1
|
Zhang Y, Zhang H, Liu L. Integration of single-cell and bulk RNA sequencing identifies and validates T cell-related prognostic model in hepatocellular carcinoma. PLoS One 2025; 20:e0322706. [PMID: 40315269 PMCID: PMC12047759 DOI: 10.1371/journal.pone.0322706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/23/2025] [Indexed: 05/04/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a lethal malignancy, and predicting patient prognosis remains a significant challenge in clinical treatment. T cells play a crucial role in the tumor microenvironment, influencing tumorigenesis and progression. In this study, we constructed a T cell-related prognostic model for HCC. Using single-cell RNA sequencing (scRNA-seq) data from the Gene Expression Omnibus (GEO) database, we identified 6,281 T cells from 10 HCC patients and subsequently identified 855 T cell-related genes. Comprehensive analyses were conducted on T cells and their associated genes, including enrichment analysis, cell-cell communication, trajectory analysis, and transcription factor analysis. By integrating scRNA-seq and bulk RNA-seq data with prognostic information from The Cancer Genome Atlas (TCGA), we identified T cell-related prognostic genes and constructed a model using LASSO regression. The model, incorporating PTTG1, LMNB1, SLC38A1, and BATF, was externally validated using the International Cancer Genome Consortium (ICGC) database. It effectively stratified patients into high- and low-risk groups based on risk scores, revealing significant differences in immune cell infiltration between these groups. Differential expression levels of PTTG1 and BATF between HCC and adjacent non-tumor tissues were further validated by immunohistochemistry (IHC) in 25 patient tissue samples. Moreover, a Cox regression analysis was performed to integrate risk scores with clinical features, resulting in a nomogram capable of predicting patient survival probabilities. This study introduces a novel prognostic risk model for HCC patients, aimed at stratifying patients by risk, enhancing personalized treatment strategies, and offering new insights into the role of T cell-related genes in HCC progression.
Collapse
Affiliation(s)
- Yuzhi Zhang
- Department of Gastroenterology and Hepatology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Haiyan Zhang
- Department of Gastroenterology and Hepatology, The First Hospital of Shanxi Medical University, Taiyuan, China
- Experimental Center of Science and Research, The First Hospital of Shanxi Medical University, Taiyuan, China
- Key Laboratory of Prevention and Treatment of Liver Injury and Digestive System Neoplasms, Provincial Committee of the Medical and Health, Taiyuan, China
| | - Lixin Liu
- Department of Gastroenterology and Hepatology, The First Hospital of Shanxi Medical University, Taiyuan, China
- Experimental Center of Science and Research, The First Hospital of Shanxi Medical University, Taiyuan, China
- Key Laboratory of Prevention and Treatment of Liver Injury and Digestive System Neoplasms, Provincial Committee of the Medical and Health, Taiyuan, China
| |
Collapse
|
2
|
Zhong B, Du J, Liu F, Sun S. The Role of Yes-Associated Protein in Inflammatory Diseases and Cancer. MedComm (Beijing) 2025; 6:e70128. [PMID: 40066231 PMCID: PMC11892025 DOI: 10.1002/mco2.70128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 02/02/2025] [Accepted: 02/11/2025] [Indexed: 03/17/2025] Open
Abstract
Yes-associated protein (YAP) plays a central role in the Hippo pathway, primarily governing cell proliferation, differentiation, and apoptosis. Its significance extends to tumorigenesis and inflammatory conditions, impacting disease initiation and progression. Given the increasing relevance of YAP in inflammatory disorders and cancer, this study aims to elucidate its pathological regulatory functions in these contexts. Specifically, we aim to investigate the involvement and molecular mechanisms of YAP in various inflammatory diseases and cancers. We particularly focus on how YAP activation, whether through Hippo-dependent or independent pathways, triggers the release of inflammation and inflammatory mediators in respiratory, cardiovascular, and digestive inflammatory conditions. In cancer, YAP not only promotes tumor cell proliferation and differentiation but also modulates the tumor immune microenvironment, thereby fostering tumor metastasis and progression. Additionally, we provide an overview of current YAP-targeted therapies. By emphasizing YAP's role in inflammatory diseases and cancer, this study aims to enhance our understanding of the protein's pivotal involvement in disease processes, elucidate the intricate pathological mechanisms of related diseases, and contribute to future drug development strategies targeting YAP.
Collapse
Affiliation(s)
- Bing Zhong
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Jintao Du
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Feng Liu
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Silu Sun
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesChinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and ManagementWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| |
Collapse
|
3
|
Li G, Wen Z, Xiong S. Microenvironmental β-TrCP negates amino acid transport to trigger CD8 + T cell exhaustion in human non-small cell lung cancer. Cell Rep 2025; 44:115128. [PMID: 39754718 DOI: 10.1016/j.celrep.2024.115128] [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: 04/11/2024] [Revised: 09/05/2024] [Accepted: 12/10/2024] [Indexed: 01/06/2025] Open
Abstract
CD8+ T cell exhaustion (Tex) has been widely acknowledged in human cancer, while the underlying mechanisms remain unclear. Here, we demonstrate that reduced amino acid (aa) metabolism and mTOR inactivation are accountable for Tex in human non-small cell lung cancer (NSCLC). NSCLC cells impede the T cell-intrinsic transcription of SLC7A5 and SLC38A1, disrupting aa transport and consequently leading to mTOR inactivation. Further, the ubiquitination of YAP1 protein is the basis for NSCLC-mediated transcriptional inhibition of aa transporters. Mechanistically, NSCLC cells transfer β-TrCP-containing exosomes into T cells, inducing YAP1 ubiquitination and Tex. Consequently, inhibiting cancer-associated β-TrCP effectively restores the anti-tumor immune response of CD8+ T cells and curtails tumor growth in NSCLC patient-derived organoids. Together, our findings highlight a β-TrCP-dependent mechanism in steering intrinsic metabolic adaptation and CD8+ Tex, emphasizing microenvironmental β-TrCP as an immune checkpoint for therapeutic exploration against human NSCLC.
Collapse
Affiliation(s)
- Ge Li
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Zhenke Wen
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China.
| | - Sidong Xiong
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China.
| |
Collapse
|
4
|
He L, Cho S, Blenis J. mTORC1, the maestro of cell metabolism and growth. Genes Dev 2025; 39:109-131. [PMID: 39572234 PMCID: PMC11789495 DOI: 10.1101/gad.352084.124] [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] [Indexed: 01/11/2025]
Abstract
The mechanistic target of rapamycin (mTOR) pathway senses and integrates various environmental and intracellular cues to regulate cell growth and proliferation. As a key conductor of the balance between anabolic and catabolic processes, mTOR complex 1 (mTORC1) orchestrates the symphonic regulation of glycolysis, nucleic acid and lipid metabolism, protein translation and degradation, and gene expression. Dysregulation of the mTOR pathway is linked to numerous human diseases, including cancer, neurodegenerative disorders, obesity, diabetes, and aging. This review provides an in-depth understanding of how nutrients and growth signals are coordinated to influence mTOR signaling and the extensive metabolic rewiring under its command. Additionally, we discuss the use of mTORC1 inhibitors in various aging-associated metabolic diseases and the current and future potential for targeting mTOR in clinical settings. By deciphering the complex landscape of mTORC1 signaling, this review aims to inform novel therapeutic strategies and provide a road map for future research endeavors in this dynamic and rapidly evolving field.
Collapse
Affiliation(s)
- Long He
- Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10021, USA;
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, USA
| | - Sungyun Cho
- Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10021, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, USA
| | - John Blenis
- Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10021, USA;
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, USA
| |
Collapse
|
5
|
Hong XL, Huang CK, Qian H, Ding CH, Liu F, Hong HY, Liu SQ, Wu SH, Zhang X, Xie WF. Positive feedback between arginine methylation of YAP and methionine transporter SLC43A2 drives anticancer drug resistance. Nat Commun 2025; 16:87. [PMID: 39747898 PMCID: PMC11697449 DOI: 10.1038/s41467-024-55769-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
Yes-associated protein (YAP) activation confers resistance to chemotherapy and targeted therapy. Methionine participates in cellular processes by converting to methyl donor for the methylation of DNA, RNA and protein. However, it remains unclear whether methionine affects drug resistance by influencing YAP activity. In this study, we report that methionine deprivation remarkably suppresses the transcriptional activity of YAP-TEAD in cancer cells. Methionine promotes PRMT1-catalyzed asymmetric dimethylation at R124 of YAP (YAP R124me2a). Mimicking of YAP methylation abolishes the reduction effect of methionine-restricted diet on YAP-induced drug resistance. YAP activates the transcription of SLC43A2, the methionine transporter, to increase methionine uptake in cancer cells. Knockdown of SLC43A2 decreases the level of YAP R124me2a. BCH, the inhibitor of SLC43A2, sensitizes tumors to anticancer drugs. Thus, our results unravel the positive feedback between YAP R124 methylation and SLC43A2 that contributes to anticancer drug resistance. Disrupting this positive feedback could be a potential strategy for cancer therapy.
Collapse
Affiliation(s)
- Xia-Lu Hong
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chen-Kai Huang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hui Qian
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chen-Hong Ding
- Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fang Liu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Huan-Yu Hong
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Shu-Qing Liu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Si-Han Wu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xin Zhang
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Wei-Fen Xie
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
6
|
Hossen MS, Islam MSU, Yasin M, Ibrahim M, Das A. A Review on the Role of Human Solute Carriers Transporters in Cancer. Health Sci Rep 2025; 8:e70343. [PMID: 39807482 PMCID: PMC11725534 DOI: 10.1002/hsr2.70343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/03/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Background and Aim The high rate of tumor growth results in an increased need for amino acids. As solute carriers (SLC) transporters are capable of transporting different amino acids, cancer may develop as a result of these transporters' over-expression due to their complex formation with other biological molecules. Therefore, this review investigated the role of SLC transporters in the progression of cancer. Methods We retrieved data from Google Scholar, Web of Science, PubMed, Cochrane Library, and EMBASE regarding the influence of human SLCs on the development of cancer. Articles published in English before August 2024 were included in the study. Results The overexpression of SLCs is strongly related to tumor cell proliferation and angiogenesis in a number of cancer types including thyroid, pancreatic, lung, hepatocellular, and colon cancers. They are crucial for the stimulation of several biological signaling pathways, particularly mTOR kinase activity, which starts a signaling cascade, protein synthesis, cell growth, and proliferation, and inhibits apoptosis of cancerous cells. Furthermore, they contribute to the activation of PI3K/AKT signaling, which has an impact on the growth, invasion, and death of cancer cells. Thus, SLC transporters become a potential therapeutic target that plays a crucial role in drug resistance, tumor microenvironment regulation, and modulation of immune response. Conclusion The review recognized the crucial role of SLC transporters in different types of cancer progression. Therefore, to confirm our findings, a case-control study is required to investigate the role of amino acid transporters in cancer development.
Collapse
Affiliation(s)
- Md. Shafiul Hossen
- Department of PharmacyState University of BangladeshDhakaBangladesh
- Department of PharmacyNoakhali Science and Technology UniversitySonapurBangladesh
| | | | - Mohammad Yasin
- Department of PharmacySouthern University BangladeshChittagongBangladesh
| | - Mohammed Ibrahim
- Department of PharmacyState University of BangladeshDhakaBangladesh
| | - Abhijit Das
- Department of PharmacyNoakhali Science and Technology UniversitySonapurBangladesh
| |
Collapse
|
7
|
Dai C, Zhang Y, Gong Y, Bradley A, Tang Z, Sellick K, Shrestha S, Spears E, Covington BA, Stanley J, Jenkins R, Richardson TM, Brantley RA, Coate K, Saunders DC, Wright JJ, Brissova M, Dean ED, Powers AC, Chen W. Hyperaminoacidemia from interrupted glucagon signaling increases pancreatic acinar cell proliferation and size via mTORC1 and YAP pathways. iScience 2024; 27:111447. [PMID: 39720531 PMCID: PMC11667045 DOI: 10.1016/j.isci.2024.111447] [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: 02/23/2024] [Revised: 06/28/2024] [Accepted: 11/19/2024] [Indexed: 12/26/2024] Open
Abstract
Increased blood amino acid levels (hyperaminoacidemia) stimulate pancreas expansion by unclear mechanisms. Here, by genetic and pharmacological disruption of glucagon receptor (GCGR) in mice and zebrafish, we found that the ensuing hyperaminoacidemia promotes pancreatic acinar cell proliferation and cell hypertrophy, which can be mitigated by a low protein diet in mice. In addition to mammalian target of rapamycin complex 1 (mTORC1) signaling, acinar cell proliferation required slc38a5, the most highly expressed amino acid transporter gene in both species. Transcriptomics data revealed the activation signature of yes-associated protein (YAP) in acinar cells of mice with hyperaminoacidemia, consistent with the observed increase in YAP-expressing acinar cells. Yap1 activation also occurred in acinar cells in gcgr-/- zebrafish, which was reversed by rapamycin. Knocking down yap1 in gcgr-/- zebrafish decreased mTORC1 activity and acinar cell proliferation and hypertrophy. Thus, the study discovered a previously unrecognized role of the YAP/Taz pathway in hyperaminoacidemia-induced acinar cell hypertrophy and hyperplasia.
