1
|
Li Y, Fan W, Lo TH, Jiang JX, Fish SR, Tomilov A, Chronopoulos A, Bansal V, Mozes G, Vancza L, Kunimoto K, Ye J, Becker L, Das S, Park H, Wei Y, Ranjbarvaziri S, Bernstein D, Ramsey J, Cortopassi G, Török NJ. P46Shc Inhibits Mitochondrial ACAA2 Thiolase, Exacerbating Mitochondrial Injury and Inflammation in Aging Livers. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00443-7. [PMID: 39733992 DOI: 10.1016/j.ajpath.2024.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/08/2024] [Accepted: 10/29/2024] [Indexed: 12/31/2024]
Abstract
Mitochondrial maladaptation and dysfunction contribute to the progression of metabolic dysfunction-associated steatohepatitis (MASH). The authors recently implicated the induction of Shc in progressive MASH during aging and the cytoplasmic p52Shc isoform in the activation of redox enzyme NOX2. The mitochondrial Shc isoform p46Shc was shown to repress acetyl-coenzyme A acyltransferase 2 (ACAA2) in vitro. ACAA2 is a key enzyme for lipid β-oxidation; however, the metabolic consequences of in vivo p46Shc induction were unknown. The authors generated p46Shc-inducible mice; these and littermate controls were aged and fed chow or fast-food diet (high-fat and high-fructose). p46Shc induction increased liver injury, inflammation, and lipid peroxidation. p46Shc overexpression did not significantly change liver triglycerides. On electron microscopy studies, mitochondria were swollen with aberrant cristae. p46Shc induction reduced mitochondrial oxygen consumption as measured by Oroboros, as well as suppressed the production of β-hydroxybutyrate, the central metabolite of therapeutic ketosis. Mitochondria exhibited increased production of reactive oxidative species. By contrast, the expression of dominant negative p46Shc reduced ACAA2 thiolase activity, improved β-oxidation, and reduced lipid peroxidation and production of reactive oxidative species. In summary, these studies support the concept that p46Shc induction in aging represses ACAA2, resulting in decreased mitochondrial β-oxidation and increased lipid peroxidation. Maintaining β-oxidation and ketogenesis could prevent liver injury, and targeting Shc-related maladaptive responses could be a successful therapeutic strategy in aging/MASH.
Collapse
Affiliation(s)
- Yuan Li
- Division of Gastroenterology and Hepatology, Stanford University, Palo Alto, California
| | - Weiguo Fan
- Division of Gastroenterology and Hepatology, Stanford University, Palo Alto, California; Palo Alto VA, Palo Alto, California
| | - Tzu-Han Lo
- Division of Gastroenterology and Hepatology, Stanford University, Palo Alto, California; Palo Alto VA, Palo Alto, California
| | - Joy X Jiang
- Division of Gastroenterology and Hepatology, UC Davis Medical Center, Sacramento, California
| | - Sarah R Fish
- Division of Gastroenterology and Hepatology, UC Davis Medical Center, Sacramento, California
| | - Alexey Tomilov
- Department of Molecular Biosciences, School of Veterinary Medicine, UC Davis, Davis, California
| | - Antonios Chronopoulos
- Division of Gastroenterology and Hepatology, Stanford University, Palo Alto, California; Palo Alto VA, Palo Alto, California
| | - Vidushi Bansal
- Division of Gastroenterology and Hepatology, Stanford University, Palo Alto, California; Palo Alto VA, Palo Alto, California
| | - Gergely Mozes
- Division of Gastroenterology and Hepatology, Stanford University, Palo Alto, California; Palo Alto VA, Palo Alto, California
| | - Lorand Vancza
- Division of Gastroenterology and Hepatology, Stanford University, Palo Alto, California; Palo Alto VA, Palo Alto, California
| | - Koshi Kunimoto
- Division of Gastroenterology and Hepatology, Stanford University, Palo Alto, California; Palo Alto VA, Palo Alto, California
| | - Jiayu Ye
- Division of Gastroenterology and Hepatology, Stanford University, Palo Alto, California; Palo Alto VA, Palo Alto, California
| | - Laren Becker
- Division of Gastroenterology and Hepatology, Stanford University, Palo Alto, California; Palo Alto VA, Palo Alto, California
| | - Suvarthi Das
- Division of Gastroenterology and Hepatology, Stanford University, Palo Alto, California; Palo Alto VA, Palo Alto, California
| | - Hyesuk Park
- Division of Gastroenterology and Hepatology, Stanford University, Palo Alto, California; Palo Alto VA, Palo Alto, California
| | - Yi Wei
- Division of Gastroenterology and Hepatology, Stanford University, Palo Alto, California; Palo Alto VA, Palo Alto, California
| | | | - Daniel Bernstein
- Pediatrics-Cardiology, Stanford University, Palo Alto, California
| | - Jon Ramsey
- Department of Molecular Biosciences, School of Veterinary Medicine, UC Davis, Davis, California
| | - Gino Cortopassi
- Department of Molecular Biosciences, School of Veterinary Medicine, UC Davis, Davis, California
| | - Natalie J Török
- Division of Gastroenterology and Hepatology, Stanford University, Palo Alto, California; Palo Alto VA, Palo Alto, California.
| |
Collapse
|
2
|
Prill M, Sardão VA, Sobczak M, Nowis D, Szymanski J, Wieckowski MR. p66Shc Protein-Oxidative Stress Sensor or Redox Enzyme: Its Potential Role in Mitochondrial Metabolism of Human Breast Cancer. Cancers (Basel) 2024; 16:3324. [PMID: 39409944 PMCID: PMC11476363 DOI: 10.3390/cancers16193324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
This work presents a comprehensive evaluation of the role of p66Shc protein in mitochondrial physiology in MDA-MB-231 breast cancer cells. The use of human breast cancer cell line MDA-MB-231 and its genetically modified clones (obtained with the use of the CRISPR-Cas9 technique), expressing different levels of p66Shc protein, allowed us to demonstrate how the p66Shc protein affects mitochondrial metabolism of human breast cancer cells. Changes in the level of p66Shc (its overexpression, and overexpressing of its Serine 36-mutated version, as well as the knockout of p66Shc) exert different effects in breast cancer cells. Interestingly, knocking out p66Shc caused significant changes observed mostly in mitochondrial bioenergetic parameters. We have shown that an MDA-MB-231 (which is a strong metastatic type of breast cancer) clone lacking p66Shc protein is characterized by a significant shift in the metabolic phenotype in comparison to other MDA-MB-231 clones. Additionally, this clone is significantly more vulnerable to doxorubicin treatment. We have proved that p66Shc adaptor protein in human breast cancer cells may exert a different role than in noncancerous cells (e.g., fibroblasts).
Collapse
Affiliation(s)
- Monika Prill
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology PAS, 02-093 Warsaw, Poland
- Cellular Immunotherapy Center, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
| | - Vilma A. Sardão
- CNC-Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-531 Coimbra, Portugal;
| | - Mateusz Sobczak
- Laboratory of Experimental Medicine, Faculty of Medicine, Medial University of Warsaw, 02-091 Warsaw, Poland (D.N.)
| | - Dominika Nowis
- Laboratory of Experimental Medicine, Faculty of Medicine, Medial University of Warsaw, 02-091 Warsaw, Poland (D.N.)
| | - Jedrzej Szymanski
- Laboratory of Imaging Tissue Structure and Function, Nencki Institute of Experimental Biology PAS, 02-093 Warsaw, Poland;
| | - Mariusz R. Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology PAS, 02-093 Warsaw, Poland
| |
Collapse
|
3
|
Pan Y, Chen X, Zhou H, Xu M, Li Y, Wang Q, Xu Z, Ren C, Liu L, Liu X. Identification and validation of SHC1 and FGFR1 as novel immune-related oxidative stress biomarkers of non-obstructive azoospermia. Front Endocrinol (Lausanne) 2024; 15:1356959. [PMID: 39391879 PMCID: PMC11466301 DOI: 10.3389/fendo.2024.1356959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/22/2024] [Indexed: 10/12/2024] Open
Abstract
Background Non-obstructive azoospermia (NOA) is a major contributor of male infertility. Herein, we used existing datasets to identify novel biomarkers for the diagnosis and prognosis of NOA, which could have great significance in the field of male infertility. Methods NOA datasets were obtained from the Gene Expression Omnibus (GEO) database. CIBERSORT was utilized to analyze the distributions of 22 immune cell populations. Hub genes were identified by applying weighted gene co-expression network analysis (WGCNA), machine learning methods, and protein-protein interaction (PPI) network analysis. The expression of hub genes was verified in external datasets and was assessed by receiver operating characteristic (ROC) curve analysis. Gene set enrichment analysis (GSEA) was applied to explore the important functions and pathways of hub genes. The mRNA-microRNA (miRNA)-transcription factors (TFs) regulatory network and potential drugs were predicted based on hub genes. Single-cell RNA sequencing data from the testes of patients with NOA were applied for analyzing the distribution of hub genes in single-cell clusters. Furthermore, testis tissue samples were obtained from patients with NOA and obstructive azoospermia (OA) who underwent testicular biopsy. RT-PCR and Western blot were used to validate hub gene expression. Results Two immune-related oxidative stress hub genes (SHC1 and FGFR1) were identified. Both hub genes were highly expressed in NOA samples compared to control samples. ROC curve analysis showed a remarkable prediction ability (AUCs > 0.8). GSEA revealed that hub genes were predominantly enriched in toll-like receptor and Wnt signaling pathways. A total of 24 TFs, 82 miRNAs, and 111 potential drugs were predicted based on two hub genes. Single-cell RNA sequencing data in NOA patients indicated that SHC1 and FGFR1 were highly expressed in endothelial cells and Leydig cells, respectively. RT-PCR and Western blot results showed that mRNA and protein levels of both hub genes were significantly upregulated in NOA testis tissue samples, which agree with the findings from analysis of the microarray data. Conclusion It appears that SHC1 and FGFR1 could be significant immune-related oxidative stress biomarkers for detecting and managing patients with NOA. Our findings provide a novel viewpoint for illustrating potential pathogenesis in men suffering from infertility.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Li Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoqiang Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
4
|
Guo Z, Cai C, Zhou K, Song L, Wang X, Chen D, Weng G, Huang S. SHC1 serves as a prognostic and immunological biomarker in clear cell renal cell carcinoma: a comprehensive bioinformatics and experimental analysis. Sci Rep 2024; 14:20150. [PMID: 39209911 PMCID: PMC11362144 DOI: 10.1038/s41598-024-70897-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
SHC1 plays a crucial regulatory role in various tumors, but its significance in predicting prognosis and immune response in clear cell renal cell carcinoma (ccRCC) is yet to be determined. In this study, we conducted a bioinformatics analysis of SHC1 expression, prognosis, and immunological functions in ccRCC using multiple databases. The association between SHC1 and immune infiltration, immune escape, and immunotherapy in ccRCC was systematically established. In addition, we validated our results by western blot of tumor and adjacent-tumor samples from nine ccRCC patients, as well as three renal carcinoma cell lines compared to a normal renal cell line. Our analysis revealed that the mRNA expression level of SHC1 in ccRCC tissues is significantly higher than that in normal tissues. Consistently, western blot experiment showed ccRCC tissues and cell lines exhibit higher protein levels that normal tissues and cell lines. Importantly, patients with low expression of SHC1 demonstrated a higher survival rate, indicating that SHC1 could serve as an independent prognostic factor for predicting survival in ccRCC. Additionally, high expression of SHC1 was associated with increased severe immune cell infiltration, enhanced immune escape, and higher immunotherapy scores. Hence, SHC1 emerges as a novel and easily detectable biomarker for predicting clinical outcomes, immune escape, and immunotherapy response in patients with ccRCC.
Collapse
MESH Headings
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Humans
- Kidney Neoplasms/genetics
- Kidney Neoplasms/immunology
- Kidney Neoplasms/pathology
- Kidney Neoplasms/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism
- Src Homology 2 Domain-Containing, Transforming Protein 1/genetics
- Computational Biology/methods
- Prognosis
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic
- Female
- Male
- Middle Aged
Collapse
Affiliation(s)
- Zhuangyu Guo
- Laboratory of Renal Carcinoma, Ningbo Urology and Nephrology Hospital, Urology and Nephrology Institute of Ningbo University, No.998 North Qianhe Road, Yinzhou District, Ningbo, 315100, Zhejiang, China
| | - Congbo Cai
- Laboratory of Renal Carcinoma, Ningbo Urology and Nephrology Hospital, Urology and Nephrology Institute of Ningbo University, No.998 North Qianhe Road, Yinzhou District, Ningbo, 315100, Zhejiang, China
| | - Kena Zhou
- School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lingmin Song
- Laboratory of Renal Carcinoma, Ningbo Urology and Nephrology Hospital, Urology and Nephrology Institute of Ningbo University, No.998 North Qianhe Road, Yinzhou District, Ningbo, 315100, Zhejiang, China
| | - Xue Wang
- Laboratory of Renal Carcinoma, Ningbo Urology and Nephrology Hospital, Urology and Nephrology Institute of Ningbo University, No.998 North Qianhe Road, Yinzhou District, Ningbo, 315100, Zhejiang, China
| | - Dongying Chen
- Department of Community Work, Ningbo Yinzhou No.3 Hospital, Ningbo, 315100, China
| | - Guobin Weng
- Laboratory of Renal Carcinoma, Ningbo Urology and Nephrology Hospital, Urology and Nephrology Institute of Ningbo University, No.998 North Qianhe Road, Yinzhou District, Ningbo, 315100, Zhejiang, China.
| | - Shuaishuai Huang
- Laboratory of Renal Carcinoma, Ningbo Urology and Nephrology Hospital, Urology and Nephrology Institute of Ningbo University, No.998 North Qianhe Road, Yinzhou District, Ningbo, 315100, Zhejiang, China.
| |
Collapse
|
5
|
Shimada H, Powell TL, Jansson T. Regulation of placental amino acid transport in health and disease. Acta Physiol (Oxf) 2024; 240:e14157. [PMID: 38711335 PMCID: PMC11162343 DOI: 10.1111/apha.14157] [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: 01/29/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/08/2024]
Abstract
Abnormal fetal growth, i.e., intrauterine growth restriction (IUGR) or fetal growth restriction (FGR) and fetal overgrowth, is associated with increased perinatal morbidity and mortality and is strongly linked to the development of metabolic and cardiovascular disease in childhood and later in life. Emerging evidence suggests that changes in placental amino acid transport may contribute to abnormal fetal growth. This review is focused on amino acid transport in the human placenta, however, relevant animal models will be discussed to add mechanistic insights. At least 25 distinct amino acid transporters with different characteristics and substrate preferences have been identified in the human placenta. Of these, System A, transporting neutral nonessential amino acids, and System L, mediating the transport of essential amino acids, have been studied in some detail. Importantly, decreased placental Systems A and L transporter activity is strongly associated with IUGR and increased placental activity of these two amino acid transporters has been linked to fetal overgrowth in human pregnancy. An array of factors in the maternal circulation, including insulin, IGF-1, and adiponectin, and placental signaling pathways such as mTOR, have been identified as key regulators of placental Systems A and L. Studies using trophoblast-specific gene targeting in mice have provided compelling evidence that changes in placental Systems A and L are mechanistically linked to altered fetal growth. It is possible that targeting specific placental amino acid transporters or their upstream regulators represents a novel intervention to alleviate the short- and long-term consequences of abnormal fetal growth in the future.
Collapse
Affiliation(s)
- Hiroshi Shimada
- Department of Obstetrics and Gynecology University of Colorado, Anschutz Medical Campus, Aurora, CO, US
- Departments of Obstetrics & Gynecology, Sapporo Medical University, Sapporo, Japan
| | - Theresa L Powell
- Department of Obstetrics and Gynecology University of Colorado, Anschutz Medical Campus, Aurora, CO, US
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, US
| | - Thomas Jansson
- Department of Obstetrics and Gynecology University of Colorado, Anschutz Medical Campus, Aurora, CO, US
| |
Collapse
|
6
|
Biondi G, Marrano N, Borrelli A, Rella M, D’Oria R, Genchi VA, Caccioppoli C, Cignarelli A, Perrini S, Laviola L, Giorgino F, Natalicchio A. The p66 Shc Redox Protein and the Emerging Complications of Diabetes. Int J Mol Sci 2023; 25:108. [PMID: 38203279 PMCID: PMC10778847 DOI: 10.3390/ijms25010108] [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: 10/31/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
Diabetes mellitus is a chronic metabolic disease, the prevalence of which is constantly increasing worldwide. It is often burdened by disabling comorbidities that reduce the quality and expectancy of life of the affected individuals. The traditional complications of diabetes are generally described as macrovascular complications (e.g., coronary heart disease, peripheral arterial disease, and stroke), and microvascular complications (e.g., diabetic kidney disease, retinopathy, and neuropathy). Recently, due to advances in diabetes management and the increased life expectancy of diabetic patients, a strong correlation between diabetes and other pathological conditions (such as liver diseases, cancer, neurodegenerative diseases, cognitive impairments, and sleep disorders) has emerged. Therefore, these comorbidities have been proposed as emerging complications of diabetes. P66Shc is a redox protein that plays a role in oxidative stress, apoptosis, glucose metabolism, and cellular aging. It can be regulated by various stressful stimuli typical of the diabetic milieu and is involved in various types of organ and tissue damage under diabetic conditions. Although its role in the pathogenesis of diabetes remains controversial, there is strong evidence regarding the involvement of p66Shc in the traditional complications of diabetes. In this review, we will summarize the evidence supporting the role of p66Shc in the pathogenesis of diabetes and its complications, focusing for the first time on the emerging complications of diabetes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Francesco Giorgino
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, 70124 Bari, Italy (M.R.); (R.D.); (V.A.G.)
| | | |
Collapse
|
7
|
He D, Tang H, Yang X, Liu X, Zhang Y, Shi J. Elaboration and validation of a prognostic signature associated with disulfidoptosis in lung adenocarcinoma, consolidated with integration of single-cell RNA sequencing and bulk RNA sequencing techniques. Front Immunol 2023; 14:1278496. [PMID: 37965333 PMCID: PMC10641741 DOI: 10.3389/fimmu.2023.1278496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/13/2023] [Indexed: 11/16/2023] Open
Abstract
Background Lung adenocarcinoma (LUAD), the predominant subtype of non-small cell lung cancer (NSCLC), remains a pervasive global public health concern. Disulfidoptosis, a nascent form of regulated cell death (RCD), presents an emerging field of inquiry. Currently, investigations into disulfidoptosis are in their initial stages. Our undertaking sought to integrate single-cell RNA sequencing (scRNA-seq) in conjunction with traditional bulk RNA sequencing (bulk RNA-seq) methodologies, with the objective of delineating genes associated with disulfidoptosis and subsequently prognosticating the clinical outcomes of LUAD patients. Methods Initially, we conducted an in-depth examination of the cellular composition disparities existing between LUAD and normal samples using scRNA-seq data sourced from GSE149655. Simultaneously, we scrutinized the expression patterns of disulfidoptosis-associated gene sets across diverse cell types. Subsequently, leveraging the bulk RNA-seq data, we formulated disulfidoptosis-related prognostic risk signatures (DRPS) employing LASSO-Cox regression. This was accomplished by focusing on genes implicated in disulfidoptosis that exhibited differential expression within endothelial cells (ECs). Sequentially, the robustness and precision of the DRPS model were rigorously verified through both internal and external validation datasets. In parallel, we executed single-cell trajectory analysis to delve into the differentiation dynamics of ECs. Concluding our study, we undertook a comprehensive investigation encompassing various facets. These included comparative assessments of enrichment pathways, clinicopathological parameters, immune cell abundance, immune response-associated genes, impacts of immunotherapy, and drug predictions among distinct risk cohorts. Results The scrutiny of scRNA-seq data underscored discernible disparities in cellular composition between LUAD and normal samples. Furthermore, disulfidoptosis-associated genes exhibited marked discrepancies within endothelial cells (ECs). Consequently, we formulated the Disulfidoptosis-Related Prognostic Signature (DRPS) to facilitate prognostic prediction. The prognostic nomogram based on the risk score effectively demonstrated DRPS's robust capacity to prognosticate survival outcomes. This assertion was corroborated by rigorous assessments utilizing both internal and external validation sets, thus affirming the commendable predictive accuracy and enduring stability of DRPS. Functional enrichment analysis shed light on the significant correlation of DRPS with pathways intrinsic to the cell cycle. Subsequent analysis unveiled correlations between DRPS and gene mutations characteristic of LUAD, as well as indications of an immunosuppressive status. Through drug prediction, we explored potential therapeutic agents for low-risk patients. Concluding our investigation, qRT-PCR experiments confirmed the heightened expression levels of EPHX1, LDHA, SHC1, MYO6, and TLE1 in lung cancer cell lines.
Collapse
Affiliation(s)
- Dabao He
- Department of Laboratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Hengfeng Tang
- Department of Laboratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Xiaoling Yang
- Department of Laboratory Medicine, Shenzhen Baoan District Songgang People’s Hospital, Shenzhen, China
| | - Xiaohong Liu
- Department of Oncology, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Yipeng Zhang
- Department of Laboratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Junzhu Shi
- Department of Laboratory Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China
| |
Collapse
|
8
|
Mo JS, Lamichhane S, Yun KJ, Chae SC. MicroRNA 452 regulates SHC1 expression in human colorectal cancer and colitis. Genes Genomics 2023; 45:1295-1304. [PMID: 37523129 DOI: 10.1007/s13258-023-01432-3] [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: 03/21/2023] [Accepted: 07/10/2023] [Indexed: 08/01/2023]
Abstract
BACKGROUND Human microRNA 452 (MIR452) has been linked to both colorectal cancer (CRC) tissues and dextran sulfate sodium (DSS)-induced colitis. OBJECTIVE We analyzed the correlation between MIR452 and its putative target gene in human CRC cells and in mouse colitis tissues. METHODS Luciferase reporter assay confirmed that Src homologous and collagen adaptor protein 1 (SHC1) is a direct target of MIR452. Furthermore, the expression of proteins or mRNA was assessed by immunohistochemical analysis, Western blot, or quantitative RT-PCR (qRT-PCR). RESULTS We found that MIR452 has a potential binding site at 3'-UTR of SHC1. Likewise, MIR452 or siSHC1 transfection dramatically reduced the level of cellular SHC1 in CRC cells. The expression of SHC1 was frequently downregulated in both human CRC tissues and mouse colitis tissues. In CRC cells, we demonstrated that MIR452 regulated the expression of genes involved in the SHC1-mediated KRAS-MAPK signal transduction pathways. CONCLUSION These findings suggest a potential defense mechanism in which MIR452 regulation of the adaptor protein SHC1 maintains cellular homeostasis during carcinogenesis or chronic inflammation. Therefore, MIR452 may have therapeutic value for human early-stage CRC and colitis.
Collapse
Affiliation(s)
- Ji-Su Mo
- Department of Pathology, School of Medicine, Wonkwang University, Iksan, Chonbuk, 54538, Republic of Korea
- Digestive Disease Research Institute, Wonkwang University, Iksan, Chonbuk, 54538, Republic of Korea
| | - Santosh Lamichhane
- Department of Pathology, School of Medicine, Wonkwang University, Iksan, Chonbuk, 54538, Republic of Korea
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Ki-Jung Yun
- Department of Pathology, School of Medicine, Wonkwang University, Iksan, Chonbuk, 54538, Republic of Korea
| | - Soo-Cheon Chae
- Department of Pathology, School of Medicine, Wonkwang University, Iksan, Chonbuk, 54538, Republic of Korea.
- Digestive Disease Research Institute, Wonkwang University, Iksan, Chonbuk, 54538, Republic of Korea.
| |
Collapse
|
9
|
Powell AM, Edwards NA, Hunter H, Kiser P, Watson AJ, Cumming RC, Betts DH. Deletion of p66Shc Dysregulates ERK and STAT3 Activity in Mouse Embryonic Stem Cells, Enhancing Their Naive-Like Self-Renewal in the Presence of Leukemia Inhibitory Factor. Stem Cells Dev 2023; 32:434-449. [PMID: 37183401 DOI: 10.1089/scd.2022.0283] [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] [Indexed: 05/16/2023] Open
Abstract
The ShcA adapter protein is necessary for early embryonic development. The role of ShcA in development is primarily attributed to its 52 and 46 kDa isoforms that transduce receptor tyrosine kinase signaling through the extracellular signal regulated kinase (ERK). During embryogenesis, ERK acts as the primary signaling effector, driving fate acquisition and germ layer specification. P66Shc, the largest of the ShcA isoforms, has been observed to antagonize ERK in several contexts; however, its role during embryonic development remains poorly understood. We hypothesized that p66Shc could act as a negative regulator of ERK activity during embryonic development, antagonizing early lineage commitment. To explore the role of p66Shc in stem cell self-renewal and differentiation, we created a p66Shc knockout murine embryonic stem cell (mESC) line. Deletion of p66Shc enhanced basal ERK activity, but surprisingly, instead of inducing mESC differentiation, loss of p66Shc enhanced the expression of core and naive pluripotency markers. Using pharmacologic inhibitors to interrogate potential signaling mechanisms, we discovered that p66Shc deletion permits the self-renewal of naive mESCs in the absence of conventional growth factors, by increasing their responsiveness to leukemia inhibitory factor (LIF). We discovered that loss of p66Shc enhanced not only increased ERK phosphorylation but also increased phosphorylation of Signal transducer and activator of transcription in mESCs, which may be acting to stabilize their naive-like identity, desensitizing them to ERK-mediated differentiation cues. These findings identify p66Shc as a regulator of both LIF-mediated ESC pluripotency and of signaling cascades that initiate postimplantation embryonic development and ESC commitment.
Collapse
Affiliation(s)
- Andrew M Powell
- Department of Biology, The University of Western Ontario, London, Canada
| | - Nicole A Edwards
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Canada
| | - Hailey Hunter
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Canada
| | - Patti Kiser
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Canada
| | - Andrew J Watson
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Canada
- Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Canada
- Genetics and Development Division, The Children's Health Research Institute, Lawson Health Research Institute, London, Canada
| | - Robert C Cumming
- Department of Biology, The University of Western Ontario, London, Canada
- Genetics and Development Division, The Children's Health Research Institute, Lawson Health Research Institute, London, Canada
| | - Dean H Betts
- Department of Biology, The University of Western Ontario, London, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Canada
- Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Canada
- Genetics and Development Division, The Children's Health Research Institute, Lawson Health Research Institute, London, Canada
| |
Collapse
|
10
|
Zaric BL, Macvanin MT, Isenovic ER. Free radicals: Relationship to Human Diseases and Potential Therapeutic applications. Int J Biochem Cell Biol 2023; 154:106346. [PMID: 36538984 DOI: 10.1016/j.biocel.2022.106346] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/06/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
Reactive species are highly-reactive enzymatically, or non-enzymatically produced compounds with important roles in physiological and pathophysiological cellular processes. Although reactive species represent an extensively researched topic in biomedical sciences, many aspects of their roles and functions remain unclear. This review aims to systematically summarize findings regarding the biochemical characteristics of various types of reactive species and specify the localization and mechanisms of their production in cells. In addition, we discuss the specific roles of free radicals in cellular physiology, focusing on the current lines of research that aim to identify the reactive oxygen species-initiated cascades of reactions resulting in adaptive or pathological cellular responses. Finally, we present recent findings regarding the therapeutic modulations of intracellular levels of reactive oxygen species, which may have substantial significance in developing novel agents for treating several diseases.
Collapse
Affiliation(s)
- Bozidarka L Zaric
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| | - Mirjana T Macvanin
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Esma R Isenovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
11
|
Zhao J, Li J, Li G, Chen M. The role of mitochondria-associated membranes mediated ROS on NLRP3 inflammasome in cardiovascular diseases. Front Cardiovasc Med 2022; 9:1059576. [PMID: 36588561 PMCID: PMC9794868 DOI: 10.3389/fcvm.2022.1059576] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/01/2022] [Indexed: 12/15/2022] Open
Abstract
Reactive oxygen species (ROS) metabolism is essential for the homeostasis of cells. Appropriate production of ROS is an important signaling molecule, but excessive ROS production can damage cells. ROS and ROS-associated proteins can act as damage associated molecular pattern molecules (DAMPs) to activate the NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome in cardiovascular diseases. Previous studies have shown that there are connected sites, termed mitochondria-associated membranes (MAMs), between mitochondria and the endoplasmic reticulum. In cardiovascular disease progression, MAMs play multiple roles, the most important of which is the ability to mediate ROS generation, which further activates the NLPR3 inflammasome, exacerbating the progression of disease. In this review, the following topics will be covered: 1. Molecular structures on MAMs that can mediate ROS generation; 2. Specific mechanisms of molecule-mediated ROS generation and the molecules' roles in cardiovascular disease, 3. The effects of MAMs-mediated ROS on the NLRP3 inflammasome in cardiovascular disease. The purpose of this review is to provide a basis for subsequent clinical treatment development.
Collapse
Affiliation(s)
- Jiahao Zhao
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, China,Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Junli Li
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Guoyong Li
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, China,Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China,Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Mao Chen
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, China,Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Mao Chen
| |
Collapse
|
12
|
Zhang YH, Peng F, Zhang L, Kang K, Yang M, Chen C, Yu H. LONG NONCODING RNA UPREGULATES ADAPTER SHCA PROTEIN EXPRESSION TO PROMOTE COGNITIVE IMPAIRMENT AFTER CARDIAC ARREST AND RESUSCITATION. Shock 2022; 58:169-178. [PMID: 35953462 DOI: 10.1097/shk.0000000000001964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT Aim: More patients are resuscitated from cardiac arrest and cardiopulmonary resuscitation (CA/CPR) due to advances in medical care. However, the burden now lies with post-cardiac arrest cognitive impairment in CA/CPR survivors. Based on our previous study, we aimed to further confirm the correlation between the long noncoding RNA-promoting ShcA (lncRNA-PS)/Src homology and collagen A (ShcA) axis and CA/CPR-induced cognitive impairment in molecular, cellular, and tissue levels. Methods and Results: The in vivo experiments were based on a mouse model of CA/CPR, while oxygen-glucose deprivation and reoxygenation was used as a cell model in vitro. Conditional ShcA suppression in neurons of the hippocampal CA1 region was achieved by cyclization recombinase of bacteriophage P1 recognizing DNA fragment locus of x-over P1 site (Cre/LoxP recombination system). Genetic manipulation of HT22 was achieved by lentivirus targeting lncRNA-PS and ShcA. Neurological function score was remarkably decreased, and cognitive function was affected after restoration of spontaneous circulation. LncRNA-PS and ShcA overexpression after CA/CPR, mainly happened in neurons of hippocampal CA1 region, was observed by in situ hybridization and immunofluorescence. Neuronal ShcA knockdown in hippocampal CA1 region before CA/CPR attenuated cognitive impairment after CA/CPR. ShcA deficiency protected HT22 cell line against oxygen-glucose deprivation and reoxygenation by inhibiting inflammation and apoptosis. In vitro upregulation of lncRNA-PS elevated ShcA expression, which was reversed by knockdown of ShcA. Conclusions: This study revealed that lncRNA-PS/ShcA axis is critically involved in the pathogenesis of cognitive impairment after CA/CPR. By inhibiting ShcA expression in neurons of the hippocampal CA1 region could improve the survival outcomes in mice after CA/CPR.
Collapse
|
13
|
SHC1 Promotes Lung Cancer Metastasis by Interacting with EGFR. JOURNAL OF ONCOLOGY 2022; 2022:3599832. [PMID: 35706930 PMCID: PMC9192283 DOI: 10.1155/2022/3599832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/27/2022] [Accepted: 05/05/2022] [Indexed: 11/27/2022]
Abstract
The study aims to explore the biological function of SHC1 in the development and progression of lung cancer. Meanwhile, the effect of SHC1 and EGFR on lung cancer was analyzed. The expression of SHC1 in lung cancer and adjacent tissues was analyzed by bioinformatics and immunohistochemistry. Meanwhile, the relationship between SHC1 expression and prognosis was analyzed. SHC1 overexpression and knockdown cell lines were constructed by overexpression plasmid and knockdown plasmid. Cell proliferation was detected by CCK-8. Cell invasion was detected by transwell. Apoptosis was detected by TUNEL. Interaction between SHC1 and EGFR was detected. The expression of SHC1 in lung adenocarcinoma tissues was significantly higher than that in paracancer tissues. Lung cancer patients with high SHC1 expression have a poor prognosis. The proliferation and invasion of SHC1 decreased with SHC1 knockout but increased after overexpression. EGFR may be a key interaction protein of SHC1. Overexpression of EGFR increases the oncogenic effect of SHC1. In conclusion, SHC1 plays a carcinogenic role in lung cancer. EGFR expression was significantly correlated with SHC1 and maybe a key interaction protein of SHC1. SHC1 interacts with EGFR to form a protein complex, which may be a new target for lung cancer metastasis.
Collapse
|
14
|
Haslem L, Hays JM, Hays FA. p66Shc in Cardiovascular Pathology. Cells 2022; 11:cells11111855. [PMID: 35681549 PMCID: PMC9180016 DOI: 10.3390/cells11111855] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 02/06/2023] Open
Abstract
p66Shc is a widely expressed protein that governs a variety of cardiovascular pathologies by generating, and exacerbating, pro-apoptotic ROS signals. Here, we review p66Shc’s connections to reactive oxygen species, expression, localization, and discuss p66Shc signaling and mitochondrial functions. Emphasis is placed on recent p66Shc mitochondrial function discoveries including structure/function relationships, ROS identity and regulation, mechanistic insights, and how p66Shc-cyt c interactions can influence p66Shc mitochondrial function. Based on recent findings, a new p66Shc mitochondrial function model is also put forth wherein p66Shc acts as a rheostat that can promote or antagonize apoptosis. A discussion of how the revised p66Shc model fits previous findings in p66Shc-mediated cardiovascular pathology follows.
Collapse
Affiliation(s)
- Landon Haslem
- Biochemistry and Molecular Biology Department, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.H.); (J.M.H.)
| | - Jennifer M. Hays
- Biochemistry and Molecular Biology Department, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.H.); (J.M.H.)
| | - Franklin A. Hays
- Biochemistry and Molecular Biology Department, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (L.H.); (J.M.H.)
- Stephenson Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Correspondence:
| |
Collapse
|
15
|
Chen J, Gao G, Li L, Ding J, Chen X, Lei J, Long H, Wu L, Long X, He L, Shen Y, Yang J, Lu Y, Sun Y. Pan-Cancer Study of SHC-Adaptor Protein 1 (SHC1) as a Diagnostic, Prognostic and Immunological Biomarker in Human Cancer. Front Genet 2022; 13:817118. [PMID: 35601500 PMCID: PMC9115805 DOI: 10.3389/fgene.2022.817118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/15/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Recent studies highlight the carcinogenesis role of SHC-adaptor protein 1 (SHC1) in cancer initiation, development, and progression. However, its aberrant expression, diagnostic and prognostic value remain unknown in a variety of tumors. Methods: The SHC1 expression profiles were analyzed using GTEx database, TCGA database, Oncomine and CPTAC database. The survival analysis was conducted using GEPIA2, Kaplan-Meier Plotter, UALCAN, and PrognoScan. The diagnostic values of SHC1 were calculated with the “pROC” package in R software. The genetic alteration of SHC1 and mutations were analyzed using cBioPortal. TIMER2 was employed to estimate the correlations between SHC1 expression and tumor-infiltrating immune cells in the TCGA cohort. Enrichment analysis of SHC1 was conducted using the R package “clusterProfiler.” Results: SHC1 was ubiquitously highly expressed and closely associated with worse prognosis of multiple major cancer types (all p < 0.05). Further, SHC1 gene mutations were strongly linked to poor OS and DFS in SKCM (all p < 0.05). An enhanced phosphorylation level of SHC1 at the S139 site was observed in clear cell RCC. Additionally, the results revealed SHC1 expression was strongly linked to TMB, MMRs, MSI, TAMs, DNA methylation, m6A RNA methylation, tumor-associated immune infiltration, and immune checkpoints in multiple cancers (all p < 0.05). In addition, the results of the ROC analysis indicated the SHC1 exhibited strong diagnostic capability for KICH (AUC = 0.92), LIHC (AUC = 0.95), and PAAD (AUC = 0.95). Finally, enrichment analysis indicated that SHC1 may potentially involve in the regulation of numerous signaling pathways in cancer metabolism and protein phosphorylation-related functions. Conclusions: These findings highlight that SHC1 plays an important role in the tumor immune microenvironment, and SHC1 has been identified to have prognostic and diagnostic value in multiple cancers. Thus, SHC1 is a potential target for cancer immunotherapy and effective prognostic and diagnostic biomarker.
Collapse
Affiliation(s)
- Jianlin Chen
- Departments of Clinical Laboratory, Key Laboratory of medical molecular diagnostics of Liuzhou, Key Laboratory for nucleic acid molecular diagnosis and application of Guangxi health and wellness Commission, Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
| | - Gan Gao
- Departments of Clinical Laboratory of Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, China
| | - Limin Li
- Departments of Clinical Laboratory of Liuzhou People's Hospital, Liuzhou, China
| | - Junping Ding
- Departments of Clinical Laboratory, Key Laboratory of medical molecular diagnostics of Liuzhou, Key Laboratory for nucleic acid molecular diagnosis and application of Guangxi health and wellness Commission, Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
| | - Xianhua Chen
- Departments of Clinical Laboratory, Key Laboratory of medical molecular diagnostics of Liuzhou, Key Laboratory for nucleic acid molecular diagnosis and application of Guangxi health and wellness Commission, Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
| | - Jianfei Lei
- People’s Hospital of Rong’an County, Liuzhou, China
| | - Haihua Long
- Departments of Clinical Laboratory, Key Laboratory of medical molecular diagnostics of Liuzhou, Key Laboratory for nucleic acid molecular diagnosis and application of Guangxi health and wellness Commission, Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
| | - Lihua Wu
- Departments of Clinical Laboratory, Key Laboratory of medical molecular diagnostics of Liuzhou, Key Laboratory for nucleic acid molecular diagnosis and application of Guangxi health and wellness Commission, Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
| | - Xin Long
- Departments of Clinical Laboratory, Key Laboratory of medical molecular diagnostics of Liuzhou, Key Laboratory for nucleic acid molecular diagnosis and application of Guangxi health and wellness Commission, Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
| | - Lian He
- People’s Hospital of Rong’an County, Liuzhou, China
| | - Yongqi Shen
- Departments of Clinical Laboratory, Key Laboratory of medical molecular diagnostics of Liuzhou, Key Laboratory for nucleic acid molecular diagnosis and application of Guangxi health and wellness Commission, Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
| | | | - Yonggang Lu
- Departments of Clinical Laboratory, Key Laboratory of medical molecular diagnostics of Liuzhou, Key Laboratory for nucleic acid molecular diagnosis and application of Guangxi health and wellness Commission, Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
- *Correspondence: Yonggang Lu, ; Yifan Sun,
| | - Yifan Sun
- Departments of Clinical Laboratory, Key Laboratory of medical molecular diagnostics of Liuzhou, Key Laboratory for nucleic acid molecular diagnosis and application of Guangxi health and wellness Commission, Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, China
- *Correspondence: Yonggang Lu, ; Yifan Sun,
| |
Collapse
|
16
|
Modulating binding affinity, specificity and configurations by multivalent interactions. Biophys J 2022; 121:1868-1880. [PMID: 35450827 DOI: 10.1016/j.bpj.2022.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/20/2021] [Accepted: 04/14/2022] [Indexed: 11/22/2022] Open
Abstract
Biological functions of proteins rely on their specific interactions with binding partners. Many proteins contain multiple domains, which can bind to their targets that often have more than one binding site, resulting in multivalent interactions. While it has been shown that multivalent interactions play an crucial role in modulating binding affinity and specificity, other potential effects of multivalent interactions are less explored. Here, we developed a broadly applicable transfer matrix formalism and used it to investigate the binding of two-domain ligands to targets with multiple binding sites. We show that 1) ligands with two specific binding domains can drastically boost both the binding affinity and specificity and down-shift the working concentration range, compared to single-domain ligands, 2) the presence of a positive domain-domain cooperativity or containing a non-specific binding domain can down-shift the working concentration range of ligands by increasing the binding affinity without compromising the binding specificity, 3) the configuration of the bound ligands has a strong concentration dependence, providing important insights into the physical origin of phase-separation processes taking place in living cells. In line with previous studies, our results suggest that multivalent interactions are utilized by cells for highly efficient regulation of target binding involved in a diverse range of cellular processes such as signal transduction, gene transcription, antibody-antigen recognition.
Collapse
|
17
|
de Souza PPC, Henning P, Lerner UH. Stimulation of Osteoclast Formation by Oncostatin M and the Role of WNT16 as a Negative Feedback Regulator. Int J Mol Sci 2022; 23:3287. [PMID: 35328707 PMCID: PMC8953253 DOI: 10.3390/ijms23063287] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/09/2022] [Accepted: 03/15/2022] [Indexed: 02/05/2023] Open
Abstract
Oncostatin M (OSM), which belongs to the IL-6 family of cytokines, is the most potent and effective stimulator of osteoclast formation in this family, as assessed by different in vitro assays. Osteoclastogenesis induced by the IL-6 type of cytokines is mediated by the induction and paracrine stimulation of the osteoclastogenic cytokine receptor activator of nuclear factor κ-B ligand (RANKL), expressed on osteoblast cell membranes and targeting the receptor activator of nuclear factor κ-B (RANK) on osteoclast progenitor cells. The potent effect of OSM on osteoclastogenesis is due to an unusually robust induction of RANKL in osteoblasts through the OSM receptor (OSMR), mediated by a JAK-STAT/MAPK signaling pathway and by unique recruitment of the adapter protein Shc1 to the OSMR. Gene deletion of Osmr in mice results in decreased numbers of osteoclasts and enhanced trabecular bone caused by increased trabecular thickness, indicating that OSM may play a role in physiological regulation of bone remodeling. However, increased amounts of OSM, either through administration of recombinant protein or of adenoviral vectors expressing Osm, results in enhanced bone mass due to increased bone formation without any clear sign of increased osteoclast numbers, a finding which can be reconciled by cell culture experiments demonstrating that OSM can induce osteoblast differentiation and stimulate mineralization of bone nodules in such cultures. Thus, in vitro studies and gene deletion experiments show that OSM is a stimulator of osteoclast formation, whereas administration of OSM to mice shows that OSM is not a strong stimulator of osteoclastogenesis in vivo when administered to adult animals. These observations could be explained by our recent finding showing that OSM is a potent stimulator of the osteoclastogenesis inhibitor WNT16, acting in a negative feedback loop to reduce OSM-induced osteoclast formation.
Collapse
Affiliation(s)
- Pedro P. C. de Souza
- The Innovation in Biomaterials Laboratory, School of Dentistry, Federal University of Goiás, Goiânia 74690-900, Brazil;
| | - Petra Henning
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden;
| | - Ulf H. Lerner
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Osteoporosis Centre and Centre for Bone and Arthritis Research at the Sahlgrenska Academy, University of Gothenburg, 413 45 Gothenburg, Sweden;
| |
Collapse
|
18
|
Mousavi S, Khazeei Tabari MA, Bagheri A, Samieefar N, Shaterian N, Kelishadi R. The Role of p66Shc in Diabetes: A Comprehensive Review from Bench to Bedside. J Diabetes Res 2022; 2022:7703520. [PMID: 36465704 PMCID: PMC9715346 DOI: 10.1155/2022/7703520] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 11/27/2022] Open
Abstract
It is well-documented that diabetes is an inflammatory and oxidative disease, with an escalating global burden. Still, there is no definite treatment for diabetes or even prevention of its harmful complications. Therefore, understanding the molecular pathways associated with diabetes might help in finding a solution. p66Shc is a member of Shc family proteins, and it is considered as an oxidative stress sensor and regulator in cells. There are inconsistent data about the role of p66Shc in inducing diabetes, but accumulating evidence supports its role in the pathogenesis of diabetes-related complications, including macro and microangiopathies. There is growing hope that by understanding and targeting molecular pathways involved in this network, prevention of diabetes or its complications would be achievable. This review provides an overview about the role of p66Shc in the development of diabetes and its complications.
Collapse
Affiliation(s)
- SeyedehFatemeh Mousavi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, Mazandaran University of Medical Sciences, Mazandaran, Iran
- USERN Office, Mazandaran University of Medical Sciences, Mazandaran, Iran
| | - Alireza Bagheri
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Noosha Samieefar
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- USERN Office, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Negar Shaterian
- Student Research Committee, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
- USERN Office, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Roya Kelishadi
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
- USERN Office, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
19
|
Jelinek HF, Helf C, Khalaf K. Human SHC-transforming protein 1 and its isoforms p66shc: A novel marker for prediabetes. J Diabetes Investig 2021; 12:1881-1889. [PMID: 33759377 PMCID: PMC8504898 DOI: 10.1111/jdi.13551] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 03/02/2021] [Accepted: 03/18/2021] [Indexed: 12/15/2022] Open
Abstract
AIMS Prediabetes is a multifactorial condition. Current guidelines for diabetes screening recommend either the use of glycated hemoglobin (HbA1c), or blood glucose level (BGL). This research aimed to identify if p66shc a component of the Human SHC-Transforming Protein 1 (Shc1), a mitochondrial associated oxidative stress biomarker, is significantly altered in patients with elevated BGL. Furthermore, we evaluated if inflammatory and oxidative stress markers, such as p66shc, are a useful addition to the regularly used biomarkers to increase sensitivity for identification of prediabetes. METHODS All participants attended the Diabetic Health Screening at Charles Sturt University (CSU), Australia. The cross-sectional clinical study collected demographic and clinical variables from 346 participants and classified into control or prediabetes based on fasting BGL. Blood and urine samples were analyzed for oxidative stress and inflammation markers. Logistic regression was used to compare multidimensional diagnostic models for prediabetes, including p66shc/Shc1, to the current HbA1c-only model in terms of sensitivity, specificity and predictive accuracy. Significance was set at P ≤ 0.05. RESULTS A significant decrease of p66shc/Shc1 was determined in prediabetes compared to controls (P ≤ 0.05). HbA1c testing resulted in an accuracy of 62%, while adding p66shc and triglycerides increased predictive accuracy to 88.05%. When HbA1c was omitted and Shc1 was combined with 8-hydroxy-2'-deoxyguanosine (8-OHdG) and monocyte chemo-attractant protein-1 (MCP-1), a predictive accuracy of 89.5% was achieved. CONCLUSION Our findings showed a major improvement of sensitivity to identify prediabetes by including oxidative stress and inflammatory biomarkers underlining beneficial diagnostic information, which most likely improves prevention and early treatment options in prediabetes.
Collapse
Affiliation(s)
- Herbert F Jelinek
- Department of Biomedical EngineeringKhalifa UniversityAbu DhabiUnited Arab Emirates
- Health Engineering Innovation CenterKhalifa UniversityAbu DhabiUnited Arab Emirates
- Biotechnology CenterKhalifa UniversityAbu DhabiUnited Arab Emirates
| | | | - Kinda Khalaf
- Department of Biomedical EngineeringKhalifa UniversityAbu DhabiUnited Arab Emirates
- Health Engineering Innovation CenterKhalifa UniversityAbu DhabiUnited Arab Emirates
| |
Collapse
|
20
|
Batty MJ, Chabrier G, Sheridan A, Gage MC. Metabolic Hormones Modulate Macrophage Inflammatory Responses. Cancers (Basel) 2021; 13:cancers13184661. [PMID: 34572888 PMCID: PMC8467249 DOI: 10.3390/cancers13184661] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/31/2021] [Accepted: 09/13/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Macrophages are a type of immune cell which play an important role in the development of cancer. Obesity increases the risk of cancer and obesity also causes disruption to the normal levels of hormones that are produced to coordinate metabolism. Recent research now shows that these metabolic hormones also play important roles in macrophage immune responses and so through macrophages, disrupted metabolic hormone levels may promote cancer. This review article aims to highlight and summarise these recent findings so that the scientific community may better understand how important this new area of research is, and how these findings can be capitalised on for future scientific studies. Abstract Macrophages are phagocytotic leukocytes that play an important role in the innate immune response and have established roles in metabolic diseases and cancer progression. Increased adiposity in obese individuals leads to dysregulation of many hormones including those whose functions are to coordinate metabolism. Recent evidence suggests additional roles of these metabolic hormones in modulating macrophage inflammatory responses. In this review, we highlight key metabolic hormones and summarise their influence on the inflammatory response of macrophages and consider how, in turn, these hormones may influence the development of different cancer types through the modulation of macrophage functions.
Collapse
|
21
|
Jiang JX, Tomilov A, Montgomery C, Hui CK, Török NJ, Cortopassi G. Shc inhibitor idebenone ameliorates liver injury and fibrosis in dietary NASH in mice. J Biochem Mol Toxicol 2021; 35:e22876. [PMID: 34369032 DOI: 10.1002/jbt.22876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 07/20/2021] [Accepted: 07/24/2021] [Indexed: 12/20/2022]
Abstract
Shc expression rises in human nonalcoholic steatohepatitis (NASH) livers, and Shc-deficient mice are protected from NASH-thus Shc inhibition could be a novel therapeutic strategy for NASH. Idebenone was recently identified as the first small-molecule Shc inhibitor drug. We tested idebenone in the fibrotic methionine-choline deficient (MCD) diet and the metabolic fast food diet (FFD) mouse models of NASH. In the fibrotic MCD NASH model, idebenone reduced Shc expression and phosphorylation in peripheral blood mononuclear cells and Shc expression in the liver; decreased serum alanine aminotransferase and aspartate aminotransferase; and attenuated liver fibrosis as observed by quantitative polymerase chain reaction (qPCR) and hydroxyproline quantification. In the metabolic FFD model, idebenone administration improved insulin resistance, and reduced inflammation and fibrosis shown with qPCR, hydroxyproline measurement, and histology. Thus, idebenone ameliorates NASH in two mouse models. As an approved drug with a benign safety profile, Idebenone could be a reasonable human NASH therapy.
Collapse
Affiliation(s)
- Joy X Jiang
- Department of Molecular Biosciences, School of Veterinary Medicine, Davis, California, USA
| | - Alexey Tomilov
- Department of Molecular Biosciences, School of Veterinary Medicine, Davis, California, USA
| | - Claire Montgomery
- Department of Molecular Biosciences, School of Veterinary Medicine, Davis, California, USA
| | - Chun Kui Hui
- Department of Molecular Biosciences, School of Veterinary Medicine, Davis, California, USA
| | - Natalie J Török
- Gastroenterology and Hepatology, Stanford University, Palo Alto, California, USA.,VA Palo Alto, Palo Alto, California, USA
| | - Gino Cortopassi
- Department of Molecular Biosciences, School of Veterinary Medicine, Davis, California, USA
| |
Collapse
|
22
|
Wang Q, Chang H, Shen Q, Li Y, Xing D. Photobiomodulation therapy for thrombocytopenia by upregulating thrombopoietin expression via the ROS-dependent Src/ERK/STAT3 signaling pathway. J Thromb Haemost 2021; 19:2029-2043. [PMID: 33501731 DOI: 10.1111/jth.15252] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Chemotherapy-induced thrombocytopenia (CIT) can increase the risk of bleeding, which may delay or prevent the administration of anticancer treatment schedules. Photobiomodulation therapy (PBMT), a non-invasive physical treatment, has been proposed to improve thrombocytopenia; however, its underlying regulatory mechanism is not fully understood. OBJECTIVE To further investigate the mechanism of thrombopoietin (TPO) in megakaryocytopoiesis and thrombopoiesis. METHODS Multiple approaches such as western blotting, cell transfection, flow cytometry, and animal studies were utilized to explore the effect and mechanism of PBMT on thrombopoiesis. RESULTS PBMT prevented a severe drop in platelet count by increasing platelet production, and then ameliorated CIT. Mechanistically, PBMT significantly upregulated hepatic TPO expression in a thrombocytopenic mouse model, which promoted megakaryocytopoiesis and thrombopoiesis. The levels of TPO mRNA and protein increased by PBMT via the Src/ERK/STAT3 signaling pathway in hepatic cells. Furthermore, the generation of the reactive oxygen species was responsible for PBMT-induced activation of Src and its downstream target effects. CONCLUSIONS Our research suggests that PBMT is a promising therapeutic strategy for the treatment of CIT.
Collapse
Affiliation(s)
- Qiuhong Wang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Haocai Chang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Qi Shen
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Yonghua Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Da Xing
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| |
Collapse
|
23
|
Yoshizawa R, Umeki N, Yamamoto A, Okada M, Murata M, Sako Y. p52Shc regulates the sustainability of ERK activation in a RAF-independent manner. Mol Biol Cell 2021; 32:1838-1848. [PMID: 34260260 PMCID: PMC8684710 DOI: 10.1091/mbc.e21-01-0007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
p52SHC (SHC) and GRB2 are adaptor proteins involved in the RAS/MAPK (ERK) pathway mediating signals from cell-surface receptors to various cytoplasmic proteins. To further examine their roles in signal transduction, we studied the translocation of fluorescently labeled SHC and GRB2 to the cell surface, caused by the activation of ERBB receptors by heregulin (HRG). We simultaneously evaluated activated ERK translocation to the nucleus. Unexpectedly, the translocation dynamics of SHC were sustained when those of GRB2 were transient. The sustained localization of SHC positively correlated with the sustained nuclear localization of ERK, which became more transient after SHC knockdown. SHC-mediated PI3K activation was required to maintain the sustainability of the ERK translocation regulating MEK but not RAF. In cells overexpressing ERBB1, SHC translocation became transient, and the HRG-induced cell fate shifted from a differentiation to a proliferation bias. Our results indicate that SHC and GRB2 functions are not redundant but that SHC plays the critical role in the temporal regulation of ERK activation.
Collapse
Affiliation(s)
- Ryo Yoshizawa
- Cellular Informatics Lab, RIKEN, Wako, Saitama 351-0198, Japan.,Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Komaba, Meguro, Tokyo 153-8902, Japan
| | - Nobuhisa Umeki
- Cellular Informatics Lab, RIKEN, Wako, Saitama 351-0198, Japan
| | | | - Mariko Okada
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan.,Center for Drug Design and Research, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki 567-0085, Japan
| | - Masayuki Murata
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Komaba, Meguro, Tokyo 153-8902, Japan
| | - Yasushi Sako
- Cellular Informatics Lab, RIKEN, Wako, Saitama 351-0198, Japan
| |
Collapse
|
24
|
Jacobs NR, Norton PA. Role of chromosome 1q copy number variation in hepatocellular carcinoma. World J Hepatol 2021; 13:662-672. [PMID: 34239701 PMCID: PMC8239492 DOI: 10.4254/wjh.v13.i6.662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/13/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023] Open
Abstract
Chromosome 1q often has been observed to be amplified in hepatocellular carcinoma. This review summarizes literature reports of multiple genes that have been proposed as possible 1q amplification drivers. These largely fall within 1q21-1q23. In addition, publicly available copy number alteration data from The Cancer Genome Atlas project were used to identify additional candidate genes involved in carcinogenesis. The most frequent location for gene amplification was 1q22, consistent with the results of the literature search. The genes TPM3 and NUF2 were found to be candidates whose amplification and/or mRNA up-regulation was most highly associated with poorer hepatocellular carcinoma outcomes.
Collapse
Affiliation(s)
- Nathan R Jacobs
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| | - Pamela A Norton
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102, United States
| |
Collapse
|
25
|
Mushtaq U, Bashir M, Nabi S, Khanday FA. Epidermal growth factor receptor and integrins meet redox signaling through P66shc and Rac1. Cytokine 2021; 146:155625. [PMID: 34157521 DOI: 10.1016/j.cyto.2021.155625] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 05/23/2021] [Accepted: 06/08/2021] [Indexed: 12/24/2022]
Abstract
This review examines the concerted role of Epidermal Growth Factor Receptor (EGFR) and integrins in regulating Reactive oxygen species (ROS) production through different signaling pathways. ROS as such are not always deleterious to the cells but they also act as signaling molecules, that regulates numerous indespensible physiological fuctions of life. Many adaptor proteins, particularly Shc and Grb2, are involved in mediating the downstream signaling pathways stimulated by EGFR and integrins. Integrin-induced activation of EGFR and subsequent tyrosine phosphorylation of a class of acceptor sites on EGFR leads to alignment and tyrosine phosphorylation of Shc, PLCγ, the p85 subunit of PI-3 K, and Cbl, followed by activation of the downstream targets Erk and Akt/PKB. Functional interactions between these receptors result in the activation of Rac1 via these adaptor proteins, thereby leading to Reactive Oxygen Species. Both GF and integrin activation can produce oxidants independently, however synergistically there is increased ROS generation, suggesting a mutual cooperation between integrins and GFRs for redox signalling. The ROS produced further promotes feed-forward stimulation of redox signaling events such as MAPK activation and gene expression. This relationship has not been reviewed previously. The literature presented here can have multiple implications, ranging from looking at synergistic effects of integrin and EGFR mediated signaling mechanisms of different proteins to possible therapeutic interventions operated by these two receptors. Furthermore, such mutual redox regulation of crosstalk between EGFR and integrins not only add to the established models of pathological oxidative stress, but also can impart new avenues and opportunities for targeted antioxidant based therapeutics.
Collapse
Affiliation(s)
- Umar Mushtaq
- Department of Biotechnology, University of Kashmir, Srinagar, JK 190006, India; Department of Biotechnology, Central University of Kashmir, Ganderbal, JK 191201, India
| | - Muneesa Bashir
- Department of Biotechnology, University of Kashmir, Srinagar, JK 190006, India; Department of Higher Education, Government of Jammu & Kashmir, 190001, India
| | - Sumaiya Nabi
- Department of Biochemistry, University of Kashmir, Srinagar, JK 190006, India
| | - Firdous A Khanday
- Department of Biotechnology, University of Kashmir, Srinagar, JK 190006, India.
| |
Collapse
|
26
|
Liang Y, Lei Y, Du M, Liang M, Liu Z, Li X, Gao Y. The increased expression and aberrant methylation of SHC1 in non-small cell lung cancer: Integrative analysis of clinical and bioinformatics databases. J Cell Mol Med 2021; 25:7039-7051. [PMID: 34117717 PMCID: PMC8278126 DOI: 10.1111/jcmm.16717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/14/2022] Open
Abstract
Despite the previous evidence showing that SHC adaptor protein 1 (SHC1) could encode three distinct isoforms (p46SHC, p52SHC and p66SHC) that function in different activities such as regulating life span and Ras activation, the precise underlying role of SHC1 in lung cancer also remains obscure. In this study, we firstly found that SHC1 expression was up‐regulated both in lung adenocarcinoma (LUAD) and in lung squamous cell carcinoma (LUSC) tissues. Furthermore, compared to patients with lower SHC1 expression, LUAD patients with higher expression of SHC1 had poorer overall survival (OS). Moreover, higher expression of SHC1 was also associated with worse OS in patients with stages 1 and 2 but not stage 3 lung cancer. Significantly, the analysis showed that SHC1 methylation level was associated with OS in lung cancer patients. It seemed that the methylation level at specific probes within SHC1 showed negative correlations with SHC1 expression both in LUAD and in LUSC tissues. The LUAD and LUSC patients with hypermethylated SHC1 at cg12473916 and cg19356022 probes had a longer OS. Therefore, it is reasonable to conclude that SHC1 has a potential clinical significance in LUAD and LUSC patients.
Collapse
Affiliation(s)
- Yicheng Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yangyang Lei
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Minjun Du
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mei Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zixu Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xingkai Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yushun Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
27
|
Shyrokova EY, Prassolov VS, Spirin PV. The Role of the MCTS1 and DENR Proteins in Regulating the Mechanisms Associated with Malignant Cell Transformation. Acta Naturae 2021; 13:98-105. [PMID: 34377560 PMCID: PMC8327141 DOI: 10.32607/actanaturae.11181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 09/28/2020] [Indexed: 02/05/2023] Open
Abstract
The mutations associated with malignant cell transformation are believed to disrupt the expression of a significant number of normal, non-mutant genes. The proteins encoded by these genes are involved in the regulation of many signaling pathways that are responsible for differentiation and proliferation, as well as sensitivity to apoptotic signals, growth factors, and cytokines. Abnormalities in the balance of signaling pathways can lead to the transformation of a normal cell, which results in tumor formation. Detection of the target genes and the proteins they encode and that are involved in the malignant transformation is one of the major evolutions in anti-cancer biomedicine. Currently, there is an accumulation of data that shed light on the role of the MCTS1 and DENR proteins in oncogenesis.
Collapse
Affiliation(s)
- E. Y. Shyrokova
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, Moscow, 119991 Russia
- Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Moscow Region, 141701 Russia
| | - V. S. Prassolov
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, Moscow, 119991 Russia
| | - P. V. Spirin
- Engelhardt Institute of Molecular Biology, Russian Academy of Science, Moscow, 119991 Russia
| |
Collapse
|
28
|
Yoshizawa R, Umeki N, Yamamoto A, Murata M, Sako Y. Biphasic spatiotemporal regulation of GRB2 dynamics by p52SHC for transient RAS activation. Biophys Physicobiol 2021; 18:1-12. [PMID: 33665062 PMCID: PMC7902154 DOI: 10.2142/biophysico.bppb-v18.001] [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: 11/24/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022] Open
Abstract
RTK-RAS-MAPK systems are major signaling pathways for cell fate decisions. Among the several RTK species, it is known that the transient activation of ERK (MAPK) stimulates cell proliferation, whereas its sustained activation induces cell differentiation. In both instances however, RAS activation is transient, suggesting that the strict temporal regulation of its activity is critical in normal cells. RAS on the cytoplasmic side of the plasma membrane is activated by SOS through the recruitment of GRB2/SOS complex to the RTKs that are phosphorylated after stimulation with growth factors. The adaptor protein GRB2 recognizes phospho-RTKs both directly and indirectly via another adaptor protein, SHC. We here studied the regulation of GRB2 recruitment under the SHC pathway using single-molecule imaging and fluorescence correlation spectroscopy in living cells. We stimulated MCF7 cells with a differentiation factor, heregulin, and observed the translocation, complex formation, and phosphorylation of cell signaling molecules including GRB2 and SHC. Our results suggest a biphasic regulation of the GRB2/SOS-RAS pathway by SHC: At the early stage (<10 min) of stimulation, SHC increased the amplitude of RAS activity by increasing the association sites for the GRB2/SOS complex on the plasma membrane. At the later stage however, SHC suppressed RAS activation and sequestered GRB2 molecules from the membrane through the complex formation in the cytoplasm. The latter mechanism functions additively to other mechanisms of negative feedback regulation of RAS from MEK and/or ERK to complete the transient activation dynamics of RAS.
Collapse
Affiliation(s)
- Ryo Yoshizawa
- Cellular Informatics Lab, RIKEN, Wako, Saitama 351-0198, Japan.,Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Meguro-ku, Tokyo 153-8902, Japan
| | - Nobuhisa Umeki
- Cellular Informatics Lab, RIKEN, Wako, Saitama 351-0198, Japan
| | | | - Masayuki Murata
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Meguro-ku, Tokyo 153-8902, Japan
| | - Yasushi Sako
- Cellular Informatics Lab, RIKEN, Wako, Saitama 351-0198, Japan
| |
Collapse
|
29
|
Khuda-Bukhsh AR, Saha SK, Das S, Saha SS. Molecular approaches toward targeted cancer therapy with some food plant products: On the role of antioxidants and immune microenvironment. Cancer 2021. [DOI: 10.1016/b978-0-12-819547-5.00018-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
30
|
Loureiro CA, Pinto FR, Barros P, Matos P, Jordan P. A SYK/SHC1 pathway regulates the amount of CFTR in the plasma membrane. Cell Mol Life Sci 2020; 77:4997-5015. [PMID: 31974654 PMCID: PMC11105000 DOI: 10.1007/s00018-020-03448-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 12/06/2019] [Accepted: 01/02/2020] [Indexed: 11/24/2022]
Abstract
Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene cause the recessive genetic disease cystic fibrosis, where the chloride transport across the apical membrane of epithelial cells mediated by the CFTR protein is impaired. CFTR protein trafficking to the plasma membrane (PM) is the result of a complex interplay between the secretory and membrane recycling pathways that control the number of channels present at the membrane. In addition, the ion transport activity of CFTR at the PM is modulated through post-translational protein modifications. Previously we described that spleen tyrosine kinase (SYK) phosphorylates a specific tyrosine residue in the nucleotide-binding domain 1 domain and this modification can regulate the PM abundance of CFTR. Here we identified the underlying biochemical mechanism using peptide pull-down assays followed by mass spectrometry. We identified in bronchial epithelial cells that the adaptor protein SHC1 recognizes tyrosine-phosphorylated CFTR through its phosphotyrosine-binding domain and that the formation of a complex between SHC1 and CFTR is induced at the PM in the presence of activated SYK. The depletion of endogenous SHC1 expression was sufficient to promote an increase in CFTR at the PM of these cells. The results identify a SYK/SHC1 pathway that regulates the PM levels of CFTR channels, contributing to a better understanding of how CFTR-mediated chloride secretion is regulated.
Collapse
Affiliation(s)
- Cláudia Almeida Loureiro
- Department of Human Genetics, National Health Institute 'Dr. Ricardo Jorge', Avenida Padre Cruz, 1649-016, Lisbon, Portugal
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Francisco R Pinto
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Patrícia Barros
- Department of Human Genetics, National Health Institute 'Dr. Ricardo Jorge', Avenida Padre Cruz, 1649-016, Lisbon, Portugal
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Paulo Matos
- Department of Human Genetics, National Health Institute 'Dr. Ricardo Jorge', Avenida Padre Cruz, 1649-016, Lisbon, Portugal
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
- Department of Chemistry and Biochemistry, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Peter Jordan
- Department of Human Genetics, National Health Institute 'Dr. Ricardo Jorge', Avenida Padre Cruz, 1649-016, Lisbon, Portugal.
- BioISI-Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal.
| |
Collapse
|
31
|
Protective Effects of ShcA Protein Silencing for Photothrombotic Cerebral Infarction. Transl Stroke Res 2020; 12:866-878. [PMID: 33242144 DOI: 10.1007/s12975-020-00874-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 12/26/2022]
Abstract
Reactive oxygen species (ROS) exacerbate stroke-induced cell damage. We found that ShcA, a protein that regulates ROS, is highly expressed in a Rose Bengal photothrombosis model. We investigated whether ShcA is essential for mitophagy in ROS-induced cellular damage and determined whether ROS exacerbate mitochondrial dysfunction via ShcA protein expression. Ischemic stroke was generated by Rose Bengal photothrombosis in mice. To silence ShcA protein expression in the mouse brain, ShcA-targeting siRNA-encapsulated nanoparticles were intrathecally injected into the cisterna magna. Upon staining with antibodies against ShcA counterpart caspase-3 or NeuN, we found that the ShcA protein expression was increased in apoptotic neurons. In addition, mitochondrial dysfunction and excessive mitophagy were evident in photothrombotic stroke tissue. Infarct volumes were significantly reduced, and neurological deficits were diminished in the ShcA siRNA nanoparticle-treated group, compared with the negative control siRNA nanoparticle-treated group. We confirmed that the reduction of ShcA expression by nanoparticle treatment rescued the expression of genes, associated with mitochondrial dynamics and mitophagy mediation in a stroke model. This study suggests that the regulation of ShcA protein expression can be a therapeutic target for reducing brain damage with mitochondrial dysfunction caused by thrombotic infarction.
Collapse
|
32
|
Mir HA, Ali R, Mushtaq U, Khanday FA. Structure-functional implications of longevity protein p66Shc in health and disease. Ageing Res Rev 2020; 63:101139. [PMID: 32795504 DOI: 10.1016/j.arr.2020.101139] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/17/2020] [Accepted: 08/06/2020] [Indexed: 12/15/2022]
Abstract
ShcA (Src homologous- collagen homologue), family of adapter proteins, consists of three isoforms which integrate and transduce external stimuli to different signaling networks. ShcA family consists of p46Shc, p52Shc and p66Shc isoforms, characterized by having multiple protein-lipid and protein-protein interaction domains implying their functional diversity. Among the three isoforms p66Shc is structurally different containing an additional CH2 domain which attributes to its dual functionality in cell growth, mediating both cell proliferation and apoptosis. Besides, p66Shc is also involved in different biological processes including reactive oxygen species (ROS) production, cell migration, ageing, cytoskeletal reorganization and cell adhesion. Moreover, the interplay between p66Shc and ROS is implicated in the pathology of various dreadful diseases. Accordingly, here we discuss the recent structural aspects of all ShcA adaptor proteins but are highlighting the case of p66Shc as model isoform. Furthermore, this review insights the role of p66Shc in progression of chronic age-related diseases like neuro diseases, metabolic disorders (non-alcoholic fatty liver, obesity, diabetes, cardiovascular diseases, vascular endothelial dysfunction) and cancer in relation to ROS. We finally conclude that p66Shc might act as a valuable biomarker for the prognosis of these diseases and could be used as a potential therapeutic target.
Collapse
|
33
|
Zhao Y, Wang Y, Zhao E, Tan Y, Geng B, Kang C, Li X. PTRF/CAVIN1, regulated by SHC1 through the EGFR pathway, is found in urine exosomes as a potential biomarker of ccRCC. Carcinogenesis 2020; 41:274-283. [PMID: 31605605 DOI: 10.1093/carcin/bgz147] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/07/2019] [Accepted: 08/20/2019] [Indexed: 01/10/2023] Open
Abstract
Polymerase I and transcript release factor (PTRF)/Cavin1 regulates RNA polymerase I during transcription and plays a critical role in endocytosis. Abnormal expressions of PTRF were detected in multiple cancers according to increasing research. PTRF has been showed to involve in the formation and secretion of exosomes and can be detected in the exosomes, which suggests that PTRF would be a potential biomarker for diagnosis of clear cell renal cell carcinoma (ccRCC) using urine samples. Approximately 50-90% of ccRCC cases suffered abnormal epidermal growth factor receptor (EGFR), which activates a variety of signaling pathways, including the mitogen-activated protein kinase/extracellular signal-regulated kinase and Phosphoinositide 3-Kinase/Akt pathway. According to bioinformatic analysis of gene expression arrays of kidney clear cell carcinoma from The Cancer Genome Atlas, we found SHC1 was significantly overexpressed in high-grade ccRCC and correlated to poor prognosis, and also SHC1 was annotated in extracellular matrix process, which was regulated by EGFR. Further studies showed that the expression of PTRF was regulated by SHC1 through EGFR-Phosphoinositide 3-Kinase/Akt pathway. PTRF was detected in the exosomes isolated from ccRCC patients' urine and ccRCC cancer cells culture medium. It suggested that the abnormal SHC1-increased PTRF, which is detected in exosomes from urine, would be a potential marker for ccRCC diagnose and treatment.
Collapse
Affiliation(s)
- Yubo Zhao
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yunfei Wang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital; Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Enyang Zhao
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanli Tan
- Department of Pathology, Affiliated Hospital of Hebei University, Baoding, China
| | - Bo Geng
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chunsheng Kang
- Laboratory of Neuro-oncology, Tianjin Neurological Institute, Department of Neurosurgery, Tianjin Medical University General Hospital; Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Xuedong Li
- Department of Urology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
34
|
Lyu D, Tang N, Womack AW, He YJ, Lin Q. Neonatal ketamine exposure-induced hippocampal neuroapoptosis in the developing brain impairs adult spatial learning ability. Neural Regen Res 2020; 15:880-886. [PMID: 31719253 PMCID: PMC6990767 DOI: 10.4103/1673-5374.268929] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 02/25/2019] [Accepted: 06/10/2019] [Indexed: 12/19/2022] Open
Abstract
Ketamine exposure can lead to selective neuroapoptosis in the developing brain. p66ShcA, the cellular adapter protein expressed selectively in immature neurons, is a known pro-apoptotic molecule that triggers neuroapoptosis when activated. Sprague-Dawley rats at postnatal day 7 were subcutaneously injected in the neck with ketamine 20 mg/kg, six times at 2-hour intervals. At 0, 1, 3, and 6 hours after final injection, western blot assay was used to detect the expression of cleaved caspase-3, p66ShcA, and phosphorylated p66ShcA. We found that the expression of activated p66ShcA and caspase-3 increased after ketamine exposure and peaked at 3 hours. The same procedure was performed on a different group of rats. At the age of 4 weeks, spatial learning and memory abilities were tested with the Morris water maze. Latency to find the hidden platform for these rats was longer than it was for control rats, although the residence time in the target quadrant was similar. These findings indicate that ketamine exposure resulted in p66ShcA being activated in the course of an apoptotic cascade during the neonatal period. This may have contributed to the deficit in spatial learning and memory that persisted into adulthood. The experimental protocol was approved by the Institutional Animal Care and Use Committee at the University of Texas at Arlington, USA (approval No. A13.008) on January 22, 2013.
Collapse
Affiliation(s)
- Dan Lyu
- Department of Pain Management, Tianjin First Center Hospital, Tianjin, China; Department of Psychology, College of Science, University of Texas at Arlington, Arlington, TX, USA
| | - Ning Tang
- Department of Psychology, College of Science, University of Texas at Arlington, Arlington, TX, USA; Reproductive Medicine Center, the 960th Hospital of the PLA Joint Logistics Support Force, Jinan, Shandong Province, China
| | - Andrew W Womack
- Department of Psychology, College of Science, University of Texas at Arlington, Arlington, TX, USA
| | - Yong-Jin He
- Department of Pain Management, Tianjin First Center Hospital, Tianjin, China
| | - Qing Lin
- Department of Psychology, College of Science, University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
35
|
P66Shc: A Pleiotropic Regulator of B Cell Trafficking and a Gatekeeper in Chronic Lymphocytic Leukemia. Cancers (Basel) 2020; 12:cancers12041006. [PMID: 32325830 PMCID: PMC7226591 DOI: 10.3390/cancers12041006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/11/2020] [Accepted: 04/16/2020] [Indexed: 12/20/2022] Open
Abstract
Neoplastic B cells from chronic lymphocytic leukemia patients (CLL) have a profound deficiency in the expression of p66Shc, an adaptor protein with pro-apoptotic and pro-oxidant activities. This defect results in leukemic B cell resistance to apoptosis and additionally impinges on the balance between chemokine receptors that control B cell homing to secondary lymphoid organs and the sphingosine phosphate receptor S1PR1 that controls their egress therefrom, thereby favoring leukemic B cell accumulation in the pro-survival lymphoid niche. Ablation of the gene encoding p66Shc in the Eµ-TCL1 mouse model of human CLL enhances leukemogenesis and promotes leukemic cell invasiveness in both nodal and extranodal organs, providing in vivo evidence of the pathogenic role of the p66Shc defect in CLL pathogenesis. Here we present an overview of the functions of p66Shc in B lymphocytes, with a specific focus on the multiple mechanisms exploited by p66Shc to control B cell trafficking and the abnormalities in this process caused by p66Shc deficiency in CLL.
Collapse
|
36
|
Onnis A, Cassioli C, Finetti F, Baldari CT. Regulation of Selective B Cell Autophagy by the Pro-oxidant Adaptor p66SHC. Front Cell Dev Biol 2020; 8:193. [PMID: 32274384 PMCID: PMC7113388 DOI: 10.3389/fcell.2020.00193] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/06/2020] [Indexed: 01/28/2023] Open
Abstract
p66SHC is a pro-oxidant member of the SHC family of protein adaptors that acts as a negative regulator of cell survival. In lymphocytes p66SHC exploits both its adaptor and its reactive oxygen species (ROS)-elevating function to antagonize mitogenic and survival signaling and promote apoptosis. As a result, p66SHC deficiency leads to the abnormal expansion of peripheral T and B cells and lupus-like autoimmunity. Additionally, a defect in p66SHC expression is a hallmark of B cell chronic lymphocytic leukemia, where it contributes to the accumulation of long-lived neoplastic cells. We have recently provided evidence that p66SHC exerts a further layer of control on B cell homeostasis by acting as a new mitochondrial LC3-II receptor to promote the autophagic demise of dysfunctional mitochondria. Here we discuss this finding in the context of the autophagic control of B cell homeostasis, development, and differentiation in health and disease.
Collapse
Affiliation(s)
- Anna Onnis
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Chiara Cassioli
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | | |
Collapse
|
37
|
Lewis K, Kiepas A, Hudson J, Senecal J, Ha JR, Voorand E, Annis MG, Sabourin V, Ahn R, La Selva R, Tabariès S, Hsu BE, Siegel MJ, Dankner M, Canedo EC, Lajoie M, Watson IR, Brown CM, Siegel PM, Ursini-Siegel J. p66ShcA functions as a contextual promoter of breast cancer metastasis. Breast Cancer Res 2020; 22:7. [PMID: 31941526 PMCID: PMC6964019 DOI: 10.1186/s13058-020-1245-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 01/05/2020] [Indexed: 01/25/2023] Open
Abstract
Background The p66ShcA redox protein is the longest isoform of the Shc1 gene and is variably expressed in breast cancers. In response to a variety of stress stimuli, p66ShcA becomes phosphorylated on serine 36, which allows it to translocate from the cytoplasm to the mitochondria where it stimulates the formation of reactive oxygen species (ROS). Conflicting studies suggest both pro- and anti-tumorigenic functions for p66ShcA, which prompted us to examine the contribution of tumor cell-intrinsic functions of p66ShcA during breast cancer metastasis. Methods We tested whether p66ShcA impacts the lung-metastatic ability of breast cancer cells. Breast cancer cells characteristic of the ErbB2+/luminal (NIC) or basal (4T1) subtypes were engineered to overexpress p66ShcA. In addition, lung-metastatic 4T1 variants (4T1-537) were engineered to lack endogenous p66ShcA via Crispr/Cas9 genomic editing. p66ShcA null cells were then reconstituted with wild-type p66ShcA or a mutant (S36A) that cannot translocate to the mitochondria, thereby lacking the ability to stimulate mitochondrial-dependent ROS production. These cells were tested for their ability to form spontaneous metastases from the primary site or seed and colonize the lung in experimental (tail vein) metastasis assays. These cells were further characterized with respect to their migration rates, focal adhesion dynamics, and resistance to anoikis in vitro. Finally, their ability to survive in circulation and seed the lungs of mice was assessed in vivo. Results We show that p66ShcA increases the lung-metastatic potential of breast cancer cells by augmenting their ability to navigate each stage of the metastatic cascade. A non-phosphorylatable p66ShcA-S36A mutant, which cannot translocate to the mitochondria, still potentiated breast cancer cell migration, lung colonization, and growth of secondary lung metastases. However, breast cancer cell survival in the circulation uniquely required an intact p66ShcA S36 phosphorylation site. Conclusion This study provides the first evidence that both mitochondrial and non-mitochondrial p66ShcA pools collaborate in breast cancer cells to promote their maximal metastatic fitness.
Collapse
Affiliation(s)
- Kyle Lewis
- Lady Davis Institute for Medical Research, 3755 Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada.,Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montreal, QC, H3G 1Y6, Canada
| | - Alex Kiepas
- Department of Physiology, McGill University, 3655 Promenade Sir William Osler, Montreal, QC, H3G 1Y6, Canada.,Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, West, Room 513, Montreal, QC, H3A 1A3, Canada
| | - Jesse Hudson
- Lady Davis Institute for Medical Research, 3755 Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada.,Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Julien Senecal
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, West, Room 513, Montreal, QC, H3A 1A3, Canada.,Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada.,Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC, H3G 1Y6, Canada
| | - Jacqueline R Ha
- Lady Davis Institute for Medical Research, 3755 Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada.,Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Elena Voorand
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montreal, QC, H3G 1Y6, Canada.,Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, West, Room 513, Montreal, QC, H3A 1A3, Canada
| | - Matthew G Annis
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, West, Room 513, Montreal, QC, H3A 1A3, Canada.,Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC, H3G 1Y6, Canada
| | - Valerie Sabourin
- Lady Davis Institute for Medical Research, 3755 Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada
| | - Ryuhjin Ahn
- Lady Davis Institute for Medical Research, 3755 Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada.,Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Rachel La Selva
- Lady Davis Institute for Medical Research, 3755 Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada.,Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Sébastien Tabariès
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, West, Room 513, Montreal, QC, H3A 1A3, Canada.,Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC, H3G 1Y6, Canada
| | - Brian E Hsu
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, West, Room 513, Montreal, QC, H3A 1A3, Canada.,Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Matthew J Siegel
- Lady Davis Institute for Medical Research, 3755 Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada
| | - Matthew Dankner
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, West, Room 513, Montreal, QC, H3A 1A3, Canada.,Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada.,Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC, H3G 1Y6, Canada
| | - Eduardo Cepeda Canedo
- Lady Davis Institute for Medical Research, 3755 Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada.,Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montreal, QC, H3G 1Y6, Canada
| | - Mathieu Lajoie
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, West, Room 513, Montreal, QC, H3A 1A3, Canada
| | - Ian R Watson
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montreal, QC, H3G 1Y6, Canada.,Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, West, Room 513, Montreal, QC, H3A 1A3, Canada
| | - Claire M Brown
- Department of Physiology, McGill University, 3655 Promenade Sir William Osler, Montreal, QC, H3G 1Y6, Canada
| | - Peter M Siegel
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montreal, QC, H3G 1Y6, Canada. .,Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, West, Room 513, Montreal, QC, H3A 1A3, Canada. .,Department of Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC, H3G 1Y6, Canada.
| | - Josie Ursini-Siegel
- Lady Davis Institute for Medical Research, 3755 Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1E2, Canada. .,Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montreal, QC, H3G 1Y6, Canada. .,Division of Experimental Medicine, McGill University, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada. .,Gerald Bronfman Department of Oncology, McGill University, 5100 Maisonneuve Blvd West, Montreal, QC, H4A 3T2, Canada.
| |
Collapse
|
38
|
Robeson HN, Lau HR, New LA, Lalonde J, Armstrong JN, Jones N. Localization of phosphotyrosine adaptor protein ShcD/SHC4 in the adult rat central nervous system. BMC Neurosci 2019; 20:57. [PMID: 31823725 PMCID: PMC6902498 DOI: 10.1186/s12868-019-0541-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 11/30/2019] [Indexed: 12/20/2022] Open
Abstract
Background Mammalian Shc (Src homology and collagen) proteins comprise a family of four phosphotyrosine adaptor molecules which exhibit varied spatiotemporal expression and signaling functions. ShcD is the most recently discovered homologue and it is highly expressed in the developing central nervous system (CNS) and adult brain. Presently however, its localization within specific cell types of mature neural structures has yet to be characterized. Results In the current study, we examine the expression profile of ShcD in the adult rat CNS using immunohistochemistry, and compare with those of the neuronally enriched ShcB and ShcC proteins. ShcD shows relatively widespread distribution in the adult brain and spinal cord, with prominent levels of staining throughout the olfactory bulb, as well as in sub-structures of the cerebellum and hippocampus, including the subgranular zone. Co-localization studies confirm the expression of ShcD in mature neurons and progenitor cells. ShcD immunoreactivity is primarily localized to axons and somata, consistent with the function of ShcD as a cytoplasmic adaptor. Regional differences in expression are observed among neural Shc proteins, with ShcC predominating in the hippocampus, cerebellum, and some fiber tracts. Interestingly, ShcD is uniquely expressed in the olfactory nerve layer and in glomeruli of the main olfactory bulb. Conclusions Together our findings suggest that ShcD may provide a distinct signaling contribution within the olfactory system, and that overlapping expression of ShcD with other Shc proteins may allow compensatory functions in the brain.
Collapse
Affiliation(s)
- Hannah N Robeson
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Hayley R Lau
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Laura A New
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Jasmin Lalonde
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - John N Armstrong
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada
| | - Nina Jones
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, Canada.
| |
Collapse
|
39
|
Wright KD, Miller BS, El-Meanawy S, Tsaih SW, Banerjee A, Geurts AM, Sheinin Y, Sun Y, Kalyanaraman B, Rui H, Flister MJ, Sorokin A. The p52 isoform of SHC1 is a key driver of breast cancer initiation. Breast Cancer Res 2019; 21:74. [PMID: 31202267 PMCID: PMC6570928 DOI: 10.1186/s13058-019-1155-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 05/23/2019] [Indexed: 01/23/2023] Open
Abstract
Background SHC1 proteins (also called SHCA) exist in three functionally distinct isoforms (p46SHC, p52SHC, and p66SHC) that serve as intracellular adaptors for several key signaling pathways in breast cancer. Despite the broad evidence implicating SHC1 gene products as a central mediator of breast cancer, testing the isoform-specific roles of SHC1 proteins have been inaccessible due to the lack of isoform-specific inhibitors or gene knockout models. Methods Here, we addressed this issue by generating the first isoform-specific gene knockout models for p52SHC and p66SHC, using germline gene editing in the salt-sensitive rat strain. Compared with the wild-type (WT) rats, we found that genetic ablation of the p52SHC isoform significantly attenuated mammary tumor formation, whereas the p66SHC knockout had no effect. Rats were dosed with 7,12-dimethylbenz(a)anthracene (DMBA) by oral gavage to induce mammary tumors, and progression of tumor development was followed for 15 weeks. At 15 weeks, tumors were excised and analyzed by RNA-seq to determine differences between tumors lacking p66SHC or p52SHC. Results Compared with the wild-type (WT) rats, we found that genetic ablation of the p52SHC isoform significantly attenuated mammary tumor formation, whereas the p66SHC knockout had no effect. These data, combined with p52SHC being the predominant isoform that is upregulated in human and rat tumors, provide the first evidence that p52SHC is the oncogenic isoform of Shc1 gene products in breast cancer. Compared with WT tumors, 893 differentially expressed (DE; FDR < 0.05) genes were detected in p52SHC KO tumors compared with only 18 DE genes in the p66SHC KO tumors, further highlighting that p52SHC is the relevant SHC1 isoform in breast cancer. Finally, gene network analysis revealed that p52SHC KO disrupted multiple key pathways that have been previously implicated in breast cancer initiation and progression, including ESR1 and mTORC2/RICTOR. Conclusion Collectively, these data demonstrate the p52SHC isoform is the key driver of DMBA-induced breast cancer while the expression of p66SHC and p46SHC are not enough to compensate. Electronic supplementary material The online version of this article (doi:10.1186/s13058-019-1155-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kevin D Wright
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Bradley S Miller
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Sarah El-Meanawy
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Free Radical Research Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Shirng-Wern Tsaih
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Anjishnu Banerjee
- Institute for Health and Equity, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Aron M Geurts
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Yuri Sheinin
- Department of Pathology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Yunguang Sun
- Department of Pathology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Balaraman Kalyanaraman
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Free Radical Research Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Michael J Flister
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Andrey Sorokin
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA. .,Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
40
|
Soysa NS, Alles N. The role of IL-3 in bone. J Cell Biochem 2019; 120:6851-6859. [PMID: 30320936 DOI: 10.1002/jcb.27956] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 10/02/2018] [Indexed: 01/24/2023]
Abstract
In the recent past, there has been a burgeoning interest in targeting cytokines such as IL-3 for specific disease conditions of bone such as rheumatoid arthritis and multiple myeloma. Unlike other cytokines, IL-3 is a cytokine with a multilineage potential and broad spectrum of target cells and it plays a vital role in hematopoiesis. Due to its common receptor subunit, the action of IL-3 shows functional redundancy with other cytokines such as the granulocyte-macrophage colony-stimulating factor and IL-5. IL-3 has been successfully used in ameliorating radiation-induced bone marrow aplasia and similar conditions. However, the role of IL-3 in bone cells has not been fully unraveled yet; therefore, the aim of this overview is to present the effects of IL-3 in bone with a special emphasis on osteoclasts and osteoblasts in a concise manner.
Collapse
Affiliation(s)
| | - Neil Alles
- Department of Biochemistry, Faculty of Medicine, University of Peradeniya, Sri Lanka
| |
Collapse
|
41
|
P66Shc and vascular endothelial function. Biosci Rep 2019; 39:BSR20182134. [PMID: 30918103 PMCID: PMC6488855 DOI: 10.1042/bsr20182134] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/21/2019] [Accepted: 03/26/2019] [Indexed: 12/23/2022] Open
Abstract
Dysfunctional endothelium is an early change in vasculature known to be associated with atherosclerosis. Among many regulators of vascular endothelial function, p66Shc has consistently been shown to mediate endothelial dysfunction. Over more than three decades of active research in the field of the physiological function of p66Shc, regulation of vascular endothelial functions has emerged as one of the most robust effects in a broad range of pathological conditions including hyperlipidemia, diabetes, and aging. A significant understanding has been developed with respect to the molecular signaling regulating the oxidative function of p66Shc in endothelial cells and its targets and regulators. In addition, novel regulatory modifications of p66Shc controlling its oxidative function, subcellular distribution, and stability have also been reported. This review will focus on summarizing the molecular signaling regulating the oxidative function of p66Shc and its role in vascular endothelium.
Collapse
|
42
|
Modulation of Obesity and Insulin Resistance by the Redox Enzyme and Adaptor Protein p66 Shc. Int J Mol Sci 2019; 20:ijms20040985. [PMID: 30813483 PMCID: PMC6412263 DOI: 10.3390/ijms20040985] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/15/2019] [Accepted: 02/21/2019] [Indexed: 12/27/2022] Open
Abstract
Initially reported as a longevity-related protein, the 66 kDa isoform of the mammalian Shc1 locus has been implicated in several metabolic pathways, being able to act both as an adaptor protein and as a redox enzyme capable of generating reactive oxygen species (ROS) when it localizes to the mitochondrion. Ablation of p66Shc has been shown to be protective against obesity and the insurgence of insulin resistance, but not all the studies available in the literature agree on these points. This review will focus in particular on the role of p66Shc in the modulation of glucose homeostasis, obesity, body temperature, and respiration/energy expenditure. In view of the obesity and diabetes epidemic, p66Shc may represent a promising therapeutic target with enormous implications for human health.
Collapse
|
43
|
Zeglinski MR, Moghadam AR, Ande SR, Sheikholeslami K, Mokarram P, Sepehri Z, Rokni H, Mohtaram NK, Poorebrahim M, Masoom A, Toback M, Sareen N, Saravanan S, Jassal DS, Hashemi M, Marzban H, Schaafsma D, Singal P, Wigle JT, Czubryt MP, Akbari M, Dixon IM, Ghavami S, Gordon JW, Dhingra S. Myocardial Cell Signaling During the Transition to Heart Failure. Compr Physiol 2018; 9:75-125. [DOI: 10.1002/cphy.c170053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
44
|
Dinsmore CJ, Soriano P. MAPK and PI3K signaling: At the crossroads of neural crest development. Dev Biol 2018; 444 Suppl 1:S79-S97. [PMID: 29453943 PMCID: PMC6092260 DOI: 10.1016/j.ydbio.2018.02.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/06/2018] [Accepted: 02/06/2018] [Indexed: 02/08/2023]
Abstract
Receptor tyrosine kinase-mediated growth factor signaling is essential for proper formation and development of the neural crest. The many ligands and receptors implicated in these processes signal through relatively few downstream pathways, frequently converging on the MAPK and PI3K pathways. Despite decades of study, there is still considerable uncertainty about where and when these signaling pathways are required and how they elicit particular responses. This review summarizes our current understanding of growth factor-induced MAPK and PI3K signaling in the neural crest.
Collapse
Affiliation(s)
- Colin J Dinsmore
- Department of Cell, Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, NY 10029, USA
| | - Philippe Soriano
- Department of Cell, Developmental and Regenerative Biology, Tisch Cancer Institute, Icahn School of Medicine at Mt. Sinai, New York, NY 10029, USA.
| |
Collapse
|
45
|
Tomilov A, Allen S, Hui CK, Bettaieb A, Cortopassi G. Idebenone is a cytoprotective insulin sensitizer whose mechanism is Shc inhibition. Pharmacol Res 2018; 137:89-103. [PMID: 30290222 DOI: 10.1016/j.phrs.2018.09.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 01/04/2023]
Abstract
When insulin binds insulin receptor, IRS1 signaling is stimulated to trigger the maximal insulin response. p52Shc protein competes directly with IRS1, thus damping and diverting maximal insulin response. Genetic reduction of p52Shc minimizes competition with IRS1, and improves insulin signaling and glucose control in mice, and improves pathophysiological consequences of hyperglycemia. Given the multiple benefits of Shc reduction in vivo, we investigated whether any of 1680 drugs used in humans may function as Shc inhibitors, and thus potentially serve as novel anti-diabetics. Of the 1680, 30 insulin sensitizers were identified by screening in vitro, and of these 30 we demonstrated that 7 bound Shc protein. Of the 7 drugs, idebenone dose-dependently bound Shc protein in the 50-100 nM range, and induced insulin sensitivity and cytoprotection in this same 100 nM range that clinically dosed idebenone reaches in human plasma. By contrast we observe mitochondrial effects of idebenone in the 5,000 nM range that are not reached in human dosing. Multiple assays of target engagement demonstrate that idebenone physically interacts with Shc protein. Idebenone sensitizes mice to insulin in two different mouse models of prediabetes. Genetic depletion of idebenone's target eliminates idebenone's ability to insulin-sensitize in vivo. Thus, idebenone is the first-in-class member of a novel category of insulin-sensitizing and cytoprotective agents, the Shc inhibitors. Idebenone is an approved drug and could be considered for other indications such as type 2 diabetes and fatty liver disease, in which insulin resistance occurs.
Collapse
Affiliation(s)
- Alexey Tomilov
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - Sonia Allen
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - Chun Kiu Hui
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| | - Ahmed Bettaieb
- Department of Nutrition, The University of Tennessee, 1215 W. Cumberland Ave, Knoxville, TN, 37996-1920, USA.
| | - Gino Cortopassi
- Department of Molecular Biosciences, 1089 Veterinary Medicine Dr., VM3B, UC Davis, CA, 95616, USA.
| |
Collapse
|
46
|
Kakizawa S. [Functional roles of phosphotyrosine adaptor Shc in the brain]. Nihon Yakurigaku Zasshi 2018; 152:84-89. [PMID: 30101865 DOI: 10.1254/fpj.152.84] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Adaptor molecules (adaptor proteins) have indispensable roles in cellular signaling, essential for cellular proliferation, development and metabolism. Shc (Src homology and collagen homology)-family molecule is a group of adaptor molecules, and indicated to be involved in intracellular phosphotyrosine signaling. Shc family has 4 subtypes, ShcA-ShcD, and there are long and short isoforms in ShcA and ShcC whereas ShcB and ShcD have short isoform only. There are three domains conserved in all Shc-family isoforms: phosphotyrosine-binding (PTB) domain, collagen-homology 1 (CH1) domain and Src-homology 2 (SH2) domain, from the N-terminal to C-terminal. PTB and SH2 domains recognize and bind to phosphotyrosine in other molecules, and CH1 domain is recognized and bind to SH2 domain in Grb2, an adaptor molecule, when the tyrosine residues in the domain are phosphorylated. Expression of ShcA is observed in all tissues except for brain in adult animals, although ShcA mRNA is detected in brain during embryonic days. On the other hand, in adult brain, expressions of ShcB, ShcC, and ShcD are observed. Analysis of single knockout mice (ShcA (neuron specific), ShcB, ShcC) and double knockout mice for ShcB and C indicated essential roles of Shc-family molecules in proliferation and survival of cells in various brain regions as well as synaptic plasticity and higher brain functions such as learning and memory. Studies on multiple-knockout mice of Shc-family molecules may further clarify possible involvements of Shc family in physiological and pathophysiological functions in brain.
Collapse
Affiliation(s)
- Sho Kakizawa
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University
| |
Collapse
|
47
|
Abstract
Insulin-like growth factors (IGFs) bind specifically to the IGF1 receptor on the cell surface of targeted tissues. Ligand binding to the α subunit of the receptor leads to a conformational change in the β subunit, resulting in the activation of receptor tyrosine kinase activity. Activated receptor phosphorylates several substrates, including insulin receptor substrates (IRSs) and Src homology collagen (SHC). Phosphotyrosine residues in these substrates are recognized by certain Src homology 2 (SH2) domain-containing signaling molecules. These include, for example, an 85 kDa regulatory subunit (p85) of phosphatidylinositol 3-kinase (PI 3-kinase), growth factor receptor-bound 2 (GRB2) and SH2-containing protein tyrosine phosphatase 2 (SHP2/Syp). These bindings lead to the activation of downstream signaling pathways, PI 3-kinase pathway and Ras-mitogen-activated protein kinase (MAP kinase) pathway. Activation of these signaling pathways is known to be required for the induction of various bioactivities of IGFs, including cell proliferation, cell differentiation and cell survival. In this review, the well-established IGF1 receptor signaling pathways required for the induction of various bioactivities of IGFs are introduced. In addition, we will discuss how IGF signals are modulated by the other extracellular stimuli or by themselves based on our studies.
Collapse
Affiliation(s)
- Fumihiko Hakuno
- Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Shin-Ichiro Takahashi
- Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
48
|
Im YK, Najyb O, Gravel SP, McGuirk S, Ahn R, Avizonis DZ, Chénard V, Sabourin V, Hudson J, Pawson T, Topisirovic I, Pollak M, St-Pierre J, Ursini-Siegel J. Interplay between ShcA Signaling and PGC-1α Triggers Targetable Metabolic Vulnerabilities in Breast Cancer. Cancer Res 2018; 78:4826-4838. [DOI: 10.1158/0008-5472.can-17-3696] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 05/07/2018] [Accepted: 06/18/2018] [Indexed: 11/16/2022]
|
49
|
Heritable, Allele-Specific Chromosomal Looping between Tandem Promoters Specifies Promoter Usage of SHC1. Mol Cell Biol 2018; 38:MCB.00658-17. [PMID: 29440311 DOI: 10.1128/mcb.00658-17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/08/2018] [Indexed: 11/20/2022] Open
Abstract
One-half of the genes in the human genome contain alternative promoters, some of which generate products with opposing functions. Aberrant silencing or activation of such alternative promoters is associated with multiple diseases, including cancer, but little is known regarding the molecular mechanisms that control alternative promoter choice. The SHC1 gene encodes p46Shc/p52Shc and p66Shc, proteins oppositely regulating anchorage-independent growth that are produced by transcription initiated from the upstream and downstream tandem promoters of SHC1, respectively. Here we demonstrate that activation of these promoters is mutually exclusive on separate alleles in single primary endothelial cells in a heritable fashion, ensuring expression of both transcripts by the cell. Peripheral blood lymphocytes that do not transcribe p66Shc transcribed p52Shc biallelically. This distinct monoallelic transcription pattern is established by allele-specific chromosomal looping between tandem promoters, which silences the upstream promoter. Our results reveal a new mechanism to control alternative promoter usage through higher-order chromatin structure.
Collapse
|
50
|
Kuwabara T, Matsui Y, Ishikawa F, Kondo M. Regulation of T-Cell Signaling by Post-Translational Modifications in Autoimmune Disease. Int J Mol Sci 2018. [PMID: 29534522 PMCID: PMC5877680 DOI: 10.3390/ijms19030819] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The adaptive immune system involves antigen-specific host defense mechanisms mediated by T and B cells. In particular, CD4+ T cells play a central role in the elimination of pathogens. Immunological tolerance in the thymus regulates T lymphocytes to avoid self-components, including induction of cell death in immature T cells expressing the self-reactive T-cell receptor repertoire. In the periphery, mature T cells are also regulated by tolerance, e.g., via induction of anergy or regulatory T cells. Thus, T cells strictly control intrinsic signal transduction to prevent excessive responses or self-reactions. If the inhibitory effects of T cells on these mechanisms are disrupted, T cells may incorrectly attack self-components, which can lead to autoimmune disease. The functions of T cells are supported by post-translational modifications, particularly phosphorylation, of signaling molecules, the proper regulation of which is controlled by endogenous mechanisms within the T cells themselves. In recent years, molecular targeted agents against kinases have been developed for treatment of autoimmune diseases. In this review, we discuss T-cell signal transduction in autoimmune disease and provide an overview of acetylation-mediated regulation of T-cell signaling pathways.
Collapse
Affiliation(s)
- Taku Kuwabara
- Department of Molecular Immunology, Toho University School of Medicine, 5-21-16 Omori-Nishi, Ota-ku, Tokyo 143-8540, Japan.
| | - Yukihide Matsui
- Department of Molecular Immunology, Toho University School of Medicine, 5-21-16 Omori-Nishi, Ota-ku, Tokyo 143-8540, Japan.
| | - Fumio Ishikawa
- Department of Molecular Immunology, Toho University School of Medicine, 5-21-16 Omori-Nishi, Ota-ku, Tokyo 143-8540, Japan.
| | - Motonari Kondo
- Department of Molecular Immunology, Toho University School of Medicine, 5-21-16 Omori-Nishi, Ota-ku, Tokyo 143-8540, Japan.
| |
Collapse
|