspective. Front Cell Dev Biol 2020; 8:217. [PMID: 32426352 PMCID: PMC7212444 DOI: 10.3389/fcell.2020.00217] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/12/2020] [Indexed: 12/13/2022] Open
Abstract
The Adenosine diphosphate-Ribosylation Factor (ARF) family belongs to the RAS superfamily of small GTPases and is involved in a wide variety of physiological processes, such as cell proliferation, motility and differentiation by regulating membrane traffic and associating with the cytoskeleton. Like other members of the RAS superfamily, ARF family proteins are activated by Guanine nucleotide Exchange Factors (GEFs) and inactivated by GTPase-Activating Proteins (GAPs). When active, they bind effectors, which mediate downstream functions. Several studies have reported that cancer cells are able to subvert membrane traffic regulators to enhance migration and invasion. Indeed, members of the ARF family, including ARF-Like (ARL) proteins have been implicated in tumorigenesis and progression of several types of cancer. Here, we review the role of ARF family members, their GEFs/GAPs and effectors in tumorigenesis and cancer progression, highlighting the ones that can have a pro-oncogenic behavior or function as tumor suppressors. Moreover, we propose possible mechanisms and approaches to target these proteins, toward the development of novel therapeutic strategies to impair tumor progression.
Collapse
Affiliation(s)
- Cristina Casalou
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Andreia Ferreira
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Duarte C Barral
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
16
|
Design and Synthesis of Arf1-Targeting γ-Dipeptides as Potential Agents against Head and Neck Squamous Cell Carcinoma. Cells 2020; 9:cells9020286. [PMID: 31991585 PMCID: PMC7072570 DOI: 10.3390/cells9020286] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/20/2020] [Accepted: 01/21/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Head and neck squamous cell carcinoma (HNSCC) is one of the leading causes of cancer-related deaths and calls for new druggable targets. We have previously highlighted the critical role of ADP-ribosylation factor-1 (Arf1) activation in HNSCC. In the present study, we address the question whether targeting Arf1 could be proposed as a valuable strategy against HNSCC. Methods: We rationally designed and synthesized constrained ATC-based (4-amino-(methyl)-1,3-thiazole-5-carboxylic acid) γ-dipeptides to block Arf1 activation. We evaluated the effects of these γ-dipeptides in HNSCC cells: The cell viability was determined in 2D and 3D cell cultures after 72 h treatment and Arf1 protein levels and activity were assessed by GGA3 pull-down and Western blotting assays. Results: Targeting Arf1 offers a valuable strategy to counter HNSCC. Our new Arf1-targeting compounds revealed a strong in vitro cytotoxicity against HNSCC cells, through inhibiting Arf1 activation and its downstream pathways. Conclusions: Arf1-targeting γ-dipeptides developed in this study may represent a promising targeted therapeutic to improve managing the HNSCC disease.
Collapse
|
17
|
Wen Y, Zhang S, Yang J, Guo D. Identification of driver genes regulating immune cell infiltration in cervical cancer by multiple omics integration. Biomed Pharmacother 2019; 120:109546. [PMID: 31675687 DOI: 10.1016/j.biopha.2019.109546] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/04/2019] [Accepted: 10/08/2019] [Indexed: 12/24/2022] Open
Abstract
Cervical cancer (CC) is one of the most common cancers in women. However, copy number alteration (CNA)-driven dysregulated genes and their functions in CC are still rarely investigated. In the present study, we conducted integrative analysis of CNA and gene expression data from The Cancer Genome Atlas (TCGA) cervical cancer to identify dysregulated genes triggered by CNAs. The integration of copy number status and RNA expression revealed 763 amplified and 1,391 deleted genes significantly dysregulated by the CNAs (P-value < 1e-8). Among these CNA genes, five driver genes, including PI3KCA, PI3KCB, DVL3, WWTR1, and ERBB2, exhibited a strong association with immune cell infiltration, suggesting that the pathways that they participate in may be involved in regulating immune cell infiltration. Moreover, we also observed that the genes of immunotherapeutic targets were abundantly expressed in the wild-type samples, suggesting that immunotherapy based on these immunotherapeutic targets may be applied to wild-type samples. In addition, the two CNA driver genes, DVL3 and ERBB2, might be sensitive and resistant biomarkers for examining the tumor's response to chemoradiotherapy, respectively. Particularly, the expression of ERBB2 was also observed to be higher in responders of chemotherapy than non-responders. Furthermore, a subset of CNA genes was identified to predict the prognosis of cervical cancer. In summary, our systematic data analysis of these CNA genes not only improved our understanding of the veiled mechanism behind immune cell infiltration, but also provided the potential clinical application of these CNA genes in cervical cancer.
