1
|
Zhang Y, Chen S, You L, He Z, Xu P, Huang W. LINC00161 upregulated by M2-like tumor-associated macrophages promotes hepatocellular carcinoma progression by methylating HACE1 promoters. Cytotechnology 2024; 76:777-793. [PMID: 39435425 PMCID: PMC11490593 DOI: 10.1007/s10616-024-00653-y] [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: 03/19/2024] [Accepted: 08/10/2024] [Indexed: 10/23/2024] Open
Abstract
M2-like tumor-associated macrophages (M2-TAM) played an essential part in hepatocellular carcinoma (HCC) progression. Long intergenic noncoding RNA 00161 (LINC00161), is a long non-coding RNA, that was related to HCC development. However, the relationship between LINC00161 and TAM remains indistinct. HCC cells were cocultured with an M2-like conditioned medium (M2-CM). cell counting kit-8 (CCK-8), plate cloning, cell scratch, and transwell assay evaluated cell biological activities of HCC cells. The interactions among molecules were analyzed by chromatin immunoprecipitation (CHIP), dual-luciferase reporter, and RNA immunoprecipitation (RIP). The methylation status of HECT domain and ankyrin repeat-containing, E3 ubiquitin protein ligase 1 (HACE1) was evaluated using methylation-specific PCR (MSP) and bisulfite sequencing PCR (BSP). The xenograft model was established in vivo using subcutaneous nude mice. Histological analyses were performed using hematoxylin-eosin (HE) staining. The expression of molecules was determined using immunohistochemistry (IHC), western blot and quantitative real-time PCR (qPCR). LINC00161 expression was promoted in HCC. LINC00161 knockdown significantly reduced HCC cell proliferation, migration, and invasion. Additionally, M2-TAM stimulated LINC00161 transcription and expression in HCC cells by secreting hepatocyte growth factor (HGF) to activate the Met/NFκB pathway. LINC00161 suppressed HACE1 expression, and knockdown of LINC00161 decreased the methylation on the HACE1 promoter. Meanwhile, a binding relationship between the enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) and HACE1 was observed. LINC00161 overexpression increased the binding of EZH2 on the HACE1 promoter region. Furthermore, LINC00161 knockdown suppressed tumor growth in vivo and induced HACE1 expression by inhibiting its methylation. LINC00161, induced by M2-TAM, played a pivotal role in contributing to HCC development by recruiting EZH2 to promote the methylation of HACE1. This underscores the significant involvement of LINC00161 in mediating the progression of HCC.
Collapse
Affiliation(s)
- Yujunya Zhang
- Third Clinical Medical College of Xinjiang Medical University (Affiliated Cancer Hospital), Urumqi, 830011 Xinjiang Uygur China
| | - Shuying Chen
- Third Clinical Medical College of Xinjiang Medical University (Affiliated Cancer Hospital), Urumqi, 830011 Xinjiang Uygur China
| | - Lina You
- Traditional Chinese Medicine Oncology Department, The Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, 830011 Xinjiang Uygur China
| | - Zhanao He
- Interventional Diagnosis and Treatment Department, The Affiliated Cancer Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Xinshi District, Urumqi, 830011 Xinjiang Uygur China
| | - Peidong Xu
- Interventional Diagnosis and Treatment Department, The Affiliated Cancer Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Xinshi District, Urumqi, 830011 Xinjiang Uygur China
| | - Wukui Huang
- Interventional Diagnosis and Treatment Department, The Affiliated Cancer Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Xinshi District, Urumqi, 830011 Xinjiang Uygur China
| |
Collapse
|
2
|
Li D, Onodera S, Yu Q, Zhou J. The impact of alternate-day fasting on the salivary gland stem cell compartments in non-obese diabetic mice with newly established Sjögren's syndrome. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119817. [PMID: 39159683 PMCID: PMC11368138 DOI: 10.1016/j.bbamcr.2024.119817] [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: 05/16/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/21/2024]
Abstract
Intermittent fasting exerts a profound beneficial influence on a spectrum of diseases through various mechanisms including regulation of immune responses, elimination of senescent- and pathogenic cells and improvement of stem cell-based tissue regeneration in a disease- and tissue-dependent manner. Our previous study demonstrated that alternate-day fasting (ADF) led to alleviation of xerostomia and sialadenitis in non-obese diabetic (NOD) mice, a well-defined model of Sjögren's syndrome (SS). This present study delved into the previously unexplored impacts of ADF in this disease setting and revealed that ADF increases the proportion of salivary gland stem cells (SGSCs), defined as the EpCAMhi cell population among the lineage marker negative submandibular gland (SMG) cells. Furthermore, ADF downregulated the expression of p16INK4a, a cellular senescence marker, which was concomitant with increased apoptosis and decreased expression and activity of NLRP3 inflammasomes in the SMGs, particularly in the SGSC-residing ductal compartments. RNA-sequencing analysis of purified SGSCs from NOD mice revealed that the significantly downregulated genes by ADF were mainly associated with sugar metabolism, amino acid biosynthetic process and MAPK signaling pathway, whereas the significantly upregulated genes related to fatty acid metabolic processes, among others. Collectively, these findings indicate that ADF increases the SGSC proportion, accompanied by a modulation of the SGSC property and a switch from sugar- to fatty acid-based metabolism. These findings lay the foundation for further investigation into the functionality of SGSCs influenced by ADF and shed light on the cellular and molecular mechanisms by which ADF exerts beneficial actions on salivary gland restoration in SS.