Collapse
Affiliation(s)
- Chunhua Dai
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yue Zhang
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Yulong Gong
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Amber Bradley
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Zihan Tang
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Katelyn Sellick
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shristi Shrestha
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Erick Spears
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Brittney A. Covington
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jade Stanley
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Regina Jenkins
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tiffany M. Richardson
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Rebekah A. Brantley
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Katie Coate
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Diane C. Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jordan J. Wright
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Marcela Brissova
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - E. Danielle Dean
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Alvin C. Powers
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
- VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Wenbiao Chen
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
8
|
Li S, Hao L, Li N, Hu X, Yan H, Dai E, Shi X. Targeting the Hippo/YAP1 signaling pathway in hepatocellular carcinoma: From mechanisms to therapeutic drugs (Review). Int J Oncol 2024; 65:88. [PMID: 39092548 DOI: 10.3892/ijo.2024.5676] [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: 06/04/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024] Open
Abstract
The Hippo signaling pathway plays a pivotal role in regulating cell growth and organ size. Its regulatory effects on hepatocellular carcinoma (HCC) encompass diverse aspects, including cell proliferation, invasion and metastasis, tumor drug resistance, metabolic reprogramming, immunomodulatory effects and autophagy. Yes‑associated protein 1 (YAP1), a potent transcriptional coactivator and a major downstream target tightly controlled by the Hippo pathway, is influenced by various molecules and pathways. The expression of YAP1 in different cell types within the liver tumor microenvironment exerts varying effects on tumor outcomes, warranting careful consideration. Therefore, research on YAP1‑targeted therapies merits attention. This review discusses the composition and regulation mechanism of the Hippo/YAP1 signaling pathway and its relationship with HCC, offering insights for future research and cancer prevention strategies.
Collapse
Affiliation(s)
- Shenghao Li
- Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Liyuan Hao
- Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Na Li
- Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Xiaoyu Hu
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Huimin Yan
- Clinical Research Center, Shijiazhuang Fifth Hospital, Shijiazhuang, Hebei 050024, P.R. China
| | - Erhei Dai
- Clinical Research Center, Shijiazhuang Fifth Hospital, Shijiazhuang, Hebei 050024, P.R. China
| | - Xinli Shi
- Center of Experimental Management, Shanxi University of Chinese Medicine, Jinzhong, Shanxi 030619, P.R. China
| |
Collapse
|
9
|
Hushmandi K, Einollahi B, Saadat SH, Lee EHC, Farani MR, Okina E, Huh YS, Nabavi N, Salimimoghadam S, Kumar AP. Amino acid transporters within the solute carrier superfamily: Underappreciated proteins and novel opportunities for cancer therapy. Mol Metab 2024; 84:101952. [PMID: 38705513 PMCID: PMC11112377 DOI: 10.1016/j.molmet.2024.101952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/24/2024] [Accepted: 04/27/2024] [Indexed: 05/07/2024] Open
Abstract
BACKGROUND Solute carrier (SLC) transporters, a diverse family of membrane proteins, are instrumental in orchestrating the intake and efflux of nutrients including amino acids, vitamins, ions, nutrients, etc, across cell membranes. This dynamic process is critical for sustaining the metabolic demands of cancer cells, promoting their survival, proliferation, and adaptation to the tumor microenvironment (TME). Amino acids are fundamental building blocks of cells and play essential roles in protein synthesis, nutrient sensing, and oncogenic signaling pathways. As key transporters of amino acids, SLCs have emerged as crucial players in maintaining cellular amino acid homeostasis, and their dysregulation is implicated in various cancer types. Thus, understanding the intricate connections between amino acids, SLCs, and cancer is pivotal for unraveling novel therapeutic targets and strategies. SCOPE OF REVIEW In this review, we delve into the significant impact of amino acid carriers of the SLCs family on the growth and progression of cancer and explore the current state of knowledge in this field, shedding light on the molecular mechanisms that underlie these relationships and highlighting potential avenues for future research and clinical interventions. MAJOR CONCLUSIONS Amino acids transportation by SLCs plays a critical role in tumor progression. However, some studies revealed the tumor suppressor function of SLCs. Although several studies evaluated the function of SLC7A11 and SLC1A5, the role of some SLC proteins in cancer is not studied well. To exert their functions, SLCs mediate metabolic rewiring, regulate the maintenance of redox balance, affect main oncogenic pathways, regulate amino acids bioavailability within the TME, and alter the sensitivity of cancer cells to therapeutics. However, different therapeutic methods that prevent the function of SLCs were able to inhibit tumor progression. This comprehensive review provides insights into a rapidly evolving area of cancer biology by focusing on amino acids and their transporters within the SLC superfamily.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Marzieh Ramezani Farani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
10
|
Yang Y, Jia S, Zhu N, Xiao X, Ma Y, Tu K, Guo Y, Xu Q. OTUD5 promotes the growth of hepatocellular carcinoma by deubiquitinating and stabilizing SLC38A1. Biol Direct 2024; 19:31. [PMID: 38658981 PMCID: PMC11041014 DOI: 10.1186/s13062-024-00475-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Deubiquitinating enzymes (DUBs) cleave ubiquitin on substrate molecules to maintain protein stability. DUBs reportedly participate in the tumorigenesis and tumour progression of hepatocellular carcinoma (HCC). OTU deubiquitinase 5 (OTUD5), a DUB family member, has been recognized as a critical regulator in bladder cancer, breast cancer and HCC. However, the expression and biological function of OTUD5 in HCC are still controversial. RESULTS We determined that the expression of OTUD5 was significantly upregulated in HCC tissues. High levels of OTUD5 were also detected in most HCC cell lines. TCGA data analysis demonstrated that high OTUD5 expression indicated poorer overall survival in HCC patients. OTUD5 silencing prominently suppressed HCC cell proliferation, while its overexpression markedly enhanced the proliferation of HCC cells. Mass spectrometry analysis revealed solute carrier family 38 member 1 (SLC38A1) as a candidate downstream target protein of OTUD5. Coimmunoprecipitation analysis confirmed the interaction between OTUD5 and SLC38A1. OTUD5 knockdown reduced and OTUD5 overexpression increased SLC38A1 protein levels in HCC cells. However, OTUD5 alteration had no effect on SLC38A1 mRNA expression. OTUD5 maintained SLC38A1 stability by preventing its ubiquitin-mediated proteasomal degradation. SLC38A1 silencing prominently attenuated the OTUD5-induced increase in HCC cell proliferation. Finally, OTUD5 knockdown markedly suppressed the growth of HCC cells in vivo. CONCLUSIONS OTUD5 is an oncogene in HCC. OTUD5 contributes to HCC cell proliferation by deubiquitinating and stabilizing SLC38A1. These results may provide a theoretical basis for the development of new anti-HCC drugs.
Collapse
Affiliation(s)
- Yingnan Yang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, 310053, Hangzhou, China
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, China
| | - Siying Jia
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, China
| | - Ning Zhu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, China
| | - Xuelian Xiao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, China
| | - Ying Ma
- Department of Cardiovascular Medicine, The Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, 710018, Xi'an, China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, China
| | - Yong Guo
- Department of Gastrointestinal Surgery, Shangluo Central Hospital, 726000, Shangluo, China.
| | - Qiuran Xu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014, Hangzhou, China.
| |
Collapse
|
11
|
Roshanmehr F, Abdoli S, Bazi Z, Jari M, Shahbazi M. Enhancing the productivity and proliferation of CHO-K1 cells by oncoprotein YAP (Yes-associated protein). Appl Microbiol Biotechnol 2024; 108:285. [PMID: 38573360 PMCID: PMC10994876 DOI: 10.1007/s00253-024-13122-5] [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: 12/19/2023] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/05/2024]
Abstract
CHO cells are extensively employed in biological drug industry to manufacture therapeutic proteins. Nevertheless, production of biopharmaceuticals faces obstacles such as limited growth and inadequate productivity. Employing host cell engineering techniques for CHO cells serves as a valuable approach to address the constraints encountered in biologics manufacturing. Despite advancements, most techniques focus on specific genes to address individual cellular challenges. The significance of YAP, transcriptional co-activator, cannot be overstated due to its involvement in regulating organ size and tumor formation. YAP's influence extends to various cellular processes and is regulated by kinase cascade in the Hippo pathway, which phosphorylates serine residues in specific LATS recognition motifs. Activation of YAP has been observed to impact both the size and quantity of cells. This research investigates the effects of YAP5SA on proliferation, apoptosis, and productivity in CHO-K1 cells. YAP5SA, with mutations in all five LATS-target sites, is selected for its heightened activity and resistance to repression through the Hippo-LATS1/2 kinase signaling pathway. Plasmid harboring YAP5SA was transfected into EPO-CHO and the influence of YAP5SA overexpression was investigated. According to our findings, transfection of EPO-CHO cells with YAP5SA exhibited a substantial enhancement in CHO cell productivity, resulting in a 3-fold increase in total protein and EPO, as well as a 1.5-fold increase in specific productivity. Additionally, it significantly contributes in augmenting viability, size, and proliferation. Overall, the findings of this study exemplify the potential of utilizing YAP5SA to impact particular cellular mechanisms, thereby presenting an avenue for customizing cells to fulfill production demands. KEY POINTS: • YAP5SA in CHO cells boosts growth, reduces apoptosis, and significantly improves productivity. • YAP5SA regulates genes involved in proliferation, survival, and mTOR activation. • YAP5SA increases productivity by improving cell cycle, c-MYC expression, and mTOR pathway.
Collapse
Affiliation(s)
- Farnaz Roshanmehr
- Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Shahriyar Abdoli
- Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Zahra Bazi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Maryam Jari
- Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Shahbazi
- Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
- Arya Tina Gene (ATG), Biopharmaceutical Company, Gorgan, Iran.
| |
Collapse
|
12
|
Deng R, Zhu Y, Liu K, Zhang Q, Hu S, Wang M, Zhang Y. Genetic loss of Nrf1 and Nrf2 leads to distinct metabolism reprogramming of HepG2 cells by opposing regulation of the PI3K-AKT-mTOR signalling pathway. Bioorg Chem 2024; 145:107212. [PMID: 38377819 DOI: 10.1016/j.bioorg.2024.107212] [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: 11/05/2023] [Revised: 02/10/2024] [Accepted: 02/12/2024] [Indexed: 02/22/2024]
Abstract
As a vital hallmarker of cancer, the metabolic reprogramming has been shown to play a pivotal role in tumour occurrence, metastasis and drug resistance. Amongst a vast variety of signalling molecules and metabolic enzymes involved in the regulation of cancer metabolism, two key transcription factors Nrf1 and Nrf2 are required for redox signal transduction and metabolic homeostasis. However, the regulatory effects of Nrf1 and Nrf2 (both encoded by Nfe2l1 and Nfe2l2, respectively) on the metabolic reprogramming of hepatocellular carcinoma cells have been not well understood to date. Here, we found that the genetic deletion of Nrf1 and Nrf2 from HepG2 cells resulted in distinct metabolic reprogramming. Loss of Nrf1α led to enhanced glycolysis, reduced mitochondrial oxygen consumption, enhanced gluconeogenesis and activation of the pentose phosphate pathway in the hepatocellular carcinoma cells. By striking contrast, loss of Nrf2 attenuated the glycolysis and gluconeogenesis pathways, but with not any significant effects on the pentose phosphate pathway. Moreover, knockout of Nrf1α also caused fat deposition and increased amino acid synthesis and transport, especially serine synthesis, whilst Nrf2 deficiency did not cause fat deposition, but attenuated amino acid synthesis and transport. Further experiments revealed that such distinctive metabolic programming of between Nrf1α-/- and Nrf2-/- resulted from substantial activation of the PI3K-AKT-mTOR signalling pathway upon the loss of Nrf1, leading to increased expression of critical genes for the glucose uptake, glycolysis, the pentose phosphate pathway, and the de novo lipid synthesis, whereas deficiency of Nrf2 resulted in the opposite phenomenon by inhibiting the PI3K-AKT-mTOR pathway. Altogether, these provide a novel insight into the cancer metabolic reprogramming and guide the exploration of a new strategy for targeted cancer therapy.
Collapse
Affiliation(s)
- Rongzhen Deng
- Bioengineering College and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing 402260, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China
| | - Yuping Zhu
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China; school of Basic Medicine, Guizhou Medical University, No. 6 Aokang Avenue, Gui'an New District, Guizhou 561113, China
| | - Keli Liu
- Bioengineering College and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing 402260, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China
| | - Qun Zhang
- Bioengineering College and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing 402260, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China
| | - Shaofan Hu
- Bioengineering College and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing 402260, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China
| | - Meng Wang
- Bioengineering College and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China
| | - Yiguo Zhang
- Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing 402260, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing 400044, China.
| |
Collapse
|
13
|
Gao Y, Gong Y, Lu J, Hao H, Shi X. Targeting YAP1 to improve the efficacy of immune checkpoint inhibitors in liver cancer: mechanism and strategy. Front Immunol 2024; 15:1377722. [PMID: 38550587 PMCID: PMC10972981 DOI: 10.3389/fimmu.2024.1377722] [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] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 03/04/2024] [Indexed: 04/02/2024] Open
Abstract
Liver cancer is the third leading of tumor death, including hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). Immune checkpoint inhibitors (ICIs) are yielding much for sufferers to hope for patients, but only some patients with advanced liver tumor respond. Recent research showed that tumor microenvironment (TME) is critical for the effectiveness of ICIs in advanced liver tumor. Meanwhile, metabolic reprogramming of liver tumor leads to immunosuppression in TME. These suggest that regulating the abnormal metabolism of liver tumor cells and firing up TME to turn "cold tumor" into "hot tumor" are potential strategies to improve the therapeutic effect of ICIs in liver tumor. Previous studies have found that YAP1 is a potential target to improve the efficacy of anti-PD-1 in HCC. Here, we review that YAP1 promotes immunosuppression of TME, mainly due to the overstimulation of cytokines in TME by YAP1. Subsequently, we studied the effects of YAP1 on metabolic reprogramming in liver tumor cells, including glycolysis, gluconeogenesis, lipid metabolism, arachidonic acid metabolism, and amino acid metabolism. Lastly, we summarized the existing drugs targeting YAP1 in the treatment of liver tumor, including some medicines from natural sources, which have the potential to improve the efficacy of ICIs in the treatment of liver tumor. This review contributed to the application of targeted YAP1 for combined therapy with ICIs in liver tumor patients.
Collapse
Affiliation(s)
- Yuting Gao
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Yi Gong
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Junlan Lu
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Huiqin Hao
- Chinese Medicine Gene Expression Regulation Laboratory, State Administration of Traditional Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, China
- Basic Laboratory of Integrated Traditional Chinese and Western, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Xinli Shi
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, China
| |
Collapse
|
14
|
Fan Y, Xue H, Li Z, Huo M, Gao H, Guan X. Exploiting the Achilles' heel of cancer: disrupting glutamine metabolism for effective cancer treatment. Front Pharmacol 2024; 15:1345522. [PMID: 38510646 PMCID: PMC10952006 DOI: 10.3389/fphar.2024.1345522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 02/23/2024] [Indexed: 03/22/2024] Open
Abstract
Cancer cells have adapted to rapid tumor growth and evade immune attack by reprogramming their metabolic pathways. Glutamine is an important nitrogen resource for synthesizing amino acids and nucleotides and an important carbon source in the tricarboxylic acid (TCA) cycle and lipid biosynthesis pathway. In this review, we summarize the significant role of glutamine metabolism in tumor development and highlight the vulnerabilities of targeting glutamine metabolism for effective therapy. In particular, we review the reported drugs targeting glutaminase and glutamine uptake for efficient cancer treatment. Moreover, we discuss the current clinical test about targeting glutamine metabolism and the prospective direction of drug development.
Collapse
Affiliation(s)
- Yuxin Fan
- Department of Clinical Laboratory Diagnostics, School of Medical Technology, Beihua University, Jilin City, China
- Department of Basic Medicine, Medical School, Taizhou University, Taizhou, Zhejiang Province, China
| | - Han Xue
- Department of Clinical Laboratory Diagnostics, School of Medical Technology, Beihua University, Jilin City, China
- Department of Basic Medicine, Medical School, Taizhou University, Taizhou, Zhejiang Province, China
| | - Zhimin Li
- Department of Clinical Laboratory Diagnostics, School of Medical Technology, Beihua University, Jilin City, China
- Department of Basic Medicine, Medical School, Taizhou University, Taizhou, Zhejiang Province, China
| | - Mingge Huo
- Department of Clinical Laboratory Diagnostics, School of Medical Technology, Beihua University, Jilin City, China
- Department of Basic Medicine, Medical School, Taizhou University, Taizhou, Zhejiang Province, China
| | - Hongxia Gao
- Department of Clinical Laboratory Diagnostics, School of Medical Technology, Beihua University, Jilin City, China
| | - Xingang Guan
- Department of Basic Medicine, Medical School, Taizhou University, Taizhou, Zhejiang Province, China
| |
Collapse
|
15
|
Schiavoni G, Messina B, Scalera S, Memeo L, Colarossi C, Mare M, Blandino G, Ciliberto G, Bon G, Maugeri-Saccà M. Role of Hippo pathway dysregulation from gastrointestinal premalignant lesions to cancer. J Transl Med 2024; 22:213. [PMID: 38424512 PMCID: PMC10903154 DOI: 10.1186/s12967-024-05027-8] [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: 07/21/2023] [Accepted: 02/25/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND First identified in Drosophila melanogaster, the Hippo pathway is considered a major regulatory cascade controlling tissue homeostasis and organ development. Hippo signaling components include kinases whose activity regulates YAP and TAZ final effectors. In response to upstream stimuli, YAP and TAZ control transcriptional programs involved in cell proliferation, cytoskeletal reorganization and stemness. MAIN TEXT While fine tuning of Hippo cascade components is essential for maintaining the balance between proliferative and non-proliferative signals, pathway signaling is frequently dysregulated in gastrointestinal cancers. Also, YAP/TAZ aberrant activation has been described in conditions characterized by chronic inflammation that precede cancer development, suggesting a role of Hippo effectors in triggering carcinogenesis. In this review, we summarize the architecture of the Hippo pathway and discuss the involvement of signaling cascade unbalances in premalignant lesions of the gastrointestinal tract, providing a focus on the underlying molecular mechanisms. CONCLUSIONS The biology of premalignant Hippo signaling dysregulation needs further investigation in order to elucidate the evolutionary trajectories triggering cancer inititation and develop effective early therapeutic strategies targeting the Hippo/YAP pathway.
Collapse
Affiliation(s)
- Giulia Schiavoni
- Clinical Trial Center, Biostatistics and Bioinformatics Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Beatrice Messina
- Clinical Trial Center, Biostatistics and Bioinformatics Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Stefano Scalera
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Lorenzo Memeo
- Pathology Unit, Mediterranean Institute of Oncology, Viagrande, Italy
| | | | - Marzia Mare
- Medical Oncology Unit, Mediterranean Institute of Oncology, Viagrande, Italy
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Messina, Italy
| | - Giovanni Blandino
- Translational Oncology Research Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Gennaro Ciliberto
- Scientific Directorate, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giulia Bon
- Cellular Network and Molecular Therapeutic Target Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.
| | - Marcello Maugeri-Saccà
- Clinical Trial Center, Biostatistics and Bioinformatics Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
16
|
Yan R, Cai H, Zhou X, Bao G, Bai Z, Ge RL. Hypoxia-inducible factor-2α promotes fibrosis in non-alcoholic fatty liver disease by enhancing glutamine catabolism and inhibiting yes-associated protein phosphorylation in hepatic stellate cells. Front Endocrinol (Lausanne) 2024; 15:1344971. [PMID: 38501098 PMCID: PMC10946064 DOI: 10.3389/fendo.2024.1344971] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/25/2024] [Indexed: 03/20/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has a high global prevalence and affects approximately one-third of adults, owing to high-fat dietary habits and a sedentary lifestyle. The role of hypoxia-inducible factor 2α (HIF-2α) in NAFLD progression remains unknown. This study aimed to investigate the effects of chronic hypoxia on NAFLD progression by examining the role of hypoxia-inducible factor 2α (HIF-2α) activation and that of hepatic stellate cell (HSC)-derived myofibroblasts through glutaminolysis. We hypothesised that hypoxia exacerbates NAFLD by promoting HIF-2α upregulation and inhibiting phosphorylated yes-associated protein (YAP), and that increasing YAP expression enhances HSC-derived myofibroblasts. We studied patients with NAFLD living at high altitudes, as well as animal models and cultured cells. The results revealed significant increases in HSC-derived myofibroblasts and collagen accumulation caused by HIF-2α and YAP upregulation, both in patients and in a mouse model for hypoxia and NAFLD. HIF-2α and HIF-2α-dependent YAP downregulation reduced HSC activation and myofibroblast levels in persistent chronic hypoxia. Furthermore, hypoxia-induced HIF-2α upregulation promoted YAP and inhibited YAP phosphorylation, leading to glutaminase 1 (GLS1), SLC38A1, α-SMA, and Collagen-1 overexpression. Additionally, hypoxia restored mitochondrial adenosine triphosphate production and reactive oxygen species (ROS) overproduction. Thus, chronic hypoxia-induced HIF-2α activation enhances fibrosis and NAFLD progression by restoring mitochondrial ROS production and glutaminase-1-induced glutaminolysis, which is mediated through the inhibition of YAP phosphorylation and increased YAP nuclear translocation. In summary, HIF-2α plays a pivotal role in NAFLD progression during chronic hypoxia.
Collapse
Affiliation(s)
- Ranran Yan
- Qinghai-Utah Joint Key Lab for High-altitude Medicine, Medical College of Qinghai University, Xining, China
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, China
- Key Laboratory of High-Altitude Medicine in Qinghai University, Ministry of Education, Xining, China
- Key Laboratory for Application of High-Altitude Medicine in Qinghai Province, Xining, China
| | - Hao Cai
- Oncology Department, The Fifth People’s Hospital of Qinghai Provincial, Xining, China
| | - Xiaofeng Zhou
- Affiliated Hospital of Qinghai University, Xining, China
| | - Guodan Bao
- Qinghai-Utah Joint Key Lab for High-altitude Medicine, Medical College of Qinghai University, Xining, China
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, China
- Key Laboratory of High-Altitude Medicine in Qinghai University, Ministry of Education, Xining, China
- Affiliated Hospital of Qinghai University, Xining, China
| | - Zhenzhong Bai
- Qinghai-Utah Joint Key Lab for High-altitude Medicine, Medical College of Qinghai University, Xining, China
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, China
- Key Laboratory of High-Altitude Medicine in Qinghai University, Ministry of Education, Xining, China
- Key Laboratory for Application of High-Altitude Medicine in Qinghai Province, Xining, China
| | - Ri-li Ge
- Qinghai-Utah Joint Key Lab for High-altitude Medicine, Medical College of Qinghai University, Xining, China
- Research Center for High Altitude Medicine, Medical College of Qinghai University, Xining, China
- Key Laboratory of High-Altitude Medicine in Qinghai University, Ministry of Education, Xining, China
- Key Laboratory for Application of High-Altitude Medicine in Qinghai Province, Xining, China
| |
Collapse
|
17
|
Song HS, Ha SY, Kim JY, Kim M, Choi JH. The effect of genetic variants of SLC22A18 on proliferation, migration, and invasion of colon cancer cells. Sci Rep 2024; 14:3925. [PMID: 38366023 PMCID: PMC10873386 DOI: 10.1038/s41598-024-54658-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/15/2024] [Indexed: 02/18/2024] Open
Abstract
Solute carrier family (SLC) transporters are expressed in the digestive system and play important roles in maintaining physiological functions in the body. In addition, SLC transporters act as oncoproteins or tumor-suppressor proteins during the development, progression, and metastasis of various digestive system cancers. SLC22A18, a member of the SLC22 gene family, is an orphan transporter with an unknown endogenous substrate. Previous study revealed that SLC22A18 is downregulated in colorectal cancer tissues and that it acts as a suppressor in colorectal cancer, although the effects of SLC22A18 variants on colon cancer cell proliferation, migration, and invasion are unknown. Therefore, in this study, we identified SLC22A18 variants found in multiple populations by searching public databases and determined the in vitro effects of these missense variations on transporter expression and cancer progression. Our results indicated that three missense SLC22A18 variants-p.Ala6Thr, p.Arg12Gln, and p.Arg86His-had significantly lower cell expression than the wild type, possibly owing to intracellular degradation. Furthermore, these three variants caused significantly higher proliferation, migration, and invasion of colon cancer cells than the wild type. Our findings suggest that missense variants of SLC22A18 can potentially serve as biomarkers or prognostic tools that enable clinicians to predict colorectal cancer progression.
Collapse
Affiliation(s)
- Hyo Sook Song
- Department of Pharmacology, Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Republic of Korea
| | - Seung Yeon Ha
- Department of Pharmacology, Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Republic of Korea
| | - Jin-Young Kim
- Department of Pharmacology, Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Republic of Korea
| | - Minsuk Kim
- Department of Pharmacology, Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Republic of Korea
| | - Ji Ha Choi
- Department of Pharmacology, Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul, 07804, Republic of Korea.
| |
Collapse
|
18
|
Edwards AC, Stalnecker CA, Morales AJ, Taylor KE, Klomp JE, Klomp JA, Waters AM, Sudhakar N, Hallin J, Tang TT, Olson P, Post L, Christensen JG, Cox AD, Der CJ. TEAD Inhibition Overcomes YAP1/TAZ-Driven Primary and Acquired Resistance to KRASG12C Inhibitors. Cancer Res 2023; 83:4112-4129. [PMID: 37934103 PMCID: PMC10821578 DOI: 10.1158/0008-5472.can-23-2994] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 11/08/2023]
Abstract
Primary/intrinsic and treatment-induced acquired resistance limit the initial response rate to and long-term efficacy of direct inhibitors of the KRASG12C mutant in cancer. To identify potential mechanisms of resistance, we applied a CRISPR/Cas9 loss-of-function screen and observed loss of multiple components of the Hippo tumor suppressor pathway, which acts to suppress YAP1/TAZ-regulated gene transcription. YAP1/TAZ activation impaired the antiproliferative and proapoptotic effects of KRASG12C inhibitor (G12Ci) treatment in KRASG12C-mutant cancer cell lines. Conversely, genetic suppression of YAP1/WWTR1 (TAZ) enhanced G12Ci sensitivity. YAP1/TAZ activity overcame KRAS dependency through two distinct TEAD transcription factor-dependent mechanisms, which phenocopy KRAS effector signaling. First, TEAD stimulated ERK-independent transcription of genes normally regulated by ERK (BIRC5, CDC20, ECT2, FOSL1, and MYC) to promote progression through the cell cycle. Second, TEAD caused activation of PI3K-AKT-mTOR signaling to overcome apoptosis. G12Ci treatment-induced acquired resistance was also caused by YAP1/TAZ-TEAD activation. Accordingly, concurrent treatment with pharmacologic inhibitors of TEAD synergistically enhanced KRASG12C inhibitor antitumor activity in vitro and prolonged tumor suppression in vivo. In summary, these observations reveal YAP1/TAZ-TEAD signaling as a crucial driver of primary and acquired resistance to KRAS inhibition and support the use of TEAD inhibitors to enhance the antitumor efficacy of KRAS-targeted therapies. SIGNIFICANCE YAP1/TAZ-TEAD activation compensates for loss of KRAS effector signaling, establishing a mechanistic basis for concurrent inhibition of TEAD to enhance the efficacy of KRASG12C-selective inhibitor treatment of KRASG12C-mutant cancers. See related commentary by Johnson and Haigis, p. 4005.
Collapse
Affiliation(s)
- A. Cole Edwards
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Clint A. Stalnecker
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Alexis Jean Morales
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Khalilah E. Taylor
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jennifer E. Klomp
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jeffrey A. Klomp
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Andrew M. Waters
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | - Jill Hallin
- Mirati Therapeutics, Inc., San Diego, California
| | | | - Peter Olson
- Mirati Therapeutics, Inc., San Diego, California
| | - Leonard Post
- Vivace Therapeutics, Inc., San Mateo, California
| | | | - Adrienne D. Cox
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Channing J. Der
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
19
|
Deng H, Jia Q, Ming X, Sun Y, Lu Y, Liu L, Zhou J. Hippo pathway in intestinal diseases: focusing on ferroptosis. Front Cell Dev Biol 2023; 11:1291686. [PMID: 38130953 PMCID: PMC10734691 DOI: 10.3389/fcell.2023.1291686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
The incidence of intestinal diseases, such as inflammatory bowel disease, gastric cancer, and colorectal cancer, has steadily increased over the past decades. The Hippo pathway is involved in cell proliferation, tissue and organ damage, energy metabolism, tumor formation, and other physiologic processes. Ferroptosis is a form of programmed cell death characterized by the accumulation of iron and lipid peroxides. The Hippo pathway and ferroptosis are associated with various intestinal diseases; however, the crosstalk between them is unclear. This review elaborates on the current research on the Hippo pathway and ferroptosis in the context of intestinal diseases. We summarized the connection between the Hippo pathway and ferroptosis to elucidate the underlying mechanism by which these pathways influence intestinal diseases. We speculate that a mutual regulatory mechanism exists between the Hippo pathway and ferroptosis and these two pathways interact in several ways to regulate intestinal diseases.
Collapse
Affiliation(s)
- Hongwei Deng
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Luzhou, China
| | - Qiuting Jia
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Luzhou, China
| | - Xin Ming
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Luzhou, China
- School of Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Yuxin Sun
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Luzhou, China
- School of Basic Medicine, Southwest Medical University, Luzhou, China
| | - Yuxuan Lu
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Luzhou, China
| | - Li Liu
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Luzhou, China
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Jun Zhou
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Luzhou, China
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
20
|
Lv L, Zhou X. Targeting Hippo signaling in cancer: novel perspectives and therapeutic potential. MedComm (Beijing) 2023; 4:e375. [PMID: 37799806 PMCID: PMC10547939 DOI: 10.1002/mco2.375] [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: 04/19/2023] [Revised: 08/23/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023] Open
Abstract
As highly conserved among diverse species, Hippo signaling pathway regulates various biological processes, including development, cell proliferation, stem cell function, tissue regeneration, homeostasis, and organ size. Studies in the last two decades have provided a good framework for how these fundamental functions of Hippo signaling are tightly regulated by a network with numerous intracellular and extracellular factors. The Hippo signaling pathway, when dysregulated, may lead to a wide variety of diseases, especially cancer. There is growing evidence demonstrating that dysregulated Hippo signaling is closely associated with tumorigenesis, cancer cell invasion, and migration, as well as drug resistance. Therefore, the Hippo pathway is considered an appealing therapeutic target for the treatment of cancer. Promising novel agents targeting the Hippo signaling pathway for cancers have recently emerged. These novel agents have shown antitumor activity in multiple cancer models and demonstrated therapeutic potential for cancer treatment. However, the detailed molecular basis of the Hippo signaling-driven tumor biology remains undefined. Our review summarizes current advances in understanding the mechanisms by which Hippo signaling drives tumorigenesis and confers drug resistance. We also propose strategies for future preclinical and clinical development to target this pathway.
Collapse
Affiliation(s)
- Liemei Lv
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongChina
| | - Xiangxiang Zhou
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongChina
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Branch of National Clinical Research Center for Hematologic DiseasesJinanShandongChina
- National Clinical Research Center for Hematologic Diseasesthe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
21
|
Wei Y, Hui VLZ, Chen Y, Han R, Han X, Guo Y. YAP/TAZ: Molecular pathway and disease therapy. MedComm (Beijing) 2023; 4:e340. [PMID: 37576865 PMCID: PMC10412783 DOI: 10.1002/mco2.340] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/27/2023] [Accepted: 07/04/2023] [Indexed: 08/15/2023] Open
Abstract
The Yes-associated protein and its transcriptional coactivator with PDZ-binding motif (YAP/TAZ) are two homologous transcriptional coactivators that lie at the center of a key regulatory network of Hippo, Wnt, GPCR, estrogen, mechanical, and metabolism signaling. YAP/TAZ influences the expressions of downstream genes and proteins as well as enzyme activity in metabolic cycles, cell proliferation, inflammatory factor expression, and the transdifferentiation of fibroblasts into myofibroblasts. YAP/TAZ can also be regulated through epigenetic regulation and posttranslational modifications. Consequently, the regulatory function of these mechanisms implicates YAP/TAZ in the pathogenesis of metabolism-related diseases, atherosclerosis, fibrosis, and the delicate equilibrium between cancer progression and organ regeneration. As such, there arises a pressing need for thorough investigation of YAP/TAZ in clinical settings. In this paper, we aim to elucidate the signaling pathways that regulate YAP/TAZ and explore the mechanisms of YAP/TAZ-induce diseases and their potential therapeutic interventions. Furthermore, we summarize the current clinical studies investigating treatments targeting YAP/TAZ. We also address the limitations of existing research on YAP/TAZ and propose future directions for research. In conclusion, this review aims to provide fresh insights into the signaling mediated by YAP/TAZ and identify potential therapeutic targets to present innovative solutions to overcome the challenges associated with YAP/TAZ.
Collapse
Affiliation(s)
- Yuzi Wei
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Victoria Lee Zhi Hui
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Yilin Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduSichuanChina
- Department of OrthodonticsWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Ruiying Han
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduSichuanChina
- Department of OrthodonticsWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Xianglong Han
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduSichuanChina
- Department of OrthodonticsWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Yongwen Guo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduSichuanChina
- Department of OrthodonticsWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
- Department of OrthodonticsLanzhou Stomatological HospitalLanzhouGansuChina
| |
Collapse
|
22
|
Liao X, Li X, Liu R. Extracellular-matrix mechanics regulate cellular metabolism: A ninja warrior behind mechano-chemo signaling crosstalk. Rev Endocr Metab Disord 2023; 24:207-220. [PMID: 36385696 DOI: 10.1007/s11154-022-09768-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/21/2022] [Indexed: 11/18/2022]
Abstract
Mechanical forces are the indispensable constituent of environmental cues, such as gravity, barometric pressure, vibration, and contact with bodies, which are involved in pattern and organogenesis, providing mechanical input to tissues and determining the ultimate fate of cells. Extracellular matrix (ECM) stiffness, the slow elastic force, carries the external physical force load onto the cell or outputs the internal force exerted by the cell and its neighbors into the environment. Accumulating evidence illustrates the pivotal role of ECM stiffness in the regulation of organogenesis, maintenance of tissue homeostasis, and the development of multiple diseases, which is largely fulfilled through its systematical impact on cellular metabolism. This review summarizes the establishment and regulation of ECM stiffness, the mechanisms underlying how ECM stiffness is sensed by cells and signals to modulate diverse cell metabolic pathways, and the physiological and pathological significance of the ECM stiffness-cell metabolism axis.
Collapse
Affiliation(s)
- Xiaoyu Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, 14, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China
| | - Xin Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, 14, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China
| | - Rui Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, 14, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
23
|
Zhu N, Yang R, Wang X, Yuan L, Li X, Wei F, Zhang L. The Hippo signaling pathway: from multiple signals to the hallmarks of cancers. Acta Biochim Biophys Sin (Shanghai) 2023. [PMID: 36942989 DOI: 10.3724/abbs.2023035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
Abstract
Evolutionarily conserved, the Hippo signaling pathway is critical in regulating organ size and tissue homeostasis. The activity of this pathway is tightly regulated under normal circumstances, since its physical function is precisely maintained to control the rate of cell proliferation. Failure of maintenance leads to a variety of tumors. Our understanding of the mechanism of Hippo dysregulation and tumorigenesis is becoming increasingly precise, relying on the emergence of upstream inhibitor or activator and the connection linking Hippo target genes, mutations, and related signaling pathways with phenotypes. In this review, we summarize recent reports on the signaling network of the Hippo pathway in tumorigenesis and progression by exploring its critical mechanisms in cancer biology and potential targeting in cancer therapy.
Collapse
Affiliation(s)
- Ning Zhu
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ruizeng Yang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiaodong Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Liang Yuan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Xiaoyu Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Fang Wei
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lei Zhang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| |
Collapse
|
24
|
Golubev DA, Zemskaya NV, Gorbunova AA, Kukuman DV, Moskalev A, Shaposhnikov MV. Studying the Geroprotective Properties of YAP/TAZ Signaling Inhibitors on Drosophila melanogaster Model. Int J Mol Sci 2023; 24:ijms24066006. [PMID: 36983079 PMCID: PMC10058302 DOI: 10.3390/ijms24066006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/28/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
The transcriptional coactivators Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are the main downstream effectors of the evolutionarily conserved Hippo signaling pathway. YAP/TAZ are implicated in the transcriptional regulation of target genes that are involved in a wide range of key biological processes affecting tissue homeostasis and play dual roles in the aging process, depending on the cellular and tissue context. The aim of the present study was to investigate whether pharmacological inhibitors of Yap/Taz increase the lifespan of Drosophila melanogaster. Real-time qRT-PCR was performed to measure the changes in the expression of Yki (Yorkie, the Drosophila homolog of YAP/TAZ) target genes. We have revealed a lifespan-increasing effect of YAP/TAZ inhibitors that was mostly associated with decreased expression levels of the wg and E2f1 genes. However, further analysis is required to understand the link between the YAP/TAZ pathway and aging.
Collapse
Affiliation(s)
- Denis A Golubev
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| | - Nadezhda V Zemskaya
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| | - Anastasia A Gorbunova
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| | - Daria V Kukuman
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| | - Alexey Moskalev
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| | - Mikhail V Shaposhnikov
- Laboratory of Geroprotective and Radioprotective Technologies, Institute of Biology, Komi Science Center, Ural Branch, Russian Academy of Sciences, 167982 Syktyvkar, Russia
| |
Collapse
|
25
|
Integrated analysis of ferroptosis-related gene signature for overall survival prediction in Asian patients with hepatocellular carcinoma. Clin Transl Oncol 2023; 25:721-730. [PMID: 36319928 DOI: 10.1007/s12094-022-02977-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 10/07/2022] [Indexed: 01/05/2023]
Abstract
PURPOSE Hepatocellular carcinoma (HCC) is one of the most prevalent types of cancers in Asia. Accumulating evidence suggests that ferroptosis is a non-apoptotic form of cell death, and has played an important role in cancer biology. METHODS Based on the manually curated ferroptosis-related gene set and TCGA-LIHC dataset of Asian patients, we used DESeq2, Kaplan-Meier analysis, and univariate Cox regression to identify differentially expressed ferroptosis-related genes with significantly prognostic capacity. A risk signature was constructed based on the selected genes for predicting the survival of HCC patients in Asia. The survival prediction accuracy was confirmed by the time-dependent receiver operating characteristic (ROC) curve analysis. Gene set variation analysis (GSVA) was used to explore the functional associations of the signature. Ferroptosis potential index (FPI) and xCell algorithm was applied to quantify ferroptosis and immune cell infiltration, respectively. Two independent datasets from the GEO and the ICGC database were used for external validation. RESULTS The ferroptosis-related signature could accurately predict the survival outcomes of HCC patients in Asian (p value < 0.0001). We showed that the signature was an independent factor and was beneficial in elevating risk stratification of current clinicopathologic features, such as the amount of alpha-fetoprotein (AFP) and residual tumor classification. Functional characterization showed that critical processes in tumorigenesis belonged to the high-risk groups, for example inflammatory response, which may be the main driver of HCC. The high-risk group had higher FPIs and infiltrations of macrophages and T-helper cells than the low-risk group. Furthermore, two independent cohorts confirmed the prognostic value of our signature. CONCLUSION Overall, our results demonstrated potential application of ferroptosis-related genes as independent biomarkers in Asian HCC patients. Targeting ferroptosis may be clinically useful beyond known clinicopathological factors and provide benefit in immunotherapy.
Collapse
|
26
|
Prognostic 7-SLC-Gene Signature Identified via Weighted Gene Co-Expression Network Analysis for Patients with Hepatocellular Carcinoma. JOURNAL OF ONCOLOGY 2023; 2023:4364654. [PMID: 36844876 PMCID: PMC9957622 DOI: 10.1155/2023/4364654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/13/2022] [Accepted: 11/24/2022] [Indexed: 02/19/2023]
Abstract
Background Solute carrier (SLC) proteins play an important role in tumor metabolism. But SLC-associated genes' prognostic significance in hepatocellular carcinoma (HCC) remained elusive. We identified SLC-related factors and developed an SLC-related classifier to predict and improve HCC prognosis and treatment. Methods From the TCGA database, corresponding clinical data and mRNA expression profiles of 371 HCC patients were acquired, and those of 231 tumor samples were derived from the ICGC database. Genes associated with clinical features were filtered using weighted gene correlation network analysis (WGCNA). Next, univariate LASSO Cox regression studies developed SLC risk profiles, with the ICGC cohort data being used in validation. Result Univariate Cox regression analysis revealed that 31 SLC genes (P < 0.05) were related to HCC prognosis. 7 (SLC22A25, SLC2A2, SLC41A3, SLC44A1, SLC48A1, SLC4A2, and SLC9A3R1) of these genes were applied in developing a SLC gene prognosis model. Samples were classified into the low-andhigh-risk groups by the prognostic signature, with those in the high-risk group showing a significantly worse prognosis (P < 0.001 in the TCGA cohort and P=0.0068 in the ICGC cohort). ROC analysis validated the signature's prediction power. In addition, functional analyses showed enrichment of immune-related pathways and different immune status between the two risk groups. Conclusion The 7-SLC-gene prognostic signature established in this study helped predict the prognosis, and was also correlated with the tumor immune status and infiltration of different immune cells in the tumor microenvironment. The current findings may provide important clinical indications for proposing a novel combination therapy consists of targeted anti-SLC therapy and immunotherapy for HCC patients.
Collapse
|
27
|
Barker BE, Hanlon MM, Marzaioli V, Smith CM, Cunningham CC, Fletcher JM, Veale DJ, Fearon U, Canavan M. The mammalian target of rapamycin contributes to synovial fibroblast pathogenicity in rheumatoid arthritis. Front Med (Lausanne) 2023; 10:1029021. [PMID: 36817783 PMCID: PMC9936094 DOI: 10.3389/fmed.2023.1029021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023] Open
Abstract
Objectives The mammalian target of Rapamycin (mTOR) is a metabolic master regulator of both innate and adaptive immunity; however, its exact role in stromal cell biology is unknown. In this study we explored the role of the mTOR pathway on Rheumatoid Arthritis synovial fibroblast (RASF) metabolism and activation and determined if crosstalk with the Hippo-YAP pathway mediates their effects. Methods Primary RA synovial fibroblasts (RASF) were cultured with TNFα alone or in combination with the mTOR inhibitor Rapamycin or YAP inhibitor Verteporfin. Chemokine production, matrix metalloproteinase (MMP) production, and adhesion marker expression were quantified by real-time PCR, ELISA, and/or Flow Cytometry. Invasion assays were performed using Transwell invasion chambers, while wound repair assays were used to assess RASF migration. Cellular bioenergetics was assessed using the Seahorse XFe96 Analyzer. Key metabolic genes (GLUT-1, HK2, G6PD) were measured using real-time PCR. Reanalysis of RNA-Seq analysis was performed on RA (n = 151) and healthy control (HC) (n = 28) synovial tissue biopsies to detect differential gene and pathway expression. The expression of YAP was measured by Western Blot. Results Transcriptomic analysis of healthy donor and RA synovial tissue revealed dysregulated expression of several key components of the mTOR pathway in RA. Moreover, the expression of phospho-ribosomal protein S6 (pS6), the major downstream target of mTOR is specifically increased in RA synovial fibroblasts compared to healthy tissue. In the presence of TNFα, RASF display heightened phosphorylation of S6 and are responsive to mTOR inhibition via Rapamycin. Rapamycin effectively alters RASF cellular bioenergetics by inhibiting glycolysis and the expression of rate limiting glycolytic enzymes. Furthermore, we demonstrate a key role for mTOR signaling in uniquely mediating RASF migratory and invasive mechanisms, which are significantly abrogated in the presence of Rapamycin. Finally, we report a significant upregulation in several genes involved in the Hippo-YAP pathway in RA synovial tissue, which are predicted to converge with the mTOR pathway. We demonstrate crosstalk between the mTOR and YAP pathways in mediating RASF invasive mechanism whereby Rapamycin significantly abrogates YAP expression and YAP inhibition significantly inhibits RASF invasiveness. Conclusion mTOR drives pathogenic mechanisms in RASF an effect which is in part mediated via crosstalk with the Hippo-YAP pathway.
Collapse
Affiliation(s)
- Brianne E. Barker
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland,Translational Immunopathology, School of Biochemistry & Immunology and School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Megan M. Hanlon
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St. Vincent’s University Hospital, Dublin, Ireland
| | - Viviana Marzaioli
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St. Vincent’s University Hospital, Dublin, Ireland
| | - Conor M. Smith
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Clare C. Cunningham
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St. Vincent’s University Hospital, Dublin, Ireland
| | - Jean M. Fletcher
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland,School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland
| | - Douglas J. Veale
- EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St. Vincent’s University Hospital, Dublin, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland,EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St. Vincent’s University Hospital, Dublin, Ireland
| | - Mary Canavan
- Translational Immunopathology, School of Biochemistry & Immunology and School of Medicine, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin, Ireland,*Correspondence: Mary Canavan,
| |
Collapse
|
28
|
Li P, Mi Q, Yan S, Xie Y, Cui Z, Zhang S, Wang Y, Gao H, Wang Y, Li J, Du L, Wang C. Characterization of circSCL38A1 as a novel oncogene in bladder cancer via targeting ILF3/TGF-β2 signaling axis. Cell Death Dis 2023; 14:59. [PMID: 36697384 PMCID: PMC9876890 DOI: 10.1038/s41419-023-05598-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023]
Abstract
The regulatory role of circRNAs in cancer metastasis has become a focused issue in recent years. To date, however, the discovery of novel functional circRNAs and their regulatory mechanisms via binding with RBPs in bladder cancer (BC) are still lacking. Here, we screened out circSLC38A1 based on our sequencing data and followed validation with clinical tissue samples and cell lines. Functional assays showed that circSLC38A1 promoted BC cell invasion in vitro and lung metastasis of mice in vivo. By conducting RNA pull-down, mass spectrum, and RIP assays, circSLC38A1 was found to interact with Interleukin enhancer-binding factor 3 (ILF3), and stabilize ILF3 protein via modulating the ubiquitination process. By integrating our CUT&Tag-seq and RNA-seq data, TGF-β2 was identified as the functional target of the circSLC38A1-ILF3 complex. In addition, m6A methylation was enriched in circSLC38A1 and contributed to its upregulation. Clinically, circSLC38A1 was identified in serum exosomes of BC patients and could distinguish BC patients from healthy individuals with a diagnostic accuracy of 0.878. Thus, our study revealed an essential role and clinical significance of circSLC38A1 in BC via activating the transcription of TGF-β2 in an ILF3-dependent manner, extending the understanding of the importance of circRNA-mediated transcriptional regulation in BC metastasis.
Collapse
Affiliation(s)
- Peilong Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
- Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, Shandong, China
| | - Qi Mi
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Suzhen Yan
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
- Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, Shandong, China
| | - Yan Xie
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Zilian Cui
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Shujun Zhang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Yifan Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Huiru Gao
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Yunshan Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
- Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, Shandong, China
- Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, Shandong, China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China.
- Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, Shandong, China.
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China.
- Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, Shandong, China.
- Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, Shandong, China.
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China.
- Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, Shandong, China.
- Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, Shandong, China.
- Shandong Technology Innovation Center for Big Data and Precision Medicine of Cancer, Jinan, 250033, Shandong, China.
| |
Collapse
|
29
|
HPV16 E6 and E7 Oncoproteins Stimulate the Glutamine Pathway Maintaining Cell Proliferation in a SNAT1-Dependent Fashion. Viruses 2023; 15:v15020324. [PMID: 36851539 PMCID: PMC9964736 DOI: 10.3390/v15020324] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Persistent high-risk human papillomavirus infection is the main risk factor for cervical cancer establishment, where the viral oncogenes E6 and E7 promote a cancerous phenotype. Metabolic reprogramming in cancer involves alterations in glutamine metabolism, also named glutaminolysis, to provide energy for supporting cancer processes including migration, proliferation, and production of reactive oxygen species, among others. The aim of this work was to analyze the effect of HPV16 E6 and E7 oncoproteins on the regulation of glutaminolysis and its contribution to cell proliferation. We found that the E6 and E7 oncoproteins exacerbate cell proliferation in a glutamine-dependent manner. Both oncoproteins increased the levels of transporter SNAT1, as well as GLS2 and GS enzymes; E6 also increased LAT1 transporter protein levels, while E7 increased ASCT2 and xCT. Some of these alterations are also regulated at a transcriptional level. Consistently, the amount of SNAT1 protein decreased in Ca Ski cells when E6 and E7 expression was knocked down. In addition, we demonstrated that cell proliferation was partially dependent on SNAT1 in the presence of glutamine. Interestingly, SNAT1 expression was higher in cervical cancer compared with normal cervical cells. The high expression of SNAT1 was associated with poor overall survival of cervical cancer patients. Our results indicate that HPV oncoproteins exacerbate glutaminolysis supporting the malignant phenotype.
Collapse
|
30
|
Jung J, Park S, Jang Y, Lee SH, Jeong YS, Yim SY, Lee JS. Clinical Significance of Glycolytic Metabolic Activity in Hepatocellular Carcinoma. Cancers (Basel) 2022; 15:186. [PMID: 36612182 PMCID: PMC9818850 DOI: 10.3390/cancers15010186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/17/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
High metabolic activity is a hallmark of cancers, including hepatocellular carcinoma (HCC). However, the molecular features of HCC with high metabolic activity contributing to clinical outcomes and the therapeutic implications of these characteristics are poorly understood. We aimed to define the features of HCC with high metabolic activity and uncover its association with response to current therapies. By integrating gene expression data from mouse liver tissues and tumor tissues from HCC patients (n = 1038), we uncovered three metabolically distinct HCC subtypes that differ in clinical outcomes and underlying molecular biology. The high metabolic subtype is characterized by poor survival, the strongest stem cell signature, high genomic instability, activation of EPCAM and SALL4, and low potential for benefitting from immunotherapy. Interestingly, immune cell analysis showed that regulatory T cells (Tregs) are highly enriched in high metabolic HCC tumors, suggesting that high metabolic activity of cancer cells may trigger activation or infiltration of Tregs, leading to cancer cells' evasion of anti-cancer immune cells. In summary, we identified clinically and metabolically distinct subtypes of HCC, potential biomarkers associated with these subtypes, and a potential mechanism of metabolism-mediated immune evasion by HCC cells.
Collapse
Affiliation(s)
- Joann Jung
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sowon Park
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yeonwoo Jang
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sung-Hwan Lee
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Yonsei University College of Medicine, Yonsei 03722, Republic of Korea
- Division of Hepatobiliary and Pancreas, Department of Surgery, CHA Bundang Medical Center, CHA University, Seongnam 46371, Republic of Korea
| | - Yun Seong Jeong
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sun Young Yim
- Division of Hepatobiliary and Pancreas, Department of Surgery, CHA Bundang Medical Center, CHA University, Seongnam 46371, Republic of Korea
- Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
31
|
Lee SH, Yim SY, Jeong YS, Li QX, Kang SH, Sohn BH, Kumar SV, Shin JH, Choi YR, Shim JJ, Kim H, Kim J, Kim S, Guo S, Johnson RL, Kaseb A, Kang KJ, Chun YS, Jang HJ, Lee BG, Woo HG, Ha MJ, Akbani R, Roberts LR, Wheeler DA, Lee JS. Consensus subtypes of hepatocellular carcinoma associated with clinical outcomes and genomic phenotypes. Hepatology 2022; 76:1634-1648. [PMID: 35349735 PMCID: PMC9519807 DOI: 10.1002/hep.32490] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/24/2022] [Accepted: 03/12/2022] [Indexed: 01/22/2023]
Abstract
BACKGROUND AND AIMS Although many studies revealed transcriptomic subtypes of HCC, concordance of the subtypes are not fully examined. We aim to examine a consensus of transcriptomic subtypes and correlate them with clinical outcomes. APPROACH AND RESULTS By integrating 16 previously established genomic signatures for HCC subtypes, we identified five clinically and molecularly distinct consensus subtypes. STM (STeM) is characterized by high stem cell features, vascular invasion, and poor prognosis. CIN (Chromosomal INstability) has moderate stem cell features, but high genomic instability and low immune activity. IMH (IMmune High) is characterized by high immune activity. BCM (Beta-Catenin with high Male predominance) is characterized by prominent β-catenin activation, low miRNA expression, hypomethylation, and high sensitivity to sorafenib. DLP (Differentiated and Low Proliferation) is differentiated with high hepatocyte nuclear factor 4A activity. We also developed and validated a robust predictor of consensus subtype with 100 genes and demonstrated that five subtypes were well conserved in patient-derived xenograft models and cell lines. By analyzing serum proteomic data from the same patients, we further identified potential serum biomarkers that can stratify patients into subtypes. CONCLUSIONS Five HCC subtypes are correlated with genomic phenotypes and clinical outcomes and highly conserved in preclinical models, providing a framework for selecting the most appropriate models for preclinical studies.
Collapse
Affiliation(s)
- Sung Hwan Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, Yonsei University College of Medicine, Korea
- Division of Hepatobiliary and Pancreas, Department of Surgery, CHA Bundang Medical Center, CHA University, Korea
| | - Sun Young Yim
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Yun Seong Jeong
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qi-Xiang Li
- Crown Bioscience, Inc., 3375 Scott Blvd, Suite 108, Santa Clara, CA, USA
| | - Sang-Hee Kang
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Surgery, Korea University Guro Hospital, Seoul, Korea
| | - Bo Hwa Sohn
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shwetha V. Kumar
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ji-Hyun Shin
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - You Rhee Choi
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jae-Jun Shim
- Department of Internal Medicine, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Hayeon Kim
- Department of Pathology, Korea University Guro Hospital, Seoul, Korea
| | - Jihoon Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Shin Kim
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Korea
| | - Sheng Guo
- Crown Bioscience (Suzhou), Inc., 218 Xinhu St, Suzhou, China
| | - Randy L. Johnson
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ahmed Kaseb
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koo Jeong Kang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Keimyung University Dongsan Medical Center, Daegu, Korea
| | - Yun Shin Chun
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hee Jin Jang
- Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Byoung Gill Lee
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hyun Goo Woo
- Department of Physiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Min Jin Ha
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rehan Akbani
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lewis R. Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - David A. Wheeler
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
32
|
Clark KL, George JW, Przygrodzka E, Plewes MR, Hua G, Wang C, Davis JS. Hippo Signaling in the Ovary: Emerging Roles in Development, Fertility, and Disease. Endocr Rev 2022; 43:1074-1096. [PMID: 35596657 PMCID: PMC9695108 DOI: 10.1210/endrev/bnac013] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Indexed: 01/09/2023]
Abstract
Emerging studies indicate that the Hippo pathway, a highly conserved pathway that regulates organ size control, plays an important role in governing ovarian physiology, fertility, and pathology. Specific to the ovary, the spatiotemporal expression of the major components of the Hippo signaling cascade are observed throughout the reproductive lifespan. Observations from multiple species begin to elucidate the functional diversity and molecular mechanisms of Hippo signaling in the ovary in addition to the identification of interactions with other signaling pathways and responses to various external stimuli. Hippo pathway components play important roles in follicle growth and activation, as well as steroidogenesis, by regulating several key biological processes through mechanisms of cell proliferation, migration, differentiation, and cell fate determination. Given the importance of these processes, dysregulation of the Hippo pathway contributes to loss of follicular homeostasis and reproductive disorders such as polycystic ovary syndrome (PCOS), premature ovarian insufficiency, and ovarian cancers. This review highlights what is currently known about the Hippo pathway core components in ovarian physiology, including ovarian development, follicle development, and oocyte maturation, while identifying areas for future research to better understand Hippo signaling as a multifunctional pathway in reproductive health and biology.
Collapse
Affiliation(s)
- Kendra L Clark
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Jitu W George
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Emilia Przygrodzka
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Michele R Plewes
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Guohua Hua
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science & Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Cheng Wang
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - John S Davis
- Olson Center for Women’s Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Veterans Affairs Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA
| |
Collapse
|
33
|
Sun Y, Hu L, Tao Z, Jarugumilli GK, Erb H, Singh A, Li Q, Cotton JL, Greninger P, Egan RK, Tony Ip Y, Benes CH, Che J, Mao J, Wu X. Pharmacological blockade of TEAD-YAP reveals its therapeutic limitation in cancer cells. Nat Commun 2022; 13:6744. [PMID: 36347861 PMCID: PMC9643419 DOI: 10.1038/s41467-022-34559-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/28/2022] [Indexed: 11/09/2022] Open
Abstract
Targeting TEAD autopalmitoylation has been proposed as a therapeutic approach for YAP-dependent cancers. Here we show that TEAD palmitoylation inhibitor MGH-CP1 and analogues block cancer cell "stemness", organ overgrowth and tumor initiation in vitro and in vivo. MGH-CP1 sensitivity correlates significantly with YAP-dependency in a large panel of cancer cell lines. However, TEAD inhibition or YAP/TAZ knockdown leads to transient inhibition of cell cycle progression without inducing cell death, undermining their potential therapeutic utilities. We further reveal that TEAD inhibition or YAP/TAZ silencing leads to VGLL3-mediated transcriptional activation of SOX4/PI3K/AKT signaling axis, which contributes to cancer cell survival and confers therapeutic resistance to TEAD inhibitors. Consistently, combination of TEAD and AKT inhibitors exhibits strong synergy in inducing cancer cell death. Our work characterizes the therapeutic opportunities and limitations of TEAD palmitoylation inhibitors in cancers, and uncovers an intrinsic molecular mechanism, which confers potential therapeutic resistance.
Collapse
Affiliation(s)
- Yang Sun
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, MA, USA.
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Lu Hu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, MA, USA
| | - Zhipeng Tao
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, MA, USA
| | - Gopala K Jarugumilli
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, MA, USA
| | - Hannah Erb
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, MA, USA
| | - Alka Singh
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, MA, USA
| | - Qi Li
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, MA, USA
| | - Jennifer L Cotton
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, MA, USA
| | - Patricia Greninger
- Massachusetts General Hospital Cancer Center, and Department of Medicine, Harvard Medical School, Charlestown, Massachusetts, MA, USA
| | - Regina K Egan
- Massachusetts General Hospital Cancer Center, and Department of Medicine, Harvard Medical School, Charlestown, Massachusetts, MA, USA
| | - Y Tony Ip
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, MA, USA
| | - Cyril H Benes
- Massachusetts General Hospital Cancer Center, and Department of Medicine, Harvard Medical School, Charlestown, Massachusetts, MA, USA
| | - Jianwei Che
- Department of Cancer Biology, Dana Farber Cancer Institute, and Harvard Medical School, Boston, Massachusetts, MA, USA
| | - Junhao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, MA, USA.
| | - Xu Wu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, MA, USA.
| |
Collapse
|
34
|
Targeting the Hippo Pathway in Gastric Cancer and Other Malignancies in the Digestive System: From Bench to Bedside. Biomedicines 2022; 10:biomedicines10102512. [PMID: 36289774 PMCID: PMC9599207 DOI: 10.3390/biomedicines10102512] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
The Hippo pathway is an evolutionally conserved signaling cascade that controls organ size and tissue regeneration under physiological conditions, and its aberrations have been well studied to promote tumor initiation and progression. Dysregulation of the Hippo tumor suppressor signaling frequently occurs in gastric cancer (GC) and other solid tumors and contributes to cancer development through modulating multiple aspects, including cell proliferation, survival, metastasis, and oncotherapy resistance. In the clinic, Hippo components also possess diagnostic and prognostic values for cancer patients. Considering its crucial role in driving tumorigenesis, targeting the Hippo pathway may greatly benefit developing novel cancer therapies. This review summarizes the current research progress regarding the core components and regulation of the Hippo pathway, as well as the mechanism and functional roles of their dysregulation in gastrointestinal malignancies, especially in GC, and discusses the therapeutic potential of targeting the Hippo pathway against cancers.
Collapse
|
35
|
Dekker Y, Le Dévédec SE, Danen EHJ, Liu Q. Crosstalk between Hypoxia and Extracellular Matrix in the Tumor Microenvironment in Breast Cancer. Genes (Basel) 2022; 13:genes13091585. [PMID: 36140753 PMCID: PMC9498429 DOI: 10.3390/genes13091585] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/28/2022] [Accepted: 08/31/2022] [Indexed: 11/24/2022] Open
Abstract
Even though breast cancer is the most diagnosed cancer among women, treatments are not always successful in preventing its progression. Recent studies suggest that hypoxia and the extracellular matrix (ECM) are important in altering cell metabolism and tumor metastasis. Therefore, the aim of this review is to study the crosstalk between hypoxia and the ECM and to assess their impact on breast cancer progression. The findings indicate that hypoxic signaling engages multiple mechanisms that directly contribute to ECM remodeling, ultimately increasing breast cancer aggressiveness. Second, hypoxia and the ECM cooperate to alter different aspects of cell metabolism. They mutually enhance aerobic glycolysis through upregulation of glucose transport, glycolytic enzymes, and by regulating intracellular pH. Both alter lipid and amino acid metabolism by stimulating lipid and amino acid uptake and synthesis, thereby providing the tumor with additional energy for growth and metastasis. Third, YAP/TAZ signaling is not merely regulated by the tumor microenvironment and cell metabolism, but it also regulates it primarily through its target c-Myc. Taken together, this review provides a better understanding of the crosstalk between hypoxia and the ECM in breast cancer. Additionally, it points to a role for the YAP/TAZ mechanotransduction pathway as an important link between hypoxia and the ECM in the tumor microenvironment, driving breast cancer progression.
Collapse
Affiliation(s)
- Yasmin Dekker
- Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Sylvia E. Le Dévédec
- Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| | - Erik H. J. Danen
- Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
- Correspondence: (E.H.J.D.); (Q.L.)
| | - Qiuyu Liu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100102, China
- Correspondence: (E.H.J.D.); (Q.L.)
| |
Collapse
|
36
|
Wang Y, Chen H, Yu J, Kang W, To KF. Recent insight into the role and therapeutic potential of YAP/TAZ in gastrointestinal cancers. Biochim Biophys Acta Rev Cancer 2022; 1877:188787. [PMID: 36041574 DOI: 10.1016/j.bbcan.2022.188787] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/25/2022] [Accepted: 08/23/2022] [Indexed: 11/18/2022]
Abstract
With the rapid development of cancer treatment, gastrointestinal (GI) cancers are still the most prevalent malignancies with high morbidity and mortality worldwide. Dysregulation of the Hippo signaling pathway has been recognized to play a critical role during cancer development and adopted for monitoring disease progression and therapy response. Despite the well-documented tumor proliferation and metastasis, recent efforts in two core Hippo components, Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), have identified as the driving forces behind cancer metabolism, stemness, tumor immunity, and therapy resistance. Understanding the molecular mechanisms by which YAP/TAZ facilitates the tumorigenesis and progression of GI cancer, and identifying novel therapeutic strategies for targeting YAP/TAZ are crucial to GI cancer treatment and prevention. In this study, we summarize the latest findings on the function and regulatory mechanisms of YAP/TAZ in GI cancers, and highlight the translational significance of targeting YAP/TAZ for cancer therapies.
Collapse
Affiliation(s)
- Yifei Wang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Centre, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Huarong Chen
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jun Yu
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Centre, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Centre, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
37
|
Mranda GM, Xiang ZP, Liu JJ, Wei T, Ding Y. Advances in prognostic and therapeutic targets for hepatocellular carcinoma and intrahepatic cholangiocarcinoma: The hippo signaling pathway. Front Oncol 2022; 12:937957. [PMID: 36033517 PMCID: PMC9411807 DOI: 10.3389/fonc.2022.937957] [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] [Received: 05/09/2022] [Accepted: 07/13/2022] [Indexed: 01/07/2023] Open
Abstract
Primary liver cancer is the sixth most frequently diagnosed cancer worldwide and the third leading cause of cancer-related death. The majority of the primary liver cancer cases are hepatocellular carcinoma and intrahepatic cholangiocarcinoma. Worldwide, there is an increasing incidence of primary liver cancer cases due to multiple risk factors ranging from parasites and viruses to metabolic diseases and lifestyles. Often, patients are diagnosed at advanced stages, depriving them of surgical curability benefits. Moreover, the efficacy of the available chemotherapeutics is limited in advanced stages. Furthermore, tumor metastases and recurrence make primary liver cancer management exceptionally challenging. Thus, exploring the molecular mechanisms for the development and progression of primary liver cancer is critical in improving diagnostic, treatment, prognostication, and surveillance modalities. These mechanisms facilitate the discovery of specific targets that are critical for novel and more efficient treatments. Consequently, the Hippo signaling pathway executing a pivotal role in organogenesis, hemostasis, and regeneration of tissues, regulates liver cells proliferation, and apoptosis. Cell polarity or adhesion molecules and cellular metabolic status are some of the biological activators of the pathway. Thus, understanding the mechanisms exhibited by the Hippo pathway is critical to the development of novel targeted therapies. This study reviews the advances in identifying therapeutic targets and prognostic markers of the Hippo pathway for primary liver cancer in the past six years.
Collapse
|
38
|
Ong YT, Andrade J, Armbruster M, Shi C, Castro M, Costa ASH, Sugino T, Eelen G, Zimmermann B, Wilhelm K, Lim J, Watanabe S, Guenther S, Schneider A, Zanconato F, Kaulich M, Pan D, Braun T, Gerhardt H, Efeyan A, Carmeliet P, Piccolo S, Grosso AR, Potente M. A YAP/TAZ-TEAD signalling module links endothelial nutrient acquisition to angiogenic growth. Nat Metab 2022; 4:672-682. [PMID: 35726026 PMCID: PMC9236904 DOI: 10.1038/s42255-022-00584-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/13/2022] [Indexed: 12/13/2022]
Abstract
Angiogenesis, the process by which endothelial cells (ECs) form new blood vessels from existing ones, is intimately linked to the tissue's metabolic milieu and often occurs at nutrient-deficient sites. However, ECs rely on sufficient metabolic resources to support growth and proliferation. How endothelial nutrient acquisition and usage are regulated is unknown. Here we show that these processes are instructed by Yes-associated protein 1 (YAP)/WW domain-containing transcription regulator 1 (WWTR1/TAZ)-transcriptional enhanced associate domain (TEAD): a transcriptional module whose function is highly responsive to changes in the tissue environment. ECs lacking YAP/TAZ or their transcriptional partners, TEAD1, 2 and 4 fail to divide, resulting in stunted vascular growth in mice. Conversely, activation of TAZ, the more abundant paralogue in ECs, boosts proliferation, leading to vascular hyperplasia. We find that YAP/TAZ promote angiogenesis by fuelling nutrient-dependent mTORC1 signalling. By orchestrating the transcription of a repertoire of cell-surface transporters, including the large neutral amino acid transporter SLC7A5, YAP/TAZ-TEAD stimulate the import of amino acids and other essential nutrients, thereby enabling mTORC1 activation. Dissociating mTORC1 from these nutrient inputs-elicited by the loss of Rag GTPases-inhibits mTORC1 activity and prevents YAP/TAZ-dependent vascular growth. Together, these findings define a pivotal role for YAP/TAZ-TEAD in controlling endothelial mTORC1 and illustrate the essentiality of coordinated nutrient fluxes in the vasculature.
Collapse
Affiliation(s)
- Yu Ting Ong
- Angiogenesis & Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Jorge Andrade
- Angiogenesis & Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Max Armbruster
- Angiogenesis & Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Chenyue Shi
- Angiogenesis & Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Marco Castro
- Angiogenesis & Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ana S H Costa
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Toshiya Sugino
- Angiogenesis & Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, and Department of Oncology and Leuven Cancer Institute, VIB and KU Leuven, Leuven, Belgium
| | - Barbara Zimmermann
- Angiogenesis & Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Kerstin Wilhelm
- Angiogenesis & Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Joseph Lim
- Angiogenesis & Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Shuichi Watanabe
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Guenther
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Andre Schneider
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Francesca Zanconato
- Department of Molecular Medicine, University of Padua School of Medicine, Padua, Italy
| | - Manuel Kaulich
- Institute of Biochemistry II, Goethe University, Frankfurt (Main), Germany
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Holger Gerhardt
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Vascular Patterning Laboratory, Center for Cancer Biology, VIB and KU Leuven, Leuven, Belgium
| | - Alejo Efeyan
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre, Madrid, Spain
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, and Department of Oncology and Leuven Cancer Institute, VIB and KU Leuven, Leuven, Belgium
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Laboratory of Angiogenesis and Vascular Heterogeneity, Department of Biomedicine, Aarhus, Denmark
| | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua School of Medicine, Padua, Italy
- IFOM-ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Ana Rita Grosso
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Michael Potente
- Angiogenesis & Metabolism Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- DZHK (German Center for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| |
Collapse
|
39
|
Paulusma CC, Lamers W, Broer S, van de Graaf SFJ. Amino acid metabolism, transport and signalling in the liver revisited. Biochem Pharmacol 2022; 201:115074. [PMID: 35568239 DOI: 10.1016/j.bcp.2022.115074] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 11/02/2022]
Abstract
The liver controls the systemic exposure of amino acids entering via the gastro-intestinal tract. For most amino acids except branched chain amino acids, hepatic uptake is very efficient. This implies that the liver orchestrates amino acid metabolism and also controls systemic amino acid exposure. Although many amino acid transporters have been identified, cloned and investigated with respect to substrate specificity, transport mechanism, and zonal distribution, which of these players are involved in hepatocellular amino acid transport remains unclear. Here, we aim to provide a review of current insight into the molecular machinery of hepatic amino acid transport. Furthermore, we place this information in a comprehensive overview of amino acid transport, signalling and metabolism.
Collapse
Affiliation(s)
- Coen C Paulusma
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands; Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Wouter Lamers
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands; Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, Netherlands; Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands
| | - Stefan Broer
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, Netherlands; Research School of Biology, Australian National University, Canberra, Australia
| | - Stan F J van de Graaf
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands; Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam, The Netherlands; Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
40
|
Russell JO, Camargo FD. Hippo signalling in the liver: role in development, regeneration and disease. Nat Rev Gastroenterol Hepatol 2022; 19:297-312. [PMID: 35064256 PMCID: PMC9199961 DOI: 10.1038/s41575-021-00571-w] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/14/2021] [Indexed: 02/07/2023]
Abstract
The Hippo signalling pathway has emerged as a major player in many aspects of liver biology, such as development, cell fate determination, homeostatic function and regeneration from injury. The regulation of Hippo signalling is complex, with activation of the pathway by diverse upstream inputs including signals from cellular adhesion, mechanotransduction and crosstalk with other signalling pathways. Pathological activation of the downstream transcriptional co-activators yes-associated protein 1 (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ, encoded by WWTR1), which are negatively regulated by Hippo signalling, has been implicated in multiple aspects of chronic liver disease, such as the development of liver fibrosis and tumorigenesis. Thus, development of pharmacological inhibitors of YAP-TAZ signalling has been an area of great interest. In this Review, we summarize the diverse roles of Hippo signalling in liver biology and highlight areas where outstanding questions remain to be investigated. Greater understanding of the mechanisms of Hippo signalling in liver function should help facilitate the development of novel therapies for the treatment of liver disease.
Collapse
Affiliation(s)
- Jacquelyn O Russell
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Fernando D Camargo
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
41
|
Role of Amino Acid Transporter SNAT1/SLC38A1 in Human Melanoma. Cancers (Basel) 2022; 14:cancers14092151. [PMID: 35565278 PMCID: PMC9099705 DOI: 10.3390/cancers14092151] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/20/2022] [Accepted: 04/24/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Malignant melanoma originates from melanocytes. Due to its high metastatic potential and its increasing incidence, it is one of the most aggressive types of cancer. Cancer cells generally exhibit an elevated metabolism, consequently adapting their expression of transport proteins to meet the increased demand of nutrients, such as amino acids. The aim of this study was to analyze the expression and function of the amino acid transporter SNAT1 in human melanoma. In addition, we wanted to determine its role in development and progression of malignant melanoma. We revealed that SNAT1 is overexpressed in melanoma tissue samples, as well as primary and metastatic cell lines. Moreover, we were able to show that SNAT1 plays an important role in forcing proliferation, colony formation, migration and invasion, and inhibiting senescence of melanoma cells. Amino acid transporters like SNAT1 are therefore promising targets for the development of novel therapeutic strategies against melanoma. Abstract The tumor metabolism is an important driver of cancer cell survival and growth, as rapidly dividing tumor cells exhibit a high demand for energetic sources and must adapt to microenvironmental changes. Therefore, metabolic reprogramming of cancer cells and the associated deregulation of nutrient transporters are a hallmark of cancer cells. Amino acids are essential for cancer cells to synthesize the necessary amount of protein, DNA, and RNA. Although cancer cells can synthesize glutamine de novo, most cancer cells show an increased uptake of glutamine from the tumor microenvironment. Especially SNAT1/SLC38A1, a member of the sodium neutral amino acid transporter (SNAT) family, plays an essential role during major net import of glutamine. In this study, we revealed a significant upregulation of SNAT1 expression in human melanoma tissue in comparison to healthy epidermis and an increased SNAT1 expression level in human melanoma cell lines when compared to normal human melanocytes (NHEMs). We demonstrated that functional inhibition of SNAT1 with α-(methylamino) isobutyric acid (MeAIB), as well as siRNA-mediated downregulation reduces cancer cell growth, cellular migration, invasion, and leads to induction of senescence in melanoma cells. Consequently, these results demonstrate that the amino acid transporter SNAT1 is essential for cancer growth, and indicates a potential target for cancer chemotherapy.
Collapse
|
42
|
Jeon HY, Choi J, Kraaier L, Kim YH, Eisenbarth D, Yi K, Kang JG, Kim JW, Shim HS, Lee JH, Lim DS. Airway secretory cell fate conversion via YAP-mTORC1-dependent essential amino acid metabolism. EMBO J 2022; 41:e109365. [PMID: 35285539 PMCID: PMC9016350 DOI: 10.15252/embj.2021109365] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 02/17/2022] [Accepted: 02/17/2022] [Indexed: 12/24/2022] Open
Abstract
Tissue homeostasis requires lineage fidelity of stem cells. Dysregulation of cell fate specification and differentiation leads to various diseases, yet the cellular and molecular mechanisms governing these processes remain elusive. We demonstrate that YAP/TAZ activation reprograms airway secretory cells, which subsequently lose their cellular identity and acquire squamous alveolar type 1 (AT1) fate in the lung. This cell fate conversion is mediated via distinctive transitional cell states of damage-associated transient progenitors (DATPs), recently shown to emerge during injury repair in mouse and human lungs. We further describe a YAP/TAZ signaling cascade to be integral for the fate conversion of secretory cells into AT1 fate, by modulating mTORC1/ATF4-mediated amino acid metabolism in vivo. Importantly, we observed aberrant activation of the YAP/TAZ-mTORC1-ATF4 axis in the altered airway epithelium of bronchiolitis obliterans syndrome, including substantial emergence of DATPs and AT1 cells with severe pulmonary fibrosis. Genetic and pharmacologic inhibition of mTORC1 activity suppresses lineage alteration and subepithelial fibrosis driven by YAP/TAZ activation, proposing a potential therapeutic target for human fibrotic lung diseases.
Collapse
Affiliation(s)
- Hae Yon Jeon
- Department of Biological Sciences, National Creative Research Center for Cell Plasticity, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Jinwook Choi
- Jeffrey Cheah Biomedical Centre, Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Lianne Kraaier
- Jeffrey Cheah Biomedical Centre, Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Young Hoon Kim
- Department of Biological Sciences, National Creative Research Center for Cell Plasticity, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - David Eisenbarth
- Department of Biological Sciences, National Creative Research Center for Cell Plasticity, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Kijong Yi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.,GenomeInsight Inc., Daejeon, South Korea
| | - Ju-Gyeong Kang
- Department of Biological Sciences, National Creative Research Center for Cell Plasticity, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Jin Woo Kim
- Department of Biological Sciences, National Creative Research Center for Cell Plasticity, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Hyo Sup Shim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Joo-Hyeon Lee
- Jeffrey Cheah Biomedical Centre, Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.,Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Dae-Sik Lim
- Department of Biological Sciences, National Creative Research Center for Cell Plasticity, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| |
Collapse
|
43
|
Du D, Liu C, Qin M, Zhang X, Xi T, Yuan S, Hao H, Xiong J. Metabolic dysregulation and emerging therapeutical targets for hepatocellular carcinoma. Acta Pharm Sin B 2022; 12:558-580. [PMID: 35256934 PMCID: PMC8897153 DOI: 10.1016/j.apsb.2021.09.019] [Citation(s) in RCA: 333] [Impact Index Per Article: 111.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is an aggressive human cancer with increasing incidence worldwide. Multiple efforts have been made to explore pharmaceutical therapies to treat HCC, such as targeted tyrosine kinase inhibitors, immune based therapies and combination of chemotherapy. However, limitations exist in current strategies including chemoresistance for instance. Tumor initiation and progression is driven by reprogramming of metabolism, in particular during HCC development. Recently, metabolic associated fatty liver disease (MAFLD), a reappraisal of new nomenclature for non-alcoholic fatty liver disease (NAFLD), indicates growing appreciation of metabolism in the pathogenesis of liver disease, including HCC, thereby suggesting new strategies by targeting abnormal metabolism for HCC treatment. In this review, we introduce directions by highlighting the metabolic targets in glucose, fatty acid, amino acid and glutamine metabolism, which are suitable for HCC pharmaceutical intervention. We also summarize and discuss current pharmaceutical agents and studies targeting deregulated metabolism during HCC treatment. Furthermore, opportunities and challenges in the discovery and development of HCC therapy targeting metabolism are discussed.
Collapse
Key Words
- 1,3-BPG, 1,3-bisphosphoglycerate
- 2-DG, 2-deoxy-d-glucose
- 3-BrPA, 3-bromopyruvic acid
- ACC, acetyl-CoA carboxylase
- ACLY, adenosine triphosphate (ATP) citrate lyase
- ACS, acyl-CoA synthease
- AKT, protein kinase B
- AML, acute myeloblastic leukemia
- AMPK, adenosine mono-phosphate-activated protein kinase
- ASS1, argininosuccinate synthase 1
- ATGL, adipose triacylglycerol lipase
- CANA, canagliflozin
- CPT, carnitine palmitoyl-transferase
- CYP4, cytochrome P450s (CYPs) 4 family
- Cancer therapy
- DNL, de novo lipogenesis
- EMT, epithelial-to-mesenchymal transition
- ER, endoplasmic reticulum
- ERK, extracellular-signal regulated kinase
- FABP1, fatty acid binding protein 1
- FASN, fatty acid synthase
- FBP1, fructose-1,6-bisphosphatase 1
- FFA, free fatty acid
- Fatty acid β-oxidation
- G6PD, glucose-6-phosphate dehydrogenase
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- GLS1, renal-type glutaminase
- GLS2, liver-type glutaminase
- GLUT1, glucose transporter 1
- GOT1, glutamate oxaloacetate transaminase 1
- Glutamine metabolism
- Glycolysis
- HCC, hepatocellular carcinoma
- HIF-1α, hypoxia-inducible factor-1 alpha
- HK, hexokinase
- HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase
- HSCs, hepatic stellate cells
- Hepatocellular carcinoma
- IDH2, isocitrate dehydrogenase 2
- LCAD, long-chain acyl-CoA dehydrogenase
- LDH, lactate dehydrogenase
- LPL, lipid lipase
- LXR, liver X receptor
- MAFLD, metabolic associated fatty liver disease
- MAGL, monoacyglycerol lipase
- MCAD, medium-chain acyl-CoA dehydrogenase
- MEs, malic enzymes
- MMP9, matrix metallopeptidase 9
- Metabolic dysregulation
- NADPH, nicotinamide adenine nucleotide phosphate
- NAFLD, non-alcoholic fatty liver disease
- NASH, non-alcoholic steatohepatitis
- OTC, ornithine transcarbamylase
- PCK1, phosphoenolpyruvate carboxykinase 1
- PFK1, phosphofructokinase 1
- PGAM1, phosphoglycerate mutase 1
- PGK1, phosphoglycerate kinase 1
- PI3K, phosphoinositide 3-kinase
- PKM2, pyruvate kinase M2
- PPARα, peroxisome proliferator-activated receptor alpha
- PPP, pentose phosphate pathway
- Pentose phosphate pathway
- ROS, reactive oxygen species
- SCD1, stearoyl-CoA-desaturase 1
- SGLT2, sodium-glucose cotransporter 2
- SLC1A5/ASCT2, solute carrier family 1 member 5/alanine serine cysteine preferring transporter 2
- SLC7A5/LAT1, solute carrier family 7 member 5/L-type amino acid transporter 1
- SREBP1, sterol regulatory element-binding protein 1
- TAGs, triacylglycerols
- TCA cycle, tricarboxylic acid cycle
- TKIs, tyrosine kinase inhibitors
- TKT, transketolase
- Tricarboxylic acid cycle
- VEGFR, vascular endothelial growth factor receptor
- WD-fed MC4R-KO, Western diet (WD)-fed melanocortin 4 receptor-deficient (MC4R-KO)
- WNT, wingless-type MMTV integration site family
- mIDH, mutant IDH
- mTOR, mammalian target of rapamycin
Collapse
Affiliation(s)
- Danyu Du
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Chan Liu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Mengyao Qin
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao Zhang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tao Xi
- Research Center of Biotechnology, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Shengtao Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Haiping Hao
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
- Corresponding authors.
| | - Jing Xiong
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Corresponding authors.
| |
Collapse
|
44
|
Lin Z, Ji X, Tian N, Gan Y, Ke L. Mapping Intellectual Structure for the Long Non-Coding RNA in Hepatocellular Carcinoma Development Research. Front Genet 2022; 12:771810. [PMID: 35047004 PMCID: PMC8762053 DOI: 10.3389/fgene.2021.771810] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/22/2021] [Indexed: 01/09/2023] Open
Abstract
Background: Emerging research suggests that long non-coding RNAs (lncRNAs) play an important role in a variety of developmental or physiological processes of hepatocellular carcinoma (HCC). Various differentially expressed lncRNAs have been identified in HCC. Thus, a deeper analysis of recent research concerning lncRNA and HCC development could provide scientists with a valuable reference for future studies. Methods: Related publications were retrieved from the Web of Science Core Collection database. CiteSpace version 5.6.R4 was employed to conduct bibliometric analysis. Several network maps were constructed to evaluate the collaborations between different countries, institutions, authors, journals, and keywords. Results: A total of 2,667 records were initially found from the year of 2010–2020. The annual related publications output had increased dramatically during these years. Although China was the most prolific country in terms of research publication, the United States played a leading role in collaborative network. The Nanjing Medical University was the most productive institute in the field of lncRNAs in HCC development. Gang Chen was the most prolific researcher, while Yang F was the most frequently co-cited author. Oncotarget, Cell, and Oncogene were the most highly co-cited journals. The most recent burst keywords were interaction, database, and pathway. Conclusion: This study provides a comprehensive overview for the field of lncRNAs in HCC development based on bibliometric and visualized methods. The results would provide a reference for scholars focusing on this field.
Collapse
Affiliation(s)
- Zhifeng Lin
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Medical Record, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaohui Ji
- Department of Obstetrics and Gynaecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Nana Tian
- Department of Medical Record, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yu Gan
- Department of Medical Record, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Li Ke
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Department of Medical Record, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
45
|
Li Y, Qiu X, Wang X, Liu H, Geck RC, Tewari AK, Xiao T, Font-Tello A, Lim K, Jones KL, Morrow M, Vadhi R, Kao PL, Jaber A, Yerrum S, Xie Y, Chow KH, Cejas P, Nguyen QD, Long HW, Liu XS, Toker A, Brown M. FGFR-inhibitor-mediated dismissal of SWI/SNF complexes from YAP-dependent enhancers induces adaptive therapeutic resistance. Nat Cell Biol 2021; 23:1187-1198. [PMID: 34737445 DOI: 10.1038/s41556-021-00781-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 09/26/2021] [Indexed: 12/20/2022]
Abstract
How cancer cells adapt to evade the therapeutic effects of drugs targeting oncogenic drivers is poorly understood. Here we report an epigenetic mechanism leading to the adaptive resistance of triple-negative breast cancer (TNBC) to fibroblast growth factor receptor (FGFR) inhibitors. Prolonged FGFR inhibition suppresses the function of BRG1-dependent chromatin remodelling, leading to an epigenetic state that derepresses YAP-associated enhancers. These chromatin changes induce the expression of several amino acid transporters, resulting in increased intracellular levels of specific amino acids that reactivate mTORC1. Consistent with this mechanism, addition of mTORC1 or YAP inhibitors to FGFR blockade synergistically attenuated the growth of TNBC patient-derived xenograft models. Collectively, these findings reveal a feedback loop involving an epigenetic state transition and metabolic reprogramming that leads to adaptive therapeutic resistance and provides potential therapeutic strategies to overcome this mechanism of resistance.
Collapse
Affiliation(s)
- Yihao Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Xintao Qiu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Xiaoqing Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hui Liu
- Department of Pathology, and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Renee C Geck
- Department of Pathology, and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alok K Tewari
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Tengfei Xiao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alba Font-Tello
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Klothilda Lim
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kristen L Jones
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, Boston, MA, USA
| | - Murry Morrow
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, Boston, MA, USA
| | - Raga Vadhi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Pei-Lun Kao
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Center for Patient Derived Models, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Aliya Jaber
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Center for Patient Derived Models, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Smitha Yerrum
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Center for Patient Derived Models, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yingtian Xie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kin-Hoe Chow
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Center for Patient Derived Models, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Paloma Cejas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Quang-Dé Nguyen
- Lurie Family Imaging Center, Center for Biomedical Imaging in Oncology, Dana-Farber Cancer Institute, Boston, Boston, MA, USA
| | - Henry W Long
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - X Shirley Liu
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Data Science, Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alex Toker
- Department of Pathology, and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
| | - Myles Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA. .,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA. .,Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
46
|
Saba JA, Liakath-Ali K, Green R, Watt FM. Translational control of stem cell function. Nat Rev Mol Cell Biol 2021; 22:671-690. [PMID: 34272502 DOI: 10.1038/s41580-021-00386-2] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2021] [Indexed: 12/22/2022]
Abstract
Stem cells are characterized by their ability to self-renew and differentiate into many different cell types. Research has focused primarily on how these processes are regulated at a transcriptional level. However, recent studies have indicated that stem cell behaviour is strongly coupled to the regulation of protein synthesis by the ribosome. In this Review, we discuss how different translation mechanisms control the function of adult and embryonic stem cells. Stem cells are characterized by low global translation rates despite high levels of ribosome biogenesis. The maintenance of pluripotency, the commitment to a specific cell fate and the switch to cell differentiation depend on the tight regulation of protein synthesis and ribosome biogenesis. Translation regulatory mechanisms that impact on stem cell function include mTOR signalling, ribosome levels, and mRNA and tRNA features and amounts. Understanding these mechanisms important for stem cell self-renewal and differentiation may also guide our understanding of cancer grade and metastasis.
Collapse
Affiliation(s)
- James A Saba
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kifayathullah Liakath-Ali
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Rachel Green
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Fiona M Watt
- King's College London Centre for Stem Cells and Regenerative Medicine, Guy's Hospital, London, UK.
| |
Collapse
|
47
|
Zavorka Thomas ME, Lu X, Talebi Z, Jeon JY, Buelow DR, Gibson AA, Uddin ME, Brinton LT, Nguyen J, Collins M, Lodi A, Sweeney SR, Campbell MJ, Sweet DH, Sparreboom A, Lapalombella R, Tiziani S, Baker SD. Gilteritinib Inhibits Glutamine Uptake and Utilization in FLT3-ITD-Positive AML. Mol Cancer Ther 2021; 20:2207-2217. [PMID: 34518298 DOI: 10.1158/1535-7163.mct-21-0071] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/17/2021] [Accepted: 09/01/2021] [Indexed: 11/16/2022]
Abstract
Acute myeloid leukemia (AML) with an FLT3 internal tandem duplication (FLT3-ITD) mutation is an aggressive hematologic malignancy associated with frequent relapse and poor overall survival. The tyrosine kinase inhibitor gilteritinib is approved for the treatment of relapse/refractory AML with FLT3 mutations, yet its mechanism of action is not completely understood. Here, we sought to identify additional therapeutic targets that can be exploited to enhance gilteritinib's antileukemic effect. Based on unbiased transcriptomic analyses, we identified the glutamine transporter SNAT1 (SLC38A1) as a novel target of gilteritinib that leads to impaired glutamine uptake and utilization within leukemic cells. Using metabolomics and metabolic flux analyses, we found that gilteritinib decreased glutamine metabolism through the TCA cycle and cellular levels of the oncometabolite 2-hydroxyglutarate. In addition, gilteritinib treatment was associated with decreased ATP production and glutathione synthesis and increased reactive oxygen species, resulting in cellular senescence. Finally, we found that the glutaminase inhibitor CB-839 enhanced antileukemic effect of gilteritinib in ex vivo studies using human primary FLT3-ITD-positive AML cells harboring mutations in the enzyme isocitrate dehydrogenase, which catalyzes the oxidative decarboxylation of isocitrate, producing α-ketoglutarate. Collectively, this work has identified a previously unrecognized, gilteritinib-sensitive metabolic pathway downstream of SLC38A1 that causes decreased glutaminolysis and disruption of redox homeostasis. These findings provide a rationale for the development and therapeutic exploration of targeted combinatorial treatment strategies for this subset of relapse/refractory AML.
Collapse
Affiliation(s)
- Megan E Zavorka Thomas
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Xiyuan Lu
- Department of Nutritional Sciences and Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Zahra Talebi
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Jae Yoon Jeon
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Daelynn R Buelow
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Alice A Gibson
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Muhammad Erfan Uddin
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Lindsey T Brinton
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Julie Nguyen
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, Virginia
| | - Meghan Collins
- Department of Nutritional Sciences and Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Alessia Lodi
- Department of Nutritional Sciences and Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Shannon R Sweeney
- Department of Nutritional Sciences and Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Moray J Campbell
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Douglas H Sweet
- Department of Pharmaceutics, Virginia Commonwealth University, Richmond, Virginia
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Rosa Lapalombella
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio.,Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Stefano Tiziani
- Department of Nutritional Sciences and Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Sharyn D Baker
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
48
|
Pearson JD, Huang K, Pacal M, McCurdy SR, Lu S, Aubry A, Yu T, Wadosky KM, Zhang L, Wang T, Gregorieff A, Ahmad M, Dimaras H, Langille E, Cole SPC, Monnier PP, Lok BH, Tsao MS, Akeno N, Schramek D, Wikenheiser-Brokamp KA, Knudsen ES, Witkiewicz AK, Wrana JL, Goodrich DW, Bremner R. Binary pan-cancer classes with distinct vulnerabilities defined by pro- or anti-cancer YAP/TEAD activity. Cancer Cell 2021; 39:1115-1134.e12. [PMID: 34270926 PMCID: PMC8981970 DOI: 10.1016/j.ccell.2021.06.016] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/17/2020] [Accepted: 06/24/2021] [Indexed: 12/13/2022]
Abstract
Cancer heterogeneity impacts therapeutic response, driving efforts to discover over-arching rules that supersede variability. Here, we define pan-cancer binary classes based on distinct expression of YAP and YAP-responsive adhesion regulators. Combining informatics with in vivo and in vitro gain- and loss-of-function studies across multiple murine and human tumor types, we show that opposite pro- or anti-cancer YAP activity functionally defines binary YAPon or YAPoff cancer classes that express or silence YAP, respectively. YAPoff solid cancers are neural/neuroendocrine and frequently RB1-/-, such as retinoblastoma, small cell lung cancer, and neuroendocrine prostate cancer. YAP silencing is intrinsic to the cell of origin, or acquired with lineage switching and drug resistance. The binary cancer groups exhibit distinct YAP-dependent adhesive behavior and pharmaceutical vulnerabilities, underscoring clinical relevance. Mechanistically, distinct YAP/TEAD enhancers in YAPoff or YAPon cancers deploy anti-cancer integrin or pro-cancer proliferative programs, respectively. YAP is thus pivotal across cancer, but in opposite ways, with therapeutic implications.
Collapse
Affiliation(s)
- Joel D Pearson
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada; Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON M5T 3A9, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Katherine Huang
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Marek Pacal
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Sean R McCurdy
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Suying Lu
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Arthur Aubry
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada; Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON M5T 3A9, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tao Yu
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Kristine M Wadosky
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Letian Zhang
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Tao Wang
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Alex Gregorieff
- Department of Pathology, McGill University and Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, ON H4A 3J1, Canada
| | - Mohammad Ahmad
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Helen Dimaras
- Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON M5T 3A9, Canada; The Department of Ophthalmology & Vision Sciences, Child Health Evaluative Sciences Program, and Center for Global Child Health