Collapse
Affiliation(s)
- Yanqi Wen
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China
| | - Silin Zhang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China.
| | - Duanying Guo
- Longgang District People's Hospital of Shenzhen, Shenzhen, China.
| |
Collapse
|
18
|
Zhang YP, Liu KL, Yang Z, Lu BS, Qi JC, Han ZW, Yin YW, Zhang M, Chen DM, Wang XW, Li W, Xin H. The involvement of FBP1 in prostate cancer cell epithelial mesenchymal transition, invasion and metastasis by regulating the MAPK signaling pathway. Cell Cycle 2019; 18:2432-2446. [PMID: 31448674 DOI: 10.1080/15384101.2019.1648956] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Prostate cancer (PCa) is a frequently occurring malignancy in males, and epithelial mesenchymal transition (EMT) plays a critical role in PCa metastasis. Thus, developing biomarkers inhibiting EMT may provide significance for treatment of PCa. Hence, the aim of the current study was to investigate the mechanism by which FBP1 gene silencing influences PCa cell EMT, invasion and metastasis by mediating the MAPK pathway. PCa cell lines exhibiting the highest FBP1 expression were selected and treated with plasmids of siRNA-FBP1 sequence 1 and 2, pcDNA3.1-Flag-FBP1 (over-expression plasmid of FBP1), U0126 (an inhibitor of the ERK signaling pathway) and PD98059 (an inhibitor of the MEK signaling pathway). Cell proliferation, migration and invasion were detected by MTT assay, wound healing assay and Transwell assay, respectively. The mRNA and protein expression of related factors of EMT and MAPK signaling were determined by RT-qPCR and western blot analysis, respectively. Xenograft tumor growth after inoculation of DU145 cells was regularly analyzed in the nude mice. The positive expression of EMT markers was determined by immunohistochemistry. DU-145 and PC-3 cells displaying the highest FBP1 expression were selected for further analysis. The PCa cells treated with siRNA-FBP1 exhibited increased proliferation, migration rate and invasion, in addition to facilitated xenograft tumor growth. Notably, siRNA-FBP1 was identified to accelerate PCa cell EMT by elevating the expression of Vimentin and N-cadherin while diminishing E-cadherin expression via activation of the MAPK signaling pathway. The aforementioned results were reversed in PCa cells treated by pcDNA3.1-Flag-FBP1. Evidence has been provided in this study that FBP1 gene silencing activates the MAPK pathway, which ultimately promotes cell EMT, invasion and metastasis in PCa.
Collapse
Affiliation(s)
- Yan-Ping Zhang
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Kai-Long Liu
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Zhan Yang
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Bao-Sai Lu
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Jin-Chun Qi
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Zhen-Wei Han
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Yue-Wei Yin
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Ming Zhang
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - De-Min Chen
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Xiao-Wei Wang
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Wei Li
- Department of Urology, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| | - Hong Xin
- Department of Obstetrics, The Second Hospital of Hebei Medical University , Shijiazhuang , P.R. China
| |
Collapse
|
19
|
Kolluru V, Tyagi A, Chandrasekaran B, Damodaran C. Profiling of differentially expressed genes in cadmium-induced prostate carcinogenesis. Toxicol Appl Pharmacol 2019; 375:57-63. [PMID: 31082426 DOI: 10.1016/j.taap.2019.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/03/2019] [Accepted: 05/07/2019] [Indexed: 12/21/2022]
Abstract
The aim of the present study was to investigate the genetic signatures of cadmium-transformed prostate epithelial (CTPE) cells and to identify the potential molecular signaling involved in their malignant transformation. The dataset contained normal prostate epithelial (RWPE-1) and CTPE cells. To further examine the biological functions of the identified differentially expressed genes (DEGs), Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Reactome pathway enrichment analyses were performed. In total, 2357 DEGs were identified, including 1083 upregulated genes and 1274 downregulated genes. GO, KEGG, and Reactome pathway enrichment analyses indicated that upregulated genes were significantly enriched in ECM-receptor, focal adhesion, TGFβ signaling, and syndecan interactions, while downregulated genes were mainly involved in cell cycle regulation, arachidonic acid metabolism, oxidative phosphorylation, and folate biosynthesis (p < .05). The top upregulated (SATB1 (p < .0001), EYA2 (p < .0001) and KPNA7 (p < .0027)) and downregulated (PITX2 (p < .0007), PDLIM4 (p < .0020) and FABP5 (p < .0007)) genes were further validated via qRT-PCR analysis. In conclusion, the present study profiled DEGs in RWPE-1 and CTPE cells and identified gene pathways that may be associated with malignant transformation and tumor progression.