Collapse
Affiliation(s)
- Dongfang Li
- The ADA Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA
| | - Shoko Onodera
- The ADA Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA; Department of Biochemistry, Tokyo Dental College, Tokyo, Japan
| | - Qing Yu
- The ADA Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA
| | - Jing Zhou
- The ADA Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA.
| |
Collapse
|
3
|
Sun Z, Ren M, Niu J, Tang G, Li Y, Kong F, Song X. miR-29b-3p targetedly regulates VEGF to inhibit tumor progression and cisplatin resistance through Nrf2/HO-1 signaling pathway in non-small cell lung cancer. ENVIRONMENTAL TOXICOLOGY 2024; 39:3956-3966. [PMID: 38587027 DOI: 10.1002/tox.24253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/09/2024] [Accepted: 03/14/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUNDS Non-small cell lung carcinoma (NSCLC) is a common type of lung cancer. Prior investigations have elucidated the pivotal role of miR-29b-3p in restraining tumor growth and metastasis. Nonetheless, it remains to be determined whether miR-29b-3p can effectively suppress NSCLC progression and enhance the sensitivity of NSCLC cells to cisplatin. This investigation sought to determine the mechanism by which miR-29b-3p inhibited the advancement of NSCLC and mitigated resistance to cisplatin. METHODS We initially assessed miR-29b-3p and VEGF levels in NSCLC tissues and cell lines. Next, miR-29b-3p expression was elevated in NSCLC cell lines H1975 and A549 by overexpression plasmid transfection. Following this, a sequence of molecular biology experiments was conducted to evaluate the impact of miR-29b-3p on the biological behaviors of NSCLC cells and their resistance to cisplatin. Additionally, we predicted VEGF was a target gene of miR-29b-3p by bioinformatics analysis. We next employed western blot to evaluate the protein expression of Nrf2 and HO-1 in NSCLC cells. Finally, we elucidated the effects of VEGF and Nrf2/HO-1pathway on NSCLC progression and cisplatin resistance by in vitro assays. RESULTS In comparison to paracancerous tissues and human normal lung epithelial cells, the expression of miR-29b-3p was notably reduced and VEGF expression was clearly elevated in NSCLC tissues and cells. Moreover, miR-29b-3p upregulated obviously suppressed the biological activities of NSCLC cells and increased their sensitivity to cisplatin. Furthermore, in NSCLC cells, miR-29b-3p bound to VEGF and negatively regulate its transcription. Additionally, miR-29b-3p overexpression also inhibited the Nrf2/HO-1 signaling pathway. Finally, the overexpression of VEGF and the activation of the Nrf2/HO-1 pathway reversed miR-29b-3p-mediated inhibitory effect on biological behaviors of NSCLC cells and increased the cisplatin resistance. CONCLUSION Our findings indicate that miR-29b-3p impedes NSCLC cells' biological behaviors and augments their sensitivity to cisplatin by targeting VEGF to modulate the Nfr2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Zhen Sun
- Department of Thoracic Surgery, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Mingming Ren
- Department of Thoracic Surgery, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Jieting Niu
- Department of Geriatrics, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Guojie Tang
- Department of Thoracic Surgery, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Yanguang Li
- Department of Thoracic Surgery, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Fanyi Kong
- Department of Thoracic Surgery, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Xiang Song
- Department of Thoracic Surgery, Cangzhou Central Hospital, Cangzhou, Hebei, China
| |
Collapse
|
4
|
Wang Y, Peng J, Yang D, Xing Z, Jiang B, Ding X, Jiang C, Ouyang B, Su L. From metabolism to malignancy: the multifaceted role of PGC1α in cancer. Front Oncol 2024; 14:1383809. [PMID: 38774408 PMCID: PMC11106418 DOI: 10.3389/fonc.2024.1383809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/16/2024] [Indexed: 05/24/2024] Open
Abstract
PGC1α, a central player in mitochondrial biology, holds a complex role in the metabolic shifts seen in cancer cells. While its dysregulation is common across major cancers, its impact varies. In some cases, downregulation promotes aerobic glycolysis and progression, whereas in others, overexpression escalates respiration and aggression. PGC1α's interactions with distinct signaling pathways and transcription factors further diversify its roles, often in a tissue-specific manner. Understanding these multifaceted functions could unlock innovative therapeutic strategies. However, challenges exist in managing the metabolic adaptability of cancer cells and refining PGC1α-targeted approaches. This review aims to collate and present the current knowledge on the expression patterns, regulators, binding partners, and roles of PGC1α in diverse cancers. We examined PGC1α's tissue-specific functions and elucidated its dual nature as both a potential tumor suppressor and an oncogenic collaborator. In cancers where PGC1α is tumor-suppressive, reinstating its levels could halt cell proliferation and invasion, and make the cells more receptive to chemotherapy. In cancers where the opposite is true, halting PGC1α's upregulation can be beneficial as it promotes oxidative phosphorylation, allows cancer cells to adapt to stress, and promotes a more aggressive cancer phenotype. Thus, to target PGC1α effectively, understanding its nuanced role in each cancer subtype is indispensable. This can pave the way for significant strides in the field of oncology.
Collapse
Affiliation(s)
- Yue Wang
- Department of Surgery, Nanjing Central Hospital, Nanjing, China
| | - Jianing Peng
- Division of Biosciences, University College London, London, United Kingdom
| | - Dengyuan Yang
- Department of Surgery, Nanjing Central Hospital, Nanjing, China
| | - Zhongjie Xing
- Department of Surgery, Nanjing Central Hospital, Nanjing, China
| | - Bo Jiang
- Department of General Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Xu Ding
- Department of Surgery, Nanjing Central Hospital, Nanjing, China
| | - Chaoyu Jiang
- Department of General Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
| | - Bing Ouyang
- Department of Surgery, Nanjing Central Hospital, Nanjing, China
| | - Lei Su
- Department of General Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
- Department of General Surgery, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
5
|
YAP regulates the liver size during the fasting-refeeding transition in mice. Acta Pharm Sin B 2022; 13:1588-1599. [PMID: 37139422 PMCID: PMC10149903 DOI: 10.1016/j.apsb.2022.12.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 11/11/2022] [Accepted: 11/18/2022] [Indexed: 03/09/2023] Open
Abstract
Liver is the central hub regulating energy metabolism during feeding-fasting transition. Evidence suggests that fasting and refeeding induce dynamic changes in liver size, but the underlying mechanisms remain unclear. Yes-associated protein (YAP) is a key regulator of organ size. This study aims to explore the role of YAP in fasting- and refeeding-induced changes in liver size. Here, fasting significantly reduced liver size, which was recovered to the normal level after refeeding. Moreover, hepatocyte size was decreased and hepatocyte proliferation was inhibited after fasting. Conversely, refeeding promoted hepatocyte enlargement and proliferation compared to fasted state. Mechanistically, fasting or refeeding regulated the expression of YAP and its downstream targets, as well as the proliferation-related protein cyclin D1 (CCND1). Furthermore, fasting significantly reduced the liver size in AAV-control mice, which was mitigated in AAV Yap (5SA) mice. Yap overexpression also prevented the effect of fasting on hepatocyte size and proliferation. Besides, the recovery of liver size after refeeding was delayed in AAV Yap shRNA mice. Yap knockdown attenuated refeeding-induced hepatocyte enlargement and proliferation. In summary, this study demonstrated that YAP plays an important role in dynamic changes of liver size during fasting-refeeding transition, which provides new evidence for YAP in regulating liver size under energy stress.
Collapse
|
6
|
Is Fasting Good When One Is at Risk of Liver Cancer? Cancers (Basel) 2022; 14:cancers14205084. [PMID: 36291868 PMCID: PMC9600146 DOI: 10.3390/cancers14205084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC), one of the leading causes of cancer-related deaths worldwide, is a multistep process that usually develops in the background of cirrhosis, but also in a non-cirrhotic state in patients with non-alcoholic fatty liver disease (NAFLD) or viral hepatis. Emerging evidence suggests that intermittent fasting can reduce the risk of cancer development and could improve response and tolerance to treatment through the metabolic and hormonal adaptations induced by the low energy availability that finally impairs cancer cells’ adaptability, survival and growth. The current review will outline the beneficial effects of fasting in NAFLD/NASH patients and the possible mechanisms that can prevent HCC development, including circadian clock re-synchronization, with a special focus on the possibility of applying this dietary intervention to cirrhotic patients.
Collapse
|
7
|
Bai Y, Cheng M, Jin J, Zhang H, He L, Zhou W, Zhang S, Xu J. SET8, a novel regulator to ameliorate vascular calcification via activating PI3K/Akt mediated anti-apoptotic effects. Biochem Cell Biol 2021; 100:104-114. [PMID: 34846946 DOI: 10.1139/bcb-2021-0322] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previous studies have shown that the apoptosis of vascular smooth muscle cells (VSMCs) underlies the mechanism of pathological calcification in patients with chronic kidney disease (CKD). SET domain-containing protein 8 (SET8) is an efficient protein that modulates apoptosis in hepatocellular carcinoma cells, esophageal squamous cells, and neuronal cells by regulating pathological processes, such as cell cycle progression and transcription regulation. However, whether SET8 is involved in high phosphorus-induced vascular calcification by mediating apoptosis remains unclear. Here, we report that SET8 is located both in the nucleus and cytoplasm and is significantly downregulated in calcification models. SET8 deficiency promoted apoptosis of VSMCs, as indicated by the increased Bax/Bcl-2 and cleaved caspase-3/total caspase-3 ratios. Mechanistically, the PI3K/Akt pathway was mediated by SET8, and inhibition of the PI3K/Akt signaling pathway by administering LY294002 or transfecting the Akt phosphorylation-inactivated mutation plasmid increased apoptosis and calcification. Akt phosphorylation constitutively activated mutations can reduce the apoptosis and calcification of VSMCs. Furthermore, exogenous overexpression of SET8 reversed the effect of PI3K/Akt inhibition on VSMC apoptosis and calcification. In summary, our research suggests that SET8 overexpression ameliorates high phosphorus-induced calcification of VSMCs by activating PI3K/Akt mediated anti-apoptotic effects.
Collapse
Affiliation(s)
- Yaling Bai
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China.,Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Meijuan Cheng
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China.,Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Jingjing Jin
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China.,Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Huiran Zhang
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China.,Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Lei He
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China.,Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Wei Zhou
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China.,Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Shenglei Zhang
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China.,Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| | - Jinsheng Xu
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China.,Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, P.R. China
| |
Collapse
|
8
|
Emara MH, Soliman HH, Elnadry M, Mohamed Said E, Abd-Elsalam S, Elbatae HE, Zaher TI, Ezzeldin S Bazeed S, Abdel-Razik A, Youssef Mohamed S, Elfert A. Ramadan fasting and liver diseases: A review with practice advices and recommendations. Liver Int 2021; 41:436-448. [PMID: 33369880 DOI: 10.1111/liv.14775] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 02/05/2023]
Abstract
Ramadan fasting is obligatory for Muslim healthy adults. However, there are many exemptions from fasting; including patients, whose diseases will be aggravated by fasting. Muslim patients with different liver diseases are frequently seen in the clinics discussing their intent to fast this month with their treating physicians. To answer our patients' inquiries about the expected benefits and/or risks of fasting and delivering them the best care, we carried out this review and we draw advices and recommendations based on the available evidence. A web-based search, combining multiple keywords representing different liver diseases with Ramadan fasting had been carried out. To answer the research question: Do adult Muslim patients with different liver diseases who fast the month of Ramadan have had a deleterious effect on their health in comparison to those who did not fast? Relevant publications were retrieved. No randomized controlled trials were focusing on Ramadan fasting and liver diseases in the filtered databases, eg Cochrane library. Consequently, non-filtered databases, eg PubMed, Google Scholar and Egyptian Knowledge Bank searched and full-text high-quality research articles were carefully analysed to draw recommendations. Other relevant publications with low quality of evidence like case studies and short communications were also reviewed to address practice advices. Although Ramadan fasting was found beneficial for patients with NAFLD, it was found deleterious to patients with Child B and C cirrhosis and patients with peptic ulcer. Patients with chronic hepatitis, Child A cirrhosis and those with non-complicated liver transplant can fast with prefasting assessment and strict follow up.
Collapse
Affiliation(s)
- Mohamed H Emara
- Faculty of Medicine, Department of Hepatology, Gastroenterology and infectious diseases, Kafrelsheikh University, Kafr El-Shikh, Egypt
| | - Hanan H Soliman
- Faculty of Medicine, Tropical medicine and infectious diseases Department, Tanta University, Tanta, Egypt
| | - Mohamed Elnadry
- Faculty of Medicine, Hepato-gastroenterology and infectious diseases Department, Al-Azhar University, Cairo, Egypt
| | - Ebada Mohamed Said
- Faculty of Medicine, Hepatology, Gastroenterology and Infectious Diseases Department, Benha University, Benha, Egypt
| | - Sherief Abd-Elsalam
- Faculty of Medicine, Tropical medicine and infectious diseases Department, Tanta University, Tanta, Egypt
| | - Hassan E Elbatae
- Faculty of Medicine, Department of Hepatology, Gastroenterology and infectious diseases, Kafrelsheikh University, Kafr El-Shikh, Egypt
| | - Tarik I Zaher
- Faculty of Medicine, Department of Tropical Medicine, Zagazig University, Zagazig, Egypt
| | - Shamardan Ezzeldin S Bazeed
- Faculty of Medicine, Tropical Medicine and Gastroenterology Department, South Valley University, Qena, Egypt
| | - Ahmed Abdel-Razik
- Faculty of Medicine, Department of Tropical Medicine, Mansoura University, Mansoura, Egypt
| | - Salem Youssef Mohamed
- Faculty of Medicine, Department of Internal Medicine, Zagazig University, Zagazig, Egypt
| | - Asem Elfert
- Faculty of Medicine, Tropical medicine and infectious diseases Department, Tanta University, Tanta, Egypt
| | | |
Collapse
|
9
|
Piao L, Che N, Li H, Li M, Feng Y, Liu X, Kim S, Jin Y, Xuan Y. SETD8 promotes stemness characteristics and is a potential prognostic biomarker of gastric adenocarcinoma. Exp Mol Pathol 2020; 117:104560. [PMID: 33127342 DOI: 10.1016/j.yexmp.2020.104560] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/15/2020] [Accepted: 10/22/2020] [Indexed: 12/26/2022]
Abstract
SETD8 is a lysine methyltransferase containing an SET domain, which is involved in the carcinogenesis of many cancer types through monomethylation of the histone H4 lysine 20. However, its prognostic value and underlying mechanisms in gastric adenocarcinoma (GA) have not been extensively studied. Here, we assessed SETD8 expression and its relationship with clinicopathological parameters, cancer stemness-related proteins, cell cycle-related proteins, and PI3K/Akt pathway proteins in GA. SETD8 expression in GA tissues was correlated with the primary tumor stage, lymph node metastasis, tumor size, gross type, and clinical stage. SETD8 was an independent predictor of poor overall survival of patients with GA. Cox regression analysis showed that SETD8 is a potential biomarker of unfavorable clinical outcomes in patients with GA. Moreover, SETD8 overexpression was associated with cancer stemness-related genes, cell cycle-related genes, and PI3K/Akt/NF-κB pathway genes in clinical GA tissue samples. SETD8 silencing downregulated the expression of cancer stemness-associated genes (LSD1 and SOX2) and inhibited GA cell proliferation, spheroid formation, invasion, and migration. Additionally, LY294002 significantly reduced the expression of SETD8, pAkt-Ser473, pPI3K-p85, and NFκB-p65 in MKN74 and MKN28 cells. SETD8 may be a novel cancer stemness-associated protein and potential prognostic biomarker in GA.
Collapse
Affiliation(s)
- Lihua Piao
- Institute for Regenerative Medicine, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China; Department of Histology and Embryology, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China
| | - Nan Che
- Institute for Regenerative Medicine, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China; Department of Pathology, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China
| | - Haoyue Li
- Institute for Regenerative Medicine, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China; Department of Pathology, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China
| | - Mengxuan Li
- Institute for Regenerative Medicine, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China; Department of Anatomy, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China
| | - Ying Feng
- Institute for Regenerative Medicine, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China; Department of Pathology, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China
| | - Xingzhe Liu
- Institute for Regenerative Medicine, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China; Department of Pathology, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China
| | - Seokhyung Kim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University College of Medicine, Seoul 110-745, Republic of Korea
| | - Yu Jin
- Institute for Regenerative Medicine, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China; Department of Anatomy, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China.
| | - Yanhua Xuan
- Institute for Regenerative Medicine, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China; Department of Pathology, Yanbian University College of Medicine, Yanji 133002, Jilin Province, PR China.
| |
Collapse
|