1
|
Mizukami H. Pathological evaluation of the pathogenesis of diabetes mellitus and diabetic peripheral neuropathy. Pathol Int 2024; 74:438-453. [PMID: 38888200 PMCID: PMC11551828 DOI: 10.1111/pin.13458] [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: 04/03/2024] [Revised: 05/29/2024] [Accepted: 06/02/2024] [Indexed: 06/20/2024]
Abstract
Currently, there are more than 10 million patients with diabetes mellitus in Japan. Therefore, the need to explore the pathogenesis of diabetes and the complications leading to its cure is becoming increasingly urgent. Pathological examination of pancreatic tissues from patients with type 2 diabetes reveals a decrease in the volume of beta cells because of a combination of various stresses. In human type 2 diabetes, islet amyloid deposition is a unique pathological change characterized by proinflammatory macrophage (M1) infiltration into the islets. The pathological changes in the pancreas with islet amyloid were different according to clinical factors, which suggests that type 2 diabetes can be further subclassified based on islet pathology. On the other hand, diabetic peripheral neuropathy is the most frequent diabetic complication. In early diabetic peripheral neuropathy, M1 infiltration in the sciatic nerve evokes oxidative stress or attenuates retrograde axonal transport, as clearly demonstrated by in vitro live imaging. Furthermore, islet parasympathetic nerve density and beta cell volume were inversely correlated in type 2 diabetic Goto-Kakizaki rats, suggesting that diabetic peripheral neuropathy itself may contribute to the decrease in beta cell volume. These findings suggest that the pathogenesis of diabetes mellitus and diabetic peripheral neuropathy may be interrelated.
Collapse
Affiliation(s)
- Hiroki Mizukami
- Department of Pathology and Molecular Medicine, Biomedical Research CenterHirosaki University Graduate School of MedicineHirosakiAomoriJapan
| |
Collapse
|
2
|
Midorikawa S, Mizukami H, Kudoh K, Takeuchi Y, Sasaki T, Kushibiki H, Wang Z, Itakura Y, Murakami K, Kudo N, Nagaki T, Wakasa T, Nakamura Y, Matsubara A. Diabetes can increase the prevalence of EBV infection and worsen the prognosis of nasopharyngeal carcinoma. Pathology 2024; 56:65-74. [PMID: 38071160 DOI: 10.1016/j.pathol.2023.09.013] [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: 01/30/2023] [Revised: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 01/24/2024]
Abstract
Epstein‒Barr virus (EBV) infection is a primary oncogenic factor of nasopharyngeal carcinoma (NPC) that elicits epithelial-mesenchymal transition (EMT). Although diabetic patients are more susceptible to various infectious diseases, the pathological association with virus-related NPC has not yet been clarified. Herein, we evaluated the influence of diabetes on the clinicopathological changes of 70 patients with NPC. Disease-specific survival (DSS) modified by viral infection was also analysed. The proportion of NPC patients with diabetes was 32.9% (23/70 cases), and 91.3% (21/23 cases) were infected with EBV detected by EBER-I in situ hybridisation. NPC with diabetes showed an effect on EMT evaluated by immunostaining for E-cadherin and vimentin, which was correlated with HbA1c levels. Receiver operating characteristic (ROC) curve analysis determined a HbA1c level of 6.5% as the cut-off value for primary disease death at 2 years [area under the curve (AUC) 0.76; sensitivity 0.64; and specificity 0.81]. High HbA1c levels (≥6.5%) significantly increased the number of lymph node metastases in NPC compared to low HbA1c levels (<6.5%, p<0.01). Diabetic NPC patients had a significantly poorer prognosis than all non-diabetic patients (DSS, 72 months vs not reached, p<0.05). Diabetic EBV-positive NPC patients had a significantly poorer prognosis than non-diabetic EBV-positive patients (DSS, 35 months vs not reached, p<0.01). Multivariate analysis using the Cox proportional hazards model also suggested that HbA1c ≥6.5% was a significant factor in poor prognosis, with a hazard ratio of 6.84 (p<0.05). Collectively, our results revealed for the first time a high prevalence of EBV infection, poor prognosis and the importance of proper glycaemic control in diabetic NPC patients.
Collapse
Affiliation(s)
- Shin Midorikawa
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan; Department of Otolaryngology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Hiroki Mizukami
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan.
| | - Kazuhiro Kudoh
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yuki Takeuchi
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Takanori Sasaki
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Hanae Kushibiki
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Zhenchao Wang
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yuko Itakura
- Department of Pathology, Japanese Red Cross Ishinomaki Hospital, Ishinomaki, Miyagi, Japan
| | - Kotaro Murakami
- Department of Pathology and Molecular Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Naomi Kudo
- Department of Otolaryngology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Takahiko Nagaki
- Department of Otolaryngology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Tomoko Wakasa
- Department of Diagnostic Pathology, Kindai University Nara Hospital, Nara, Japan
| | - Yasuhiro Nakamura
- Division of Pathology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Atsushi Matsubara
- Department of Otolaryngology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| |
Collapse
|
3
|
Gao X, Qian J, Zhang Y, Wang H, Cui J, Yang Y. Analysis of differential membrane proteins related to matrix stiffness-mediated metformin resistance in hepatocellular carcinoma cells. Proteome Sci 2023; 21:14. [PMID: 37740172 PMCID: PMC10517517 DOI: 10.1186/s12953-023-00216-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/01/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND Our previous work shows that increased matrix stiffness not only alters malignant characteristics of hepatocellular carcinoma (HCC) cells, but also attenuates metformin efficacy in treating HCC cells. Here, we identified differential membrane proteins related to matrix stiffness-mediated metformin resistance for better understand therapeutic resistance of metformin in HCC. METHODS Differential membrane proteins in HCC cells grown on different stiffness substrates before and after metformin intervention were screened and identified using isobaric tags for relative and absolute quantification (iTRAQ) labeling coupled with the liquid chromatography-tandem mass spectrometry (LC-MS/MS), then bioinformatic analysis were applied to determine candidate membrane protein and their possible signaling pathway. RESULTS A total of 5159 proteins were identified and 354 differential membrane proteins and membrane associated proteins, which might be associated with matrix stiffness-mediated metformin resistance were discovered. Then 94 candidate membrane proteins including 21 up-regulated protein molecules and 73 down-regulated protein molecules were further obtained. Some of them such as Annexin A2 (ANXA2), Filamin-A (FLNA), Moesin (MSN), Myosin-9 (MYH9), Elongation factor 2 (eEF2), and Tax1 binding Protein 3 (TAX1BP3) were selected for further validation. Their expressions were all downregulated in HCC cells grown on different stiffness substrates after metformin intervention. More importantly, the degree of decrease was obviously weakened on the higher stiffness substrate compared with that on the lower stiffness substrate, indicating that these candidate membrane proteins might contribute to matrix stiffness-mediated metformin resistance in HCC. CONCLUSIONS There was an obvious change in membrane proteins in matrix stiffness-mediated metformin resistance in HCC cells. Six candidate membrane proteins may reflect the response of HCC cells under high stiffness stimulation to metformin intervention, which deserve to be investigated in the future.
Collapse
Affiliation(s)
- Xiangyu Gao
- Department of Endocrinology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, PR China
| | - Jiali Qian
- Department of Endocrinology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, PR China
| | - Yang Zhang
- Institute of Biomedical Science, Fudan University, 131 Dong' an Road, Shanghai, 200032, PR China
| | - Heming Wang
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China
| | - Jiefeng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University & Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, 136 Yi Xue Yuan Road, Shanghai, 200032, PR China.
| | - Yehong Yang
- Department of Endocrinology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, PR China.
| |
Collapse
|
4
|
Hara Y, Mizukami H, Yamazaki K, Yamada T, Igawa A, Takeuchi Y, Sasaki T, Kushibiki H, Murakami K, Kudoh K, Ishido K, Hakamada K. Dual epigenetic changes in diabetes mellitus-associated pancreatic ductal adenocarcinoma correlate with downregulation of E-cadherin and worsened prognosis. J Pathol Clin Res 2023; 9:354-366. [PMID: 37246239 PMCID: PMC10397378 DOI: 10.1002/cjp2.326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/02/2023] [Accepted: 05/04/2023] [Indexed: 05/30/2023]
Abstract
Diabetes mellitus (DM) is a risk factor for pancreatic ductal adenocarcinoma (PDAC) that promotes the promoter methylation of CDH1. It is still unclear whether DM can exert other epigenetic effects, such as altering microRNA (miR) expression, in PDAC. The expression of miR-100-5p is known to be changed in DM patients and can suppress the expression of E-cadherin. In this study, the correlation between DM status and dual epigenetic changes was evaluated in PDAC specimens from patients who underwent radical surgical resection. A total of 132 consecutive patients with PDAC were clinicopathologically evaluated. E-cadherin and nuclear β-catenin expression was measured using immunohistochemistry. DNA and miRs were extracted from the main tumor site on formalin-fixed paraffin-embedded tissue sections. TaqMan miR assays were applied to assess miR-100-5p expression. Bisulfite modification was conducted on the extracted DNA, which was then subjected to methylation-specific polymerase chain reaction. Immunohistochemistry revealed that decreased E-cadherin expression and increased nuclear β-catenin expression were significantly associated with DM and poor tumor cell differentiation. The presence of long-duration DM (≥3 years) was a significant factor contributing to CDH1 promoter methylation (p < 0.01), while miR-100-5p expression was proportionally correlated with the preoperative HbA1c level (R = 0.34, p < 0.01), but not the duration of DM. The subjects with high miR-100-5p expression and CDH1 promoter methylation showed the highest level of vessel invasion and prevalence of tumor size ≥30 mm. PDAC subjects with dual epigenetic changes showed poorer overall survival (OS) than those with a single epigenetic change. miR-100-5p expression ≥4.13 and CDH1 promoter methylation independently predicted poor OS and disease-free survival (DFS) in the multivariate analysis. OS and DFS worsened in DM subjects with both HbA1c ≥ 6.5% and DM duration ≥3 years. Thus, DM is associated with two modes of epigenetic change by independent mechanisms and worsens prognosis.
Collapse
Affiliation(s)
- Yutaro Hara
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
- Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineHirosakiJapan
| | - Hiroki Mizukami
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| | - Keisuke Yamazaki
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
- Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineHirosakiJapan
| | - Takahiro Yamada
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
- Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineHirosakiJapan
| | - Akiko Igawa
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
- Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineHirosakiJapan
| | - Yuki Takeuchi
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| | - Takanori Sasaki
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| | - Hanae Kushibiki
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| | - Kotaro Murakami
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| | - Kazuhiro Kudoh
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineHirosakiJapan
| | - Keinosuke Ishido
- Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineHirosakiJapan
| | - Kenichi Hakamada
- Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineHirosakiJapan
| |
Collapse
|
5
|
Pan X, Mizukami H, Hara Y, Yamada T, Yamazaki K, Kudoh K, Takeuchi Y, Sasaki T, Kushibiki H, Igawa A, Hakamada K. Diabetes mellitus impacts on expression of DNA mismatch repair protein PMS2 and tumor microenvironment in pancreatic ductal adenocarcinoma. J Diabetes Investig 2022; 14:132-144. [PMID: 36453157 PMCID: PMC9807152 DOI: 10.1111/jdi.13929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 09/10/2022] [Accepted: 10/06/2022] [Indexed: 12/03/2022] Open
Abstract
AIMS/INTRODUCTION The mismatch repair (MMR) protein recognizes DNA replication errors and plays an important role in tumorigenesis, including pancreatic ductal adenocarcinoma (PDAC). Although PMS2, a MMR protein, is degraded under oxidative stress, the effects of diabetes are still unclear. Herein, we focused on whether diabetes affected MMR protein expression in PDAC. MATERIALS AND METHODS Tissues from 61 surgically resected PDAC subjects were clinicopathologically analyzed. Immunohistochemical analysis was performed for MMR protein expression, oxidative stress, and immune cell infiltration. The change of MMR protein expression was assessed in PDAC cell lines under stimulation with 25 mM glucose and 500 μM palmitic acid. Survival curves were analyzed by the Kaplan-Meier method with the log-rank test. RESULTS Diabetes complicated with dyslipidemia significantly decreased the expression of PMS2 in PDAC tissues with an inverse correlation with the degree of oxidative stress. Palmitic acid combined with high glucose induced degradation of PMS2 protein, enhancing oxidative stress in vitro. CD8+ T-cell infiltration was associated with a short duration of type 2 diabetes (≤4 years) and a low expression of PMS2 in PDAC tissues, while CD163+ tumor-associated macrophage infiltration was increased with a long duration of diabetes (>4 years). A short duration of diabetes exhibited a better prognosis than nondiabetic subjects with PDAC (P < 0.05), while a long duration of diabetes had a worse prognosis (P < 0.05). CONCLUSIONS The different phases of diabetes have a major impact on PDAC by altering PMS2 expression and the tumor immune microenvironment, which can be targeted by an immune checkpoint inhibitor.
Collapse
Affiliation(s)
- Xuekai Pan
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineAomoriJapan,Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineAomoriJapan
| | - Hiroki Mizukami
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineAomoriJapan
| | - Yutaro Hara
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineAomoriJapan,Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineAomoriJapan
| | - Takahiro Yamada
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineAomoriJapan,Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineAomoriJapan
| | - Keisuke Yamazaki
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineAomoriJapan,Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineAomoriJapan
| | - Kazuhiro Kudoh
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineAomoriJapan
| | - Yuki Takeuchi
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineAomoriJapan
| | - Takanori Sasaki
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineAomoriJapan
| | - Hanae Kushibiki
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineAomoriJapan
| | - Akiko Igawa
- Department of Pathology and Molecular MedicineHirosaki University Graduate School of MedicineAomoriJapan,Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineAomoriJapan
| | - Kenichi Hakamada
- Department of Gastroenterological SurgeryHirosaki University Graduate School of MedicineAomoriJapan
| |
Collapse
|
6
|
Bian Q, Li H, Wang X, Liang T, Zhang K. Multiomics Integrated Analysis Identifies SLC24A2 as a Potential Link between Type 2 Diabetes and Cancer. J Diabetes Res 2022; 2022:4629419. [PMID: 35601016 PMCID: PMC9122708 DOI: 10.1155/2022/4629419] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 04/11/2022] [Indexed: 11/24/2022] Open
Abstract
Background So far, type 2 diabetes (T2D) is considered as an independent risk factor for various cancers, but the underlying mechanism remains unclear. Methods. SLC24A2 was first identified as a key gene strongly associated with fasting plasma glucose (FPG). Then, overlapped differentially expressed genes (DEGs) between T2D verse control and SLC24A2-high verse SLC24A2-low were extracted and imported into weighted correlation network analysis. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and gene set enrichment analysis were used for functional enrichment analysis of DEGs. Least absolute shrinkage and selection operator was utilized to build a T2D prediction model. Timer and K-M plotters were employed to find the expression and prognosis of SLC24A2 in pan cancer. Results Interestingly, both DEGs between T2D verse control and SLC24A2-high verse SLC24A2-low enriched in cancer-related pathways. Moreover, a total of 3719 overlapped DEGs were divided into 8 functional modules. Grey module negatively correlated with T2D and FPG and was markedly involved in ribosome biogenesis. Ten SLC24A2-related genes (RRP36, RPF1, GRWD1, FBL, EXOSC5, BCCIP, UTP14A, TWISTNB, TBL3, and SKIV2L) were identified as hub genes, based on which the LASSO model accurately predicts the occurrence of T2D (AUC = 0.841). In addition, SLC24A2 was only expressed in islet β cells and showed abnormal expression in 17 kinds of cancers and significantly correlated with the prognosis of 10 kinds of cancers. Conclusion Taken together, SLC24A2 may link T2D and cancer by influencing the ribosome function of islet β cells and play different prognostic roles in different cancers.
Collapse
Affiliation(s)
- Qin Bian
- Department of Clinical Laboratory, Guangyuan Central Hospital, Guangyuan 628000, China
| | - Haijun Li
- Department of Clinical Laboratory, Guangyuan Central Hospital, Guangyuan 628000, China
| | - Xiaoyi Wang
- Department of Medical Imaging, Guangyuan Central Hospital, Guangyuan 628000, China
| | - Tingting Liang
- Department of Hospital-Acquired Infection Control, Guangyuan Central Hospital, Guangyuan 628000, China
| | - Kai Zhang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| |
Collapse
|
7
|
Zhang C, Liu S, Yang M. Hepatocellular Carcinoma and Obesity, Type 2 Diabetes Mellitus, Cardiovascular Disease: Causing Factors, Molecular Links, and Treatment Options. Front Endocrinol (Lausanne) 2021; 12:808526. [PMID: 35002979 PMCID: PMC8733382 DOI: 10.3389/fendo.2021.808526] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, which will affect more than a million people by the year 2025. However, current treatment options have limited benefits. Nonalcoholic fatty liver disease (NAFLD) is the fastest growing factor that causes HCC in western countries, including the United States. In addition, NAFLD co-morbidities including obesity, type 2 diabetes mellitus (T2DM), and cardiovascular diseases (CVDs) promote HCC development. Alteration of metabolites and inflammation in the tumor microenvironment plays a pivotal role in HCC progression. However, the underlying molecular mechanisms are still not totally clear. Herein, in this review, we explored the latest molecules that are involved in obesity, T2DM, and CVDs-mediated progression of HCC, as they share some common pathologic features. Meanwhile, several therapeutic options by targeting these key factors and molecules were discussed for HCC treatment. Overall, obesity, T2DM, and CVDs as chronic metabolic disease factors are tightly implicated in the development of HCC and its progression. Molecules and factors involved in these NAFLD comorbidities are potential therapeutic targets for HCC treatment.
Collapse
Affiliation(s)
- Chunye Zhang
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Shuai Liu
- The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO, United States
| |
Collapse
|
8
|
Gao C, Fang L, Zhang H, Zhang WS, Li XO, Du SY. Metformin Induces Autophagy via the AMPK-mTOR Signaling Pathway in Human Hepatocellular Carcinoma Cells. Cancer Manag Res 2020; 12:5803-5811. [PMID: 32765083 PMCID: PMC7371564 DOI: 10.2147/cmar.s257966] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 07/03/2020] [Indexed: 12/11/2022] Open
Abstract
Background Metformin may exert the anticancer effect on multiple types of cancers and some potential mechanisms have been suggested. Our study was designed to determine the effect of metformin on the cell autophagy and autophagic flux via the AMPK-mTOR signaling pathway in human hepatocellular carcinoma (HCC) cells. Methods MHCC97H and HepG2 cell lines were cultured and treated without and with metformin at various concentrations (2, 5, 10 and 20 mM) for 48 h. Then, 10 mM was determined as the optimal concentration and the HCC cells were treated with metformin for 12, 24, 48, and 72 h. MTT assay was used to assess the cell viability and Western blotting was used to determine the expression of proteins (LC3-II, p62, phospho-AMPKα, phospho-mTOR, mTOR, phospho-p70 S6 Kinase, p70 S6 Kinase, PARP1, Caspase-9 and Caspase-3). Autophagy inhibitor 3-methyladenine, EGFP-LC3 and mCherry-GFP-LC3B plasmid transfection were used for further study. Results Metformin inhibited significantly the viability of MHCC97H and HepG2 cells in a dose- and time-dependent manner. For the apoptotic properties, activation of Caspase-9 and Caspase-3 and PARP cleavage were not observed after treatment with metformin. MHCC97H cells were transfected with a EGFP-LC3 plasmid and treatment with metformin could lead to the increased level of LC3-II and decreased level of p62. In metformin-induced autophagy, AMPK expression was activated, and the phosphorylation levels of mTOR and p70 S6 Kinase were inhibited. Metformin treatment and mCherry-GFP-LC3B plasmid transfection showed that metformin could induce the autophagic flux. 3-Methyladenine (3-MA) partly abolished this effect. Conclusion Metformin could induce the autophagy, autophagic flux, and activate the AMPK-mTOR signaling pathway in human HCC cells.
Collapse
Affiliation(s)
- Chun Gao
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China
| | - Long Fang
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China
| | - Hui Zhang
- Department of Gastroenterology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, People's Republic of China
| | - Wei-Shuo Zhang
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China
| | - Xiao-Ou Li
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China
| | - Shi-Yu Du
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China
| |
Collapse
|