Collapse
|
20
|
Hong X, Yu JJ. MicroRNA-150 suppresses epithelial-mesenchymal transition, invasion, and metastasis in prostate cancer through the TRPM4-mediated β-catenin signaling pathway. Am J Physiol Cell Physiol 2019; 316:C463-C480. [PMID: 30566393 DOI: 10.1152/ajpcell.00142.2018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Prostate cancer (PCa) remains one of the leading causes of cancer-related deaths among males. The aim of the current study was to investigate the ability of microRNA-150 (miR-150) targeting transient receptor potential melastatin 4 (TRPM4) to mediate epithelial-mesenchymal transition (EMT), invasion, and metastasis through the β-catenin signaling pathway in PCa. Microarray analysis was performed to identify PCa-related differentially expressed genes, after which both the mirDIP and TargetScan databases were employed in the prediction of the miRNAs regulating TRPM4. Immunohistochemistry and RT-qPCR were conducted to determine the expression pattern of miR-150 and TRPM4 in PCa. The relationship between miR-150 and TRPM4 expression was identified. By perturbing miR-150 and TRPM4 expression in PCa cells, cell proliferation, migration, invasion, cycle, and apoptosis as well as EMT markers were determined accordingly. Finally, tumor growth and metastasis were evaluated among nude mice. Higher TRPM4 expression and lower miR-150 expression and activation of the β-catenin signaling pathway as well as EMT stimulation were detected in the PCa tissues. Our results confirmed TRPM4 as a target of miR-150. Upregulation of miR-150 resulted in inactivation of the β-catenin signaling pathway. Furthermore, the upregulation of miR-150 or knockdown of TRPM4 was observed to suppress EMT, proliferation, migration, and invasion in vitro in addition to restrained tumor growth and metastasis in vivo. The evidence provided by our study highlights the involvement of miR-150 in the translational suppression of TRPM4 and the blockade of the β-catenin signaling pathway, resulting in the inhibition of PCa progression.
Collapse
Affiliation(s)
- Xi Hong
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai , China
| | - Jian-Jun Yu
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital , Shanghai , China
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus , Shanghai , China
| |
Collapse
|
21
|
Prieto-Dominguez N, Parnell C, Teng Y. Drugging the Small GTPase Pathways in Cancer Treatment: Promises and Challenges. Cells 2019; 8:E255. [PMID: 30884855 PMCID: PMC6468615 DOI: 10.3390/cells8030255] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/08/2019] [Accepted: 03/13/2019] [Indexed: 02/07/2023] Open
Abstract
Small GTPases are a family of low molecular weight GTP-hydrolyzing enzymes that cycle between an inactive state when bound to GDP and an active state when associated to GTP. Small GTPases regulate key cellular processes (e.g., cell differentiation, proliferation, and motility) as well as subcellular events (e.g., vesicle trafficking), making them key participants in a great array of pathophysiological processes. Indeed, the dysfunction and deregulation of certain small GTPases, such as the members of the Ras and Arf subfamilies, have been related with the promotion and progression of cancer. Therefore, the development of inhibitors that target dysfunctional small GTPases could represent a potential therapeutic strategy for cancer treatment. This review covers the basic biochemical mechanisms and the diverse functions of small GTPases in cancer. We also discuss the strategies and challenges of inhibiting the activity of these enzymes and delve into new approaches that offer opportunities to target them in cancer therapy.
Collapse
Affiliation(s)
- Néstor Prieto-Dominguez
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Institute of Biomedicine (IBIOMED), University of León, León 24010, Spain.
| | | | - Yong Teng
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Department of Medical laboratory, Imaging and Radiologic Sciences, College of Allied Health, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
22
|
Xiong Y, He L, Shay C, Lang L, Loveless J, Yu J, Chemmalakuzhy R, Jiang H, Liu M, Teng Y. Nck-associated protein 1 associates with HSP90 to drive metastasis in human non-small-cell lung cancer. J Exp Clin Cancer Res 2019; 38:122. [PMID: 30867003 PMCID: PMC6417146 DOI: 10.1186/s13046-019-1124-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/28/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Metastatic lung cancer is a life-threatening condition that develops when cancer in another area of the body metastasizes, or spreads, to the lung. Despite advances in our understanding of primary lung oncogenesis, the biological basis driving the progression from primary to metastatic lung cancer remains poorly characterized. METHODS Genetic knockdown of the particular genes in cancer cells were achieved by lentiviral-mediated interference. Invasion potential was determined by Matrigel and three-dimensional invasion. The secretion of matrix metalloproteinase 2 (MMP2) and MMP9 were measured by ELISA. Protein levels were assessed by Western blotting and immunohistochemistry. Protein-protein interactions were determined by immunoprecipitation. An experimental mouse model was generated to investigate the gene regulation in tumor growth and metastasis. RESULTS Nck-associated protein 1 (NAP1/NCKAP1) is highly expressed in primary non-small-cell lung cancer (NSCLC) when compared with adjacent normal lung tissues, and its expression levels are strongly associated with the histologic tumor grade, metastasis and poor survival rate of NSCLC patients. Overexpression of NAP1 in lowly invasive NSCLC cells enhances MMP9 secretion and invasion potential, whereas NAP1 silencing in highly invasive NSCLC cells produces opposing effects in comparison. Mechanistic studies further reveal that the binding of NAP1 to the cellular chaperone heat shock protein 90 (HSP90) is required for its protein stabilization, and NAP1 plays an essential role in HSP90-mediated invasion and metastasis by provoking MMP9 activation and the epithelial-to-mesenchymal transition in NSCLC cells. CONCLUSIONS Our insights demonstrate the importance and functional regulation of the HSP90-NAP1 protein complex in cancer metastatic signaling, which spur new avenues to target this interaction as a novel approach to block NSCLC metastasis.
Collapse
Affiliation(s)
- Yuanping Xiong
- Department of Otolaryngology Head and Neck Surgery, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi China
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912 USA
| | - Leilei He
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912 USA
| | - Chloe Shay
- Division of Endocrinology and Diabetes, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA USA
| | - Liwei Lang
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912 USA
| | - Jenni Loveless
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912 USA
| | - Jieqing Yu
- Department of Otolaryngology Head and Neck Surgery, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi China
| | - Ron Chemmalakuzhy
- Department of Biology, College of Science and Mathematics, Augusta University, Augusta, GA USA
| | - Hongqun Jiang
- Department of Otolaryngology Head and Neck Surgery, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi China
| | - Manran Liu
- Key Laboratory of Laboratory Medical Diagnostics Designed by Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Yong Teng
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912 USA
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912 USA
- Department of Medical Laboratory, Imaging and Radiologic Sciences, College of Allied Health, Augusta University, 1120 15th Street, Augusta, GA 30912 USA
| |
Collapse
|
23
|
He L, Gao L, Shay C, Lang L, Lv F, Teng Y. Histone deacetylase inhibitors suppress aggressiveness of head and neck squamous cell carcinoma via histone acetylation-independent blockade of the EGFR-Arf1 axis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:84. [PMID: 30777099 PMCID: PMC6379952 DOI: 10.1186/s13046-019-1080-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 02/06/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND A promising arsenal of histone deacetylase (HDAC)-targeted treatment has emerged in the past decade, as the abnormal targeting or retention of HDACs to DNA regulatory regions often occurs in many cancers. Head and neck squamous cell carcinoma (HNSCC) is one of the most aggressive malignancies worldwide associated with poor overall survival in late-stage patients. HDAC inhibitors have great potential to treat this devastating disease; however, few has been studied regarding the beneficial role of HDAC inhibition in anti-HNSCC therapy and the underlying molecular mechanisms remain elusive. METHODS Cell migration and invasion were examined by wound closure and Transwell assays. Protein levels and interactions were assessed by Western blotting and immunoprecipitation. HDAC activity was measured with the fluorometric HDAC Activity Assay. Phospho-receptor tyrosine kinase (RTK) profiling was determined by the Proteome Profiler Human Phospho-RTK Array. RESULTS ADP-ribosylation factor 1 (Arf1), a small GTPase coordinating vesicle-mediated intracellular trafficking, can be inactivated by HDAC inhibitors through histone acetylation-independent degradation of epidermal growth factor receptor (EGFR) in HNSCC cells. Mechanistically, high levels of Arf1 activity are maintained by binding to phosphorylated EGFR which is localized on HNSCC cell plasma membrane. Decreased EGFR phosphorylation is associated with reduced EGFR protein levels in the presence of TSA, which inactivates Arf1 and eventually inhibits invasion in HNSCC cells. CONCLUSIONS Our insights explore the critical role of EGFR-Arf1 complex in driving HNSCC progression, and demonstrate the selective action of HDAC inhibitors on this specific axis for suppressing HNSCC invasion. This novel finding represents the first example of modulating the EGFR-Arf1 complex in HNSCC by small molecule agents.
Collapse
Affiliation(s)
- Leilei He
- College of Bioengineering, Chongqing University, Chongqing, 400044, People's Republic of China.,Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Lixia Gao
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, 30912, USA.,Chongqing University of Arts and Sciences, Chongqing, 402160, People's Republic of China
| | - Chloe Shay
- Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Liwei Lang
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Fenglin Lv
- College of Bioengineering, Chongqing University, Chongqing, 400044, People's Republic of China.
| | - Yong Teng
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, 30912, USA. .,Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, USA. .,Department of Medical Laboratory, Imaging and Radiologic Sciences, College of Allied Health, Augusta University, Augusta, GA, USA.
| |
Collapse
|
24
|
Zhao X, Lang L, He L, Gao L, Chyan D, Xiong Y, Li H, Peng H, Teng Y. Intracellular reduction in ATP levels contributes to CYT997-induced suppression of metastasis of head and neck squamous carcinoma. J Cell Mol Med 2019; 23:1174-1182. [PMID: 30450674 PMCID: PMC6349165 DOI: 10.1111/jcmm.14017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 11/29/2022] Open
Abstract
The incidence rate of head and neck squamous cell carcinoma (HNSCC) has steadily increased over the past decade. However, treatment options for metastatic HNSCC are often limited and the 5-year survival rate has remained static. Therefore, the development and assessment of more efficient but less toxic therapeutic strategies is an unmet need for treatment of more extensive HNSCC. Here, we report that CYT997, a novel microtubule-disrupting agent, exerts strong activity in inhibiting HNSCC cell invasion and metastasis. The loss of invasion capacity by CYT997 was accompanied by an associated increase in cell adhesion and the reversal of epithelial-mesenchymal transition (EMT). Increased expression of E-cadherin protein and decreased expression of Vimentin protein became evident in HNSCC cells following CYT997 exposure, which were consistently observed in HNSCC xenografts from the mice receiving CYT997. Moreover, the capacity of invasive HNSCC cells to form pulmonary metastases was significantly blocked with CYT997 treatment, indicating that the diminishment of EMT traits contributes to CYT997-suppressed metastasis. Intriguingly, CYT997 impaired intracellular ATP levels in HNSCC cells, at least in part, through its inhibitory effect on the mitochondrial protein IF1. The addition of ATP attenuated CYT997-induced suppression of cell invasion, coupled with down-regulation of E-Cadherin and up-regulation of Vimentin. These findings support a critical role of ATP levels in cell invasion and metastasis under the influence of CYT997. Collectively, our data unveil the mechanism involved in mediating CYT997 action, and provide preclinical rationale for possible clinical application of CYT997 as a novel therapeutic strategy against aggressive HNSCC.
Collapse
Affiliation(s)
- Xiangdong Zhao
- Department of Otorhinolaryngology, Head and Neck SurgeryGuangdong Second Provincial General HospitalGuangzhouGuangdongChina
- Department of Oral Biology and Diagnostic SciencesDental College of GeorgiaAugusta UniversityAugustaGeorgia
| | - Liwei Lang
- Department of Oral Biology and Diagnostic SciencesDental College of GeorgiaAugusta UniversityAugustaGeorgia
| | - Leilei He
- Department of Oral Biology and Diagnostic SciencesDental College of GeorgiaAugusta UniversityAugustaGeorgia
| | - Lixia Gao
- Department of Oral Biology and Diagnostic SciencesDental College of GeorgiaAugusta UniversityAugustaGeorgia
| | - David Chyan
- Department of Oral Biology and Diagnostic SciencesDental College of GeorgiaAugusta UniversityAugustaGeorgia
- Department of BiologyCollege of Science and MathematicsAugusta UniversityAugustaGeorgia
| | - Yuanping Xiong
- Department of Oral Biology and Diagnostic SciencesDental College of GeorgiaAugusta UniversityAugustaGeorgia
| | - Honglin Li
- Department of Biochemistry and Molecular BiologyGeorgia Cancer CenterMedical College of GeorgiaAugusta UniversityAugustaGeorgia
| | - Hong Peng
- Department of Otorhinolaryngology, Head and Neck SurgeryGuangdong Second Provincial General HospitalGuangzhouGuangdongChina
| | - Yong Teng
- Department of Oral Biology and Diagnostic SciencesDental College of GeorgiaAugusta UniversityAugustaGeorgia
- Department of Biochemistry and Molecular BiologyGeorgia Cancer CenterMedical College of GeorgiaAugusta UniversityAugustaGeorgia
- Department of Medical Laboratory, Imaging and Radiologic SciencesCollege of Allied HealthAugusta UniversityAugustaGeorgia
| |
Collapse
|
25
|
Almiron Bonnin DA, Havrda MC, Israel MA. Glioma Cell Secretion: A Driver of Tumor Progression and a Potential Therapeutic Target. Cancer Res 2018; 78:6031-6039. [PMID: 30333116 DOI: 10.1158/0008-5472.can-18-0345] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/30/2018] [Accepted: 08/14/2018] [Indexed: 11/16/2022]
Abstract
Cellular secretion is an important mediator of cancer progression. Secreted molecules in glioma are key components of complex autocrine and paracrine pathways that mediate multiple oncogenic pathologies. In this review, we describe tumor cell secretion in high-grade glioma and highlight potential novel therapeutic opportunities. Cancer Res; 78(21); 6031-9. ©2018 AACR.
Collapse
Affiliation(s)
- Damian A Almiron Bonnin
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire.,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Matthew C Havrda
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire.,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Mark A Israel
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. .,Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire.,Departments of Medicine and Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| |
Collapse
|
26
|
Luchsinger C, Aguilar M, Burgos PV, Ehrenfeld P, Mardones GA. Functional disruption of the Golgi apparatus protein ARF1 sensitizes MDA-MB-231 breast cancer cells to the antitumor drugs Actinomycin D and Vinblastine through ERK and AKT signaling. PLoS One 2018; 13:e0195401. [PMID: 29614107 PMCID: PMC5882166 DOI: 10.1371/journal.pone.0195401] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 03/21/2018] [Indexed: 12/26/2022] Open
Abstract
Increasing evidence indicates that the Golgi apparatus plays active roles in cancer, but a comprehensive understanding of its functions in the oncogenic transformation has not yet emerged. At the same time, the Golgi is becoming well recognized as a hub that integrates its functions of protein and lipid biosynthesis to signal transduction for cell proliferation and migration in cancer cells. Nevertheless, the active function of the Golgi apparatus in cancer cells has not been fully evaluated as a target for combined treatment. Here, we analyzed the effect of perturbing the Golgi apparatus on the sensitivity of the MDA-MB-231 breast cancer cell line to the drugs Actinomycin D and Vinblastine. We disrupted the function of ARF1, a protein necessary for the homeostasis of the Golgi apparatus. We found that the expression of the ARF1-Q71L mutant increased the sensitivity of MDA-MB-231 cells to both Actinomycin D and Vinblastine, resulting in decreased cell proliferation and cell migration, as well as in increased apoptosis. Likewise, the combined treatment of cells with Actinomycin D or Vinblastine and Brefeldin A or Golgicide A, two disrupting agents of the ARF1 function, resulted in similar effects on cell proliferation, cell migration and apoptosis. Interestingly, each combined treatment had distinct effects on ERK1/2 and AKT signaling, as indicated by the decreased levels of either phospho-ERK1/2 or phospho-AKT. Our results suggest that disruption of Golgi function could be used as a strategy for the sensitization of cancer cells to chemotherapy.
Collapse
Affiliation(s)
- Charlotte Luchsinger
- Department of Physiology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Marcelo Aguilar
- Department of Physiology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Patricia V. Burgos
- Department of Physiology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Center for Cell Biology and Biomedicine (CEBICEM), School of Medicine and Science, Universidad San Sebastián, Santiago, Chile
- Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pamela Ehrenfeld
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Department of Anatomy, Histology and Pathology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Gonzalo A. Mardones
- Department of Physiology, School of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies of the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Center for Cell Biology and Biomedicine (CEBICEM), School of Medicine and Science, Universidad San Sebastián, Santiago, Chile
- * E-mail:
| |
Collapse
|
27
|
Fang LL, Sun BF, Huang LR, Yuan HB, Zhang S, Chen J, Yu ZJ, Luo H. Potent Inhibition of miR-34b on Migration and Invasion in Metastatic Prostate Cancer Cells by Regulating the TGF-β Pathway. Int J Mol Sci 2017; 18:E2762. [PMID: 29257105 PMCID: PMC5751361 DOI: 10.3390/ijms18122762] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 12/25/2022] Open
Abstract
The importance of miRNAs in the progression of prostate cancer (PCa) has further been supported by the finding that miRNAs have been identified as potential oncogenes or tumor suppressors in PCa. Indeed, in eukaryotes, miRNAs have been found to regulate and control gene expression by degrading mRNA at the post-transcriptional level. In this study, we investigated the expression of miR-34 family members, miR-34b and miR-34c, in different PCa cell lines, and discussed the molecular mechanism of miR-34b in the invasion and migration of PCa cells in vitro. The difference analyses of the transcriptome between the DU145 and PC3 cell lines demonstrated that both miR-34b and -34c target critical pathways that are involved in metabolism, such as proliferation, and migration, and invasion. The molecular expression of miR-34b/c were lower in PC3 cells. Moreover, over-expression of miR-34b/c in PC3 cells caused profound phenotypic changes, including decreased cell proliferation, migration and invasion. Moreover, the players that regulate expression levels of transforming growth factor-β (TGF-β), TGF-β receptor 1 (TGF-βR1), and p53 or phosphorylation levels of mothers against decapentaplegic 3 (SMAD3) in the TGF-β/Smad3 signaling pathway have yet to be elucidated, and will provide novel tools for diagnosis and treatment of metastatic PCa.
Collapse
Affiliation(s)
- Li-Li Fang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China.
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550002, China.
- Department of Anatomy, Guizhou Medical University, Guiyang 550000, China.
| | - Bao-Fei Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China.
- Department of Anatomy, Guizhou Medical University, Guiyang 550000, China.
| | - Li-Rong Huang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China.
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550002, China.
| | - Hai-Bo Yuan
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China.
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550002, China.
| | - Shuo Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China.
- Department of Anatomy, Guizhou Medical University, Guiyang 550000, China.
| | - Jing Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China.
- Department of Anatomy, Guizhou Medical University, Guiyang 550000, China.
| | - Zi-Jiang Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China.
- Department of Anatomy, Guizhou Medical University, Guiyang 550000, China.
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China.
- Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550002, China.
| |
Collapse
|