1
|
Park MA, Gumpper-Fedus K, Krishna SG, Genilo-Delgado MC, Brantley S, Hart PA, Dillhoff ME, Gomez MF, Basinski TL, Mok SR, Luthra AK, Fleming JB, Mohammadi A, Centeno BA, Jiang K, Karolak A, Jeong D, Chen DT, Stewart PA, Teer JK, Cruz-Monserrate Z, Permuth JB. Molecular Pathway and Immune Profile Analysis of IPMN-Derived Versus PanIN-Derived Pancreatic Ductal Adenocarcinomas. Int J Mol Sci 2024; 25:13164. [PMID: 39684873 DOI: 10.3390/ijms252313164] [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: 09/23/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Intraductal papillary mucinous neoplasms (IPMN) are commonly detected pancreatic cysts that may transform into pancreatic ductal adenocarcinoma (PDAC). Predicting which IPMNs will progress to PDAC remains a clinical challenge. Moreover, identifying those clinically evident IPMNs for which a surveillance approach is best is a dire clinical need. Therefore, we aimed to identify molecular signatures that distinguished between PDAC with and without clinical evidence of an IPMN to identify novel molecular pathways related to IPMN-derived PDAC that could help guide biomarker development. Data from the Oncology Research Information Exchange Network (ORIEN) multi-institute sequencing project were utilized to analyze 66 PDAC cases from Moffitt Cancer Center and The Ohio State University Wexner Medical Center, for which tumor whole transcriptome sequencing datasets were generated. Cases were classified based on whether a tumor had originated from an IPMN (n = 16) or presumably through the pancreatic intraepithelial neoplasia (PanIN) pathway (n = 50). We then performed differential expression and pathway analysis using Gene-Set Enrichment Analysis (GSEA) and Pathway Analysis with Down-weighted Genes (PADOG) algorithms. We also analyzed immune profiles using the Tumor-Immune Microenvironment Deconvolution web portal for Bulk Transcriptomics (TIMEx). Both GSEA and TIMEx indicate that PanIN-derived PDAC tumors enrich inflammatory pathways (complement, hedgehog signaling, coagulation, inflammatory response, apical surface, IL-2/STAT5, IL-6/STAT3, EMT, KRAS signaling, apical junction, IFN-gamma, allograft rejection) and are comparatively richer in almost all immune cell types than those from IPMN-derived PDAC. IPMN-derived tumors were enriched for metabolic and energy-generating pathways (oxidative phosphorylation, unfolded protein response, pancreas beta cells, adipogenesis, fatty acid metabolism, protein secretion), and the most significantly upregulated genes (padj < 0.001) included mucin 2 (MUC2) and gastrokine-2 (GKN2). Further, the metabolic-linked gene signature enriched in the IPMN-derived samples is associated with a cluster of early-stage and long-survival (top 4th quartile) PDAC cases from The Cancer Genome Atlas (TCGA) expression database. Our data suggest that IPMN-derived and PanIN-derived PDACs differ in the expression of immune profiles and metabolic pathways. These initial findings warrant validation and follow-up to develop biomarker-based strategies for early PDAC detection and treatment.
Collapse
Affiliation(s)
- Margaret A Park
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Kristyn Gumpper-Fedus
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Somashekar G Krishna
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Maria C Genilo-Delgado
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Stephen Brantley
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Phil A Hart
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Mary E Dillhoff
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Maria F Gomez
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Toni L Basinski
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Shaffer R Mok
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Anjuli K Luthra
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jason B Fleming
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Amir Mohammadi
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Barbara A Centeno
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Kun Jiang
- Department of Pathology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Aleksandra Karolak
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Daniel Jeong
- Department of Radiology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Dung-Tsa Chen
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Paul A Stewart
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jamie K Teer
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology and Nutrition, The Ohio State University Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Jennifer B Permuth
- Department of Gastrointestinal (GI) Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL 33612, USA
| |
Collapse
|
2
|
Matos TL, Souza PFN, de Moraes MEA, Rabenhorst SHB, Mesquita FP, Montenegro RC. Molecular characterization and biomarker discovery in gastric cancer progression through transcriptome meta-analysis. Comput Biol Med 2024; 183:109276. [PMID: 39447404 DOI: 10.1016/j.compbiomed.2024.109276] [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: 08/13/2024] [Revised: 09/26/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
Gastric cancer (GC) is a leading cause of cancer-related deaths globally. It is a multifactorial, molecularly heterogeneous disease whose carcinogenic patterns are not yet well established, requiring the development of new tools for better understanding and identifying gastric carcinogenesis. From this point of view, this study aims to compare transcriptome profiles from The Cancer Genome Atlas Stomach Adenocarcinoma (TCGA-STAD) and a human-merged dataset to identify potential biomarkers and therapeutic targets. Principal component analysis (PCA) revealed shared and distinct gene expression patterns between datasets. Differential expression analysis identified key genes with altered expression across non-malignant and malignant samples. Six genes, including SERPINE1 and CLDN9, were significantly associated with patient survival. The findings underscore the molecular diversity of GC and highlight novel biomarkers for early diagnosis and therapeutic strategies. Further validation in clinical specimens is necessary.
Collapse
Affiliation(s)
- Thiago Loreto Matos
- Pharmacogenetics Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, 60430-160, Brazil
| | - Pedro Filho Noronha Souza
- Pharmacogenetics Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, 60430-160, Brazil
| | - Maria Elisabete Amaral de Moraes
- Pharmacogenetics Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, 60430-160, Brazil
| | - Silvia Helena Barem Rabenhorst
- Molecular Genetics Laboratory, Department of Pathology and Legal Medicine, Federal University of Ceará, Fortaleza, 60430-160, Brazil
| | - Felipe Pantoja Mesquita
- Pharmacogenetics Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, 60430-160, Brazil.
| | - Raquel Carvalho Montenegro
- Pharmacogenetics Laboratory, Drug Research and Development Center, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, 60430-160, Brazil
| |
Collapse
|
3
|
Yan Z, Yang J, Wei WT, Zhou ML, Mo DX, Wan X, Ma R, Wu MM, Huang JH, Liu YJ, Lv FH, Li MH. A time-resolved multi-omics atlas of transcriptional regulation in response to high-altitude hypoxia across whole-body tissues. Nat Commun 2024; 15:3970. [PMID: 38730227 PMCID: PMC11087590 DOI: 10.1038/s41467-024-48261-w] [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: 08/23/2023] [Accepted: 04/23/2024] [Indexed: 05/12/2024] Open
Abstract
High-altitude hypoxia acclimatization requires whole-body physiological regulation in highland immigrants, but the underlying genetic mechanism has not been clarified. Here we use sheep as an animal model for low-to-high altitude translocation. We generate multi-omics data including whole-genome sequences, time-resolved bulk RNA-Seq, ATAC-Seq and single-cell RNA-Seq from multiple tissues as well as phenotypic data from 20 bio-indicators. We characterize transcriptional changes of all genes in each tissue, and examine multi-tissue temporal dynamics and transcriptional interactions among genes. Particularly, we identify critical functional genes regulating the short response to hypoxia in each tissue (e.g., PARG in the cerebellum and HMOX1 in the colon). We further identify TAD-constrained cis-regulatory elements, which suppress the transcriptional activity of most genes under hypoxia. Phenotypic and transcriptional evidence indicate that antenatal hypoxia could improve hypoxia tolerance in offspring. Furthermore, we provide time-series expression data of candidate genes associated with human mountain sickness (e.g., BMPR2) and high-altitude adaptation (e.g., HIF1A). Our study provides valuable resources and insights for future hypoxia-related studies in mammals.
Collapse
Affiliation(s)
- Ze Yan
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ji Yang
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Wen-Tian Wei
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ming-Liang Zhou
- Sichuan Academy of Grassland Science, Chengdu, 611743, China
| | - Dong-Xin Mo
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xing Wan
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Rui Ma
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Mei-Ming Wu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jia-Hui Huang
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Ya-Jing Liu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Feng-Hua Lv
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Meng-Hua Li
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, 100193, China.
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
4
|
Zhao K, Chen L, Xie Y, Ren N, Li J, Zhai X, Zheng S, Liu K, Wang C, Qiu Q, Peng X, Wang W, Liu J, Che Q, Fan J, Hu H, Liu M. m6A/HOXA10-AS/ITGA6 axis aggravates oxidative resistance and malignant progression of laryngeal squamous cell carcinoma through regulating Notch and Keap1/Nrf2 pathways. Cancer Lett 2024; 587:216735. [PMID: 38369001 DOI: 10.1016/j.canlet.2024.216735] [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: 12/09/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 02/20/2024]
Abstract
As the second most prevalent malignant tumor of head and neck, laryngeal squamous cell carcinoma (LSCC) imposes a substantial health burden on patients worldwide. Within recent years, resistance to oxidative stress and N6-methyladenosine (m6A) of RNA have been proved to be significantly involved in tumorigenesis. In current study, we investigated the oncogenic role of m6A modified long non coding RNAs (lncRNAs), specifically HOXA10-AS, and its downstream signaling pathway in the regulation of oxidative resistance in LSCC. Bioinformatics analysis revealed that heightened expression of HOXA10-AS was associated with the poor prognosis in LSCC patients, and N (6)-Methyladenosine (m6A) methyltransferase-like 3 (METTL3) was identified as a factor in promoting m6A modification of HOXA10-AS and further intensify its RNA stability. Mechanistically, HOXA10-AS was found to play as a competitive endogenous RNA (ceRNA) by sequestering miR-29 b-3p and preventing its downregulation of Integrin subunit alpha 6 (ITGA6), ultimately enhancing the oxidative resistance of tumor cells and promoting the malignant progression of LSCC. Furthermore, our research elucidated the mechanism by which ITGA6 accelerates Keap1 proteasomal degradation via enhancing TRIM25 expression, leading to increased Nrf2 stability and exacerbating its aberrant activation. Additionally, we demonstrated that ITGA6 enhances γ-secretase-mediated Notch signaling activation, ultimately promoting RBPJ-induced TRIM25 transcription. The current study provides the evidence supporting the effect of m6A modified HOXA10-AS and its downstream miR-29 b-3p/ITGA6 axis on regulating oxidative resistance and malignant progression in LSCC through the Notch and Keap1/Nrf2 pathways, and proposed that targeting this axis holds promise as a potential therapeutic approach for treating LSCC.
Collapse
Affiliation(s)
- Kai Zhao
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572013, PR China
| | - Liwei Chen
- Department of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing 100853, PR China
| | - Yingli Xie
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572013, PR China; Medical School of Chinese PLA, Beijing, 100853, PR China
| | - Nan Ren
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572013, PR China
| | - Jianhui Li
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572013, PR China
| | - Xingyou Zhai
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572013, PR China
| | - Shikang Zheng
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572013, PR China
| | - Kun Liu
- Department of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing 100853, PR China
| | - Cheng Wang
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572013, PR China
| | - Qibing Qiu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, PR China
| | - Xin Peng
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572013, PR China
| | - Wenjia Wang
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572013, PR China; Medical School of Chinese PLA, Beijing, 100853, PR China
| | - Jinjing Liu
- Medical School of Chinese PLA, Beijing, 100853, PR China
| | - Qin Che
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572013, PR China
| | - Junda Fan
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572013, PR China
| | - Hai Hu
- Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, PR China.
| | - Mingbo Liu
- Department of Otolaryngology Head and Neck Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, 572013, PR China; Department of Otolaryngology Head and Neck Surgery, Chinese PLA General Hospital, Beijing 100853, PR China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, PR China.
| |
Collapse
|
5
|
Zhong S, Borlak J. Sex differences in the tumor promoting effects of tobacco smoke in a cRaf transgenic lung cancer disease model. Arch Toxicol 2024; 98:957-983. [PMID: 38245882 PMCID: PMC10861769 DOI: 10.1007/s00204-023-03671-5] [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: 09/05/2023] [Accepted: 12/14/2023] [Indexed: 01/23/2024]
Abstract
Tobacco smoke (TS) is the leading cause for lung cancer (LC), and female smokers are at a greater risk for LC. Yet, the underlying causes are unknown. We performed whole genome scans in TS exposed wild type and histologically characterized tumor lesions of cRaf transgenic mice. We constructed miRNA-gene and transcription factor-miRNA/gene regulatory networks and determined sex-specific gene regulations by evaluating hormone receptor activities. We validated the findings from TS exposed cRaf mice in a large cohort of smoking and never-smoking LC patients. When compared to males, TS prompted a sevenfold increase in tumor multiplicity in cRaf females. Genome-wide scans of tumor lesions identified 161 and 53 genes and miRNAs, which code for EGFR/MAPK signaling, cell proliferation, oncomirs and oncogenes, and 50% of DEGs code for immune response and tumor evasion. Outstandingly, in transgenic males, TS elicited upregulation of 20 tumor suppressors, some of which are the targets of the androgen and estrogen receptor. Conversely, in females, 18 tumor suppressors were downregulated, and five were specifically repressed by the estrogen receptor. We found TS to perturb the circadian clock in a sex-specific manner and identified a female-specific regulatory loop that consisted of the estrogen receptor, miR-22-3p and circadian genes to support LC growth. Finally, we confirmed sex-dependent tumor promoting effects of TS in a large cohort of LC patients. Our study highlights the sex-dependent genomic responses to TS and the interplay of circadian clock genes and hormone receptors in the regulation of oncogenes and oncomirs in LC growth.
Collapse
Affiliation(s)
- Shen Zhong
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Jürgen Borlak
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
6
|
Lu YY, Zhu CY, Ding YX, Wang B, Zhao SF, Lv J, Chen SM, Wang SS, Wang Y, Wang R, Qiu WS, Qi WW. Cepharanthine, a regulator of keap1-Nrf2, inhibits gastric cancer growth through oxidative stress and energy metabolism pathway. Cell Death Discov 2023; 9:450. [PMID: 38086844 PMCID: PMC10716385 DOI: 10.1038/s41420-023-01752-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 11/09/2024] Open
Abstract
Cepharanthine (CEP), a bioactive compound derived from Stephania Cephalantha Hayata, is cytotoxic to various malignancies. However, the underlying mechanism of gastric cancer is unknown. CEP inhibited the cellular activity of gastric cancer AGS, HGC27 and MFC cell lines in this study. CEP-induced apoptosis reduced Bcl-2 expression and increased cleaved caspase 3, cleaved caspase 9, Bax, and Bad expression. CEP caused a G2 cell cycle arrest and reduced cyclin D1 and cyclin-dependent kinases 2 (CDK2) expression. Meanwhile, it increased oxidative stress, decreased mitochondrial membrane potential, and enhanced reactive oxygen species (ROS) accumulation in gastric cancer cell lines. Mechanistically, CEP inhibited Kelch-like ECH-associated protein (Keap1) expression while activating NF-E2 related factor 2 (Nrf2) nuclear translocations, increasing transcription of Nrf2 target genes quinone oxidoreductase 1 (NQO1), heme oxygenase 1 (HMOX1), and glutamate-cysteine ligase modifier subunit (GCLM). Furthermore, a combined analysis of targeted energy metabolism and RNA sequencing revealed that CEP could alter the levels of metabolic substances such as D (+) - Glucose, D-Fructose 6-phosphate, citric acid, succinic acid, and pyruvic acid, thereby altering energy metabolism in AGS cells. In addition, CEP significantly inhibited tumor growth in MFC BALB/c nude mice in vivo, consistent with the in vitro findings. Overall, CEP can induce oxidative stress by regulating Nrf2/Keap1 and alter energy metabolism, resulting in anti-gastric cancer effects. Our findings suggest a potential application of CEP in gastric cancer treatment.
Collapse
Affiliation(s)
- Yang-Yang Lu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chun-Yang Zhu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yi-Xin Ding
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bing Wang
- Biomedical Centre, Qingdao University, Qingdao, China
| | - Shu-Fen Zhao
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Lv
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shu-Ming Chen
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Sha-Sha Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Rui Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wen-Sheng Qiu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Wei-Wei Qi
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
7
|
Kubota S, Pasri P, Okrathok S, Jantasaeng O, Rakngam S, Mermillod P, Khempaka S. Transcriptome analysis of the uterovaginal junction containing sperm storage tubules in heat-stressed breeder hens. Poult Sci 2023; 102:102797. [PMID: 37285691 PMCID: PMC10250161 DOI: 10.1016/j.psj.2023.102797] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/08/2023] [Accepted: 05/17/2023] [Indexed: 06/09/2023] Open
Abstract
Sperm storage tubules (SSTs) in the uterovaginal junction (UVJ) of the oviduct are major sites of sperm storage after artificial insemination or mating. Female birds may regulate sperm motility in the UVJ. Heat stress can decrease the reproductive ability of broiler breeder hens. However, its effects on UVJ remain unclear. Changes in gene expression aid in understanding heat stress-affected molecular mechanisms. Herein, we wanted to conduct a comparative transcriptomic analysis to identify the differentially expressed genes (DEGs) in the UVJ of breeder hens under thermoneutral (23°C) and heat stress (36°C for 6 h) conditions. The results indicated that cloacal temperatures and respiratory rates were significantly increased in heat-stressed breeder hens (P < 0.05). Total RNA was extracted from the hen UVJ tissues containing SSTs after heat exposure. Transcriptome analysis identified 561 DEGs, including 181 upregulated DEGs containing heat shock protein (HSP) transcripts and 380 downregulated DEGs containing immune-related genes, such as interleukin 4-induced 1, radical S-adenosyl methionine domain containing 2, and 2'-5'-oligoadenylate synthetase like, in heat-stressed hens. Gene Ontology analysis revealed the significantly enriched terms involving HSPs. Kyoto Encyclopedia of Genes and Genomes analysis identified 9 significant pathways, including the protein processing in endoplasmic reticulum (11 genes including HSPs), neuroactive ligand-receptor interaction (13 genes including luteinizing hormone/choriogonadotropin receptor), biosynthesis of amino acids (4 genes including tyrosine aminotransferase), ferroptosis (3 genes including heme oxygenase 1), and nitrogen metabolism (carbonic anhydrase [CA]-12 and CA6) pathways. Protein-protein interaction network analysis of DEGs revealed 2 large networks, one containing upregulated HSPs and the other containing downregulated interferon-stimulating genes. Overall, heat stress inhibits innate immunity in the UVJ tissues of broiler chickens, and heat-stressed chickens protect their cells by increasing the expression levels of HSPs. The identified genes are potential candidates for further exploration of the UVJ in heat-stressed hens. The identified molecular pathways and networks increase our understanding of the sperm storage reservoirs (UVJ containing SSTs) within the reproductive tract and may be used to prevent heat stress-induced fertility loss in breeder hens.
Collapse
Affiliation(s)
- Satoshi Kubota
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Phocharapon Pasri
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Supattra Okrathok
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Orapin Jantasaeng
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Sitthipong Rakngam
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Pascal Mermillod
- UMR de Physiologie de la Reproduction et des Comportements, National Research Institute for Agronomy, Food and Environment (INRAe), 37380 Nouzilly, France
| | - Sutisa Khempaka
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand.
| |
Collapse
|
8
|
Gao Y, Wang C, Jiang D, An G, Jin F, Zhang J, Han G, Cui C, Jiang P. New insights into the interplay between autophagy and oxidative and endoplasmic reticulum stress in neuronal cell death and survival. Front Cell Dev Biol 2022; 10:994037. [PMID: 36187470 PMCID: PMC9524158 DOI: 10.3389/fcell.2022.994037] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/30/2022] [Indexed: 12/03/2022] Open
Abstract
Autophagy is a dynamic process that maintains the normal homeostasis of cells by digesting and degrading aging proteins and damaged organelles. The effect of autophagy on neural tissue is still a matter of debate. Some authors suggest that autophagy has a protective effect on nerve cells, whereas others suggest that autophagy also induces the death of nerve cells and aggravates nerve injury. In mammals, oxidative stress, autophagy and endoplasmic reticulum stress (ERS) constitute important defense mechanisms to help cells adapt to and survive the stress conditions caused by physiological and pathological stimuli. Under many pathophysiological conditions, oxidative stress, autophagy and ERS are integrated and amplified in cells to promote the progress of diseases. Over the past few decades, oxidative stress, autophagy and ERS and their interactions have been a hot topic in biomedical research. In this review, we summarize recent advances in understanding the interactions between oxidative stress, autophagy and ERS in neuronal cell death and survival.
Collapse
Affiliation(s)
- Yahao Gao
- Clinical Medical School, Jining Medical University, Jining, China
| | - Changshui Wang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Di Jiang
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Gang An
- Clinical Medical School, Jining Medical University, Jining, China
| | - Feng Jin
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Junchen Zhang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Guangkui Han
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Changmeng Cui
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
- *Correspondence: Changmeng Cui, ; Pei Jiang,
| | - Pei Jiang
- Department of Clinical Pharmacy, Jining First People’s Hospital, Jining Medical University, Jining, China
- *Correspondence: Changmeng Cui, ; Pei Jiang,
| |
Collapse
|
9
|
CircOAS3 Regulates Keratinocyte Proliferation and Psoriatic Inflammation by Interacting with Hsc70 via the JNK/STAT3/NF-κB Signaling Pathway. Inflammation 2022; 45:1924-1935. [DOI: 10.1007/s10753-022-01664-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/02/2022] [Accepted: 03/14/2022] [Indexed: 02/07/2023]
|
10
|
Lv J, Xie M, Zhao S, Qiu W, Wang S, Cao M. Nestin is essential for cellular redox homeostasis and gastric cancer metastasis through the mediation of the Keap1-Nrf2 axis. Cancer Cell Int 2021; 21:603. [PMID: 34772403 PMCID: PMC8588700 DOI: 10.1186/s12935-021-02184-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/28/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Gastric cancer (GC) is a common malignancy of the digestive system. Antioxidant activity is regarded as a possible mechanism in ectopic cancer. Hence, oxidative stress regulation is being evaluated for cancer treatment. Previous research has demonstrated that Nestin is associated with antioxidative resistance via its modulation of the Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor erythroid 2-related factor 2 (Nrf2) pathway. METHODS We determined the role of Nestin-mediated redox homeostasis and tumor phenotypes in GC cells. RESULTS We found that the Nestin expression level was high in GC tissues and cell lines. Nestin knockdown in the GC cell lines SGC-7901 and MKN-45 reduced viability, induced apoptosis, decreased antioxidant enzyme generation, and repressed GC metastasis. Nestin binds to Keap1, resulting in Nrf2 degradation and influencing downstream gene expression. Nestin knockdown resulted in the downregulation of Nrf2 expression in GC cells. The restoration of Nrf2 expression or treatment with the Nrf2 activator sulforaphane counteracted the inhibitory effect of Nestin knockdown on the proliferation, migration, invasion, and antioxidant enzyme production in GC cells. Moreover, xenograft GC tumors exhibited a slower growth rate than those of the control group in vivo. CONCLUSIONS Taken together, these findings suggest that the Nestin-Keap1-Nrf2 axis confers oxidative stress resistance and plays an important role in the proliferation, migration, and invasion of GC cells.
Collapse
Affiliation(s)
- Jing Lv
- Department of Oncology, The Affiliated Hospital of Qingdao University/Key Laboratory of Cancer Molecular and Translational Research, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266000 Shandong China
| | - Meiqiang Xie
- Medical Oncology, Central People’s Hospital of Zhanjiang, Zhanjiang, 524000 Guangdong China
| | - Shufen Zhao
- Department of Oncology, The Affiliated Hospital of Qingdao University/Key Laboratory of Cancer Molecular and Translational Research, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266000 Shandong China
| | - Wensheng Qiu
- Department of Oncology, The Affiliated Hospital of Qingdao University/Key Laboratory of Cancer Molecular and Translational Research, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266000 Shandong China
| | - Shasha Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University/Key Laboratory of Cancer Molecular and Translational Research, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, 266000 Shandong China
| | - Manming Cao
- Department of Medical Oncology, Zhujiang Hospital, Southern Medical University, No.253 Gongye Road, Haizhu District, Guangzhou, 510282 Guangdong China
| |
Collapse
|
11
|
Liu Z, Zheng W, Liu Y, Zhou B, Zhang Y, Wang F. Targeting HSPA8 inhibits proliferation via downregulating BCR-ABL and enhances chemosensitivity in imatinib-resistant chronic myeloid leukemia cells. Exp Cell Res 2021; 405:112708. [PMID: 34157313 DOI: 10.1016/j.yexcr.2021.112708] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/20/2022]
Abstract
The resistance to tyrosine kinase inhibitors is currently a major problem for chronic myeloid leukemia (CML) treatment and HSPA8 is highly expressed and a hallmark of poor prognosis in several human cancers. However, its role in imatinib-resistant CML (IR-CML) cells remains undetermined. Here, we determined HSPA8 was overexpressed in IR-CML cells and associated with imatinib resistance. HSPA8 ablation could downregulate BCR-ABL/STAT5 and BCR-ABL/AKT signaling pathways, dramatically induce proliferation inhibition, autophagy, G0/G1 phase cell cycle arrest but not apoptosis in IR-CML cells. Significantly, HSPA8 ablation enhanced the antitumor activity of imatinib via promoting apoptosis in vitro and vivo. These findings unraveled that HSPA8 ablation inhibits proliferation via downregulating BCR-ABL and enhances chemosensitivity of imatinib in IR-CML cells, which investigate the role and molecular mechanism of HSPA8 in IR-CML cells and suggest that HSPA8 may be a potential target for IR-CML treatment.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, PR China.
| | - Wenlong Zheng
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, PR China
| | - Yuan Liu
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, PR China
| | - Binghe Zhou
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, PR China
| | - Yuqing Zhang
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, PR China
| | - Fan Wang
- Department of Clinical Laboratory, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, PR China
| |
Collapse
|
12
|
Zhao H, Huang C, Luo Y, Yao X, Hu Y, Wang M, Chen X, Zeng J, Hu W, Wang J, Li R, Yao X. A Correlation Study of Prognostic Risk Prediction for Colorectal Cancer Based on Autophagy Signature Genes. Front Oncol 2021; 11:595099. [PMID: 34168974 PMCID: PMC8218632 DOI: 10.3389/fonc.2021.595099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 04/26/2021] [Indexed: 01/07/2023] Open
Abstract
Autophagy plays a complex role in tumors, sometimes promoting cancer cell survival and sometimes inducing apoptosis, and its role in the colorectal tumor microenvironment is controversial. The purpose of this study was to investigate the prognostic value of autophagy-related genes (ARGs) in colorectal cancer. We identified 37 differentially expressed autophagy-related genes by collecting TCGA colorectal tumor transcriptome data. A single-factor COX regression equation was used to identify 11 key prognostic genes, and a prognostic risk prediction model was constructed based on multifactor COX analysis. We classified patients into high and low risk groups according to prognostic risk parameters (p <0.001) and determined the prognostic value they possessed by survival analysis and the receiver operating characteristic (ROC) curve in the training and test sets of internal tests. In a multifactorial independent prognostic analysis, this risk value could be used as an independent prognostic indicator (HR=1.167, 95% CI=1.078-1.264, P<0.001) and was a robust predictor without any staging interference. To make it more applicable to clinical procedures, we constructed nomogram based on risk parameters and parameters of key clinical characteristics. The area under ROC curve for 3-year and 5-year survival rates were 0.735 and 0.718, respectively. These will better enable us to monitor patient prognosis, thus improve patient outcomes.
Collapse
Affiliation(s)
- Haibi Zhao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China.,Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China
| | - Chengzhi Huang
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuwen Luo
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xiaoya Yao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China.,Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China
| | - Yong Hu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China.,Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China
| | - Muqing Wang
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Xin Chen
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China.,Medical College, Shantou University, Shantou, China
| | - Jun Zeng
- Department of General Surgery, Baoan Central Hospital, The Fifth Affiliated Hospital of Shen Zhen University, Shen Zhen, China
| | - Weixian Hu
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Junjiang Wang
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Rongjiang Li
- Department of General Surgery, Baoan Central Hospital, The Fifth Affiliated Hospital of Shen Zhen University, Shen Zhen, China
| | - Xueqing Yao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China.,Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Ganzhou Hospital (Ganzhou Municipal Hospital), Guangdong Provincial People's Hospital, Ganzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Medical College, Shantou University, Shantou, China
| |
Collapse
|
13
|
Chen R, Yang M, Huang W, Wang B. Cascades between miRNAs, lncRNAs and the NF-κB signaling pathway in gastric cancer (Review). Exp Ther Med 2021; 22:769. [PMID: 34055068 PMCID: PMC8145527 DOI: 10.3892/etm.2021.10201] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 04/28/2021] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer is a common digestive tract malignancy that is mainly treated with surgery combined with perioperative adjuvant chemoradiotherapy and biological targeted therapy. However, the diagnosis rate of early gastric cancer is low and both postoperative recurrence and distant metastasis are thorny problems. Therefore, it is essential to study the pathogenesis of gastric cancer and search for more effective means of treatment. The nuclear factor-κB (NF-κB) signaling pathway has an important role in the occurrence and development of gastric cancer and recent studies have revealed that microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are able to regulate this pathway through a variety of mechanisms. Understanding these interrelated molecular mechanisms is helpful in guiding improvements in gastric cancer treatment. In the present review, the functional associations between miRNAs, lncRNAs and the NF-κB signaling pathway in the occurrence, development and prognosis of gastric cancer were discussed. It was concluded that miRNAs and lncRNAs have complex relations with the NF-κB signaling pathway in gastric cancer. miRNAs/target genes/NF-κB/target proteins, signaling molecules/NF-κB/miRNAs/target genes, lncRNAs/miRNAs/NF-κB/genes or mRNAs, lncRNAs/target genes/NF-Κb/target proteins, and lncRNAs/NF-κB/target proteins cascades are all important factors in the occurrence and development of gastric cancer.
Collapse
Affiliation(s)
- Risheng Chen
- Department of Anesthesiology, Affiliated Nanhua Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Mingxiu Yang
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology (2016TP1015), Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Weiguo Huang
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology (2016TP1015), Cancer Research Institute, Hengyang Medical College of University of South China, Hengyang, Hunan 421001, P.R. China
| | - Baiyun Wang
- Department of Anesthesiology, Affiliated Nanhua Hospital of University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
14
|
Pu Y, Wen X, Jia Z, Xie Y, Luan C, Yu Y, Chen F, Chen P, Li D, Sun Y, Zhao J, Lv H. Association Between Polymorphisms in Gastric Cancer Related Genes and Risk of Gastric Cancer: A Case-Control Study. Front Mol Biosci 2021; 8:690665. [PMID: 34079823 PMCID: PMC8166284 DOI: 10.3389/fmolb.2021.690665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 04/30/2021] [Indexed: 11/13/2022] Open
Abstract
Gastric cancer has the second highest incidence among all the malignancies in China, just below lung cancer. Gastric cancer is likewise one of the main sources of cancer related passings. Gastric cancer therefore remains a huge threat to human health. The primary reason is absence of high sensitivity and specificity for early detection while the pathogenesis of GC is stayed muddled. During the last few decades, a lot of GC related genes have been identified. To find candidate GC related variant in these GC related genes, we conducted this case-control study. 29 tagSNPs located in 7 GC related genes were included. 228 gastric cancer patients and 299 healthy controls were enrolled. Significant differences were found between the genotype frequencies of EFNA1 rs4971066 polymorphism between gastric cancer patients and healthy controls. The result indicated that ephrin-A1 tagSNP rs4971066 GT/TT genotypes was significantly associated with reduced susceptibility of gastric cancer development.
Collapse
Affiliation(s)
- Yan Pu
- School of Medicine, Southeast University, Nanjing, China
| | - Xu Wen
- Department of General Surgery, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Zhangjun Jia
- Department of Clinical Laboratory, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Xie
- Department of Geriatrics, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Changxing Luan
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, China
| | - Youjia Yu
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, China
| | - Feng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, China
| | - Peng Chen
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, China
| | - Ding Li
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, China
| | - Yan Sun
- Department of Medical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Jian Zhao
- Department of Radiation Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Haiqin Lv
- School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
15
|
Rios J, Sequeida A, Albornoz A, Budini M. Chaperone Mediated Autophagy Substrates and Components in Cancer. Front Oncol 2021; 10:614677. [PMID: 33643916 PMCID: PMC7908825 DOI: 10.3389/fonc.2020.614677] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/14/2020] [Indexed: 12/15/2022] Open
Abstract
Chaperone-mediated autophagy (CMA) represents a specific way of lysosomal protein degradation and contrary to macro and microautophagy is independent of vesicles formation. The role of CMA in different physiopathological processes has been studied for several years. In cancer, alterations of the CMA principal components, Hsc70 and Lamp2A protein and mRNA levels, have been described in malignant cells. However, changes in the expression levels of these CMA components are not always associated with changes in CMA activity and their biological significance must be carefully interpreted case by case. The objective of this review is to discuss whether altering the CMA activity, CMA substrates or CMA components is accurate to avoid cancer progression. In particular, this review will discuss about the evidences in which alterations CMA components Lamp2A and Hsc70 are associated or not with changes in CMA activity in different cancer types. This analysis will help to better understand the role of CMA activity in cancer and to elucidate whether CMA can be considered as target for therapeutics. Further, it will help to define whether the attention of the investigation should be focused on Lamp2A and Hsc70 because they can have an independent role in cancer progression beyond of their participation in altered CMA activity.
Collapse
Affiliation(s)
- Javiera Rios
- Molecular and Cellular Pathology Laboratory, Dentistry Faculty, Institute in Dentistry Sciences, University of Chile, Santiago, Chile
| | - Alvaro Sequeida
- Molecular and Cellular Pathology Laboratory, Dentistry Faculty, Institute in Dentistry Sciences, University of Chile, Santiago, Chile
| | - Amelina Albornoz
- Fundación Ciencia & Vida, Santiago, Chile.,San Sebastian University, Santiago, Chile
| | - Mauricio Budini
- Molecular and Cellular Pathology Laboratory, Dentistry Faculty, Institute in Dentistry Sciences, University of Chile, Santiago, Chile.,Autophagy Research Center (ARC), Santiago, Chile
| |
Collapse
|
16
|
RNA Sequencing of Early-Stage Gastric Adenocarcinoma Reveals Multiple Activated Pathways and Novel Long Non-Coding RNAs in Patient Tissue Samples. Rep Biochem Mol Biol 2021; 9:478-489. [PMID: 33969142 DOI: 10.52547/rbmb.9.4.478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Background Gastric cancer is among the most common cancers worldwide that currently lacks effective diagnostic biomarkers and therapeutic targets. Next-generation RNA sequencing is a powerful tool that allows rapid and accurate transcriptome-wide profiling to detect differentially expressed transcripts involved in normal biological and pathological processes. Given the function of this technique, it has the potential to identify new molecular targets for the early diagnosis of disease, particularly in gastric adenocarcinoma. Methods In this study, whole-transcriptome analysis was performed with RNA sequencing on tumoral and non-tumoral tissue samples from patients with early-stage gastric cancer. Gene ontology and pathway enrichment analysis were used to determine the main function of the specific genes and pathways present in tissue samples. Results Analysis of the differentially expressed genes revealed 5 upregulated and 234 downregulated genes in gastric cancer tissues. Pathway enrichment analysis revealed significantly dysregulated signalling pathways, including those involved in gastric acid secretion, drug metabolism and transporters, molecular toxicology, O-linked glycosylation of mucins, immunotoxicity, metabolism of xenobiotics by cytochrome P450, and glycosylation. We also found novel downregulated non-coding RNAs present in gastric cancer tissues, including GATA6 antisense RNA 1, antisense to LYZ, antisense P4HB, overlapping ACER2, long intergenic non-protein coding RNA 2688 (LINC02688) and uncharacterized LOC25845 (PP7080). Conclusion The transcriptomic data found in this study illustrates the power of RNA-sequencing in discovering novel genes and tumorigenic pathways involved in human carcinogenesis. The anomalies present in these genes may serve as promising tools for the development of accurate diagnostic biomarkers for the detection of early-stage gastric cancer.
Collapse
|
17
|
Luan M, Shi SS, Shi DB, Liu HT, Ma RR, Xu XQ, Sun YJ, Gao P. TIPRL, a Novel Tumor Suppressor, Suppresses Cell Migration, and Invasion Through Regulating AMPK/mTOR Signaling Pathway in Gastric Cancer. Front Oncol 2020; 10:1062. [PMID: 32719745 PMCID: PMC7350861 DOI: 10.3389/fonc.2020.01062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/28/2020] [Indexed: 12/25/2022] Open
Abstract
Invasion and metastasis of gastric cancer after curative resection remain the most common lethal outcomes. However, our current understanding of the molecular mechanism underlying gastric cancer metastasis is far from complete. Herein, we identified TOR signaling pathway regulator (TIPRL) as a novel metastasis suppressor in gastric cancer through genome-wide gene expression profiling analysis using mRNA microarray. Decreased TIPRL expression was detected in clinical gastric cancer specimens, and low TIPRL expression was correlated with more-advanced TNM stage, distant metastasis, and poor clinical outcome. Moreover, TIPRL was identified as a direct target of miR-216a-5p and miR-383-5p. Functional study revealed that re-expression of TIPRL in gastric cancer cell lines suppressed their migratory and invasive capacities, whereas inverse effects were observed in TIPRL-deficient models. Mechanistically, TIPRL downstream effectors and signaling pathways were investigated using mRNA microarray. Gene expression profiling revealed that TIPRL could not modulate the downstream genes at transcriptional levels, thereby implying that the regulation might occur at the post-transcriptional levels. We further demonstrated that TIPRL induced phosphorylation/activation of AMPK, which in turn attenuated phosphorylation of mTOR, p70S6K, and 4E-BP1, thereby leading to inactivation of mTOR signaling and subsequent suppression of cell migration/invasion in gastric cancer. Taken together, TIPRL acts as a novel metastasis suppressor in gastric cancer, at least in part, through regulating AMPK/mTOR signaling, likely representing a promising target for new therapies in gastric cancer.
Collapse
Affiliation(s)
- Meng Luan
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shan-Shan Shi
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Cardiovascular Proteomics of Shandong Province, Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, China
| | - Duan-Bo Shi
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pathology, Qilu Hospital, Shandong University, Jinan, China
| | - Hai-Ting Liu
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ran-Ran Ma
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pathology, Qilu Hospital, Shandong University, Jinan, China
| | - Xiao-Qun Xu
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yu-Jing Sun
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pathology, Qilu Hospital, Shandong University, Jinan, China
| | - Peng Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pathology, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
18
|
Tripathi SK, Biswal BK. Piperlongumine, a potent anticancer phytotherapeutic: Perspectives on contemporary status and future possibilities as an anticancer agent. Pharmacol Res 2020; 156:104772. [PMID: 32283222 DOI: 10.1016/j.phrs.2020.104772] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 12/13/2022]
Abstract
Piperlongumine, a white to beige biologically active alkaloid/amide phytochemical, has high pharmacological relevance as an anticancer agent. Piperlongumine has several biological activities, including selective cytotoxicity against multiple cancer cells of different origins at a preclinical level. Several preclinical studies have documented the anticancer potential of piperlongumine through its targeting of multiple molecular mechanisms, such as cell cycle arrest, anti-angiogenesis, anti- invasive and anti-metastasis pathways, autophagy pathways, and intrinsic apoptotic pathways in vitro and in vivo. Mechanistically, piperlongumine inhibits cancer growth by resulting in the accumulation of intracellular reactive oxygen species, decreasing glutathione and chromosomal damage, or modulating key regulatory proteins, including PI3K, AKT, mTOR, NF-kβ, STATs, and cyclin D1. Furthermore, combined treatment with piperlongumine potentiates the anticancer activity of conventional chemotherapeutics and overcomes resistance to chemo- and radio- therapy. Nanoformulation of piperlongumine has been associated with increased aqueous solubility and bioavailability and lower toxicity, thus enhancing therapeutic efficacy in both preclinical and clinical settings. The current review highlights anticancer studies on the occurrence, chemical properties, chemopreventive mechanisms, toxicity, bioavailability, and pharmaceutical relevance of piperlongumine in vitro and in vivo.
Collapse
Affiliation(s)
- Surya Kant Tripathi
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha, 769008, India
| | - Bijesh Kumar Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha, 769008, India.
| |
Collapse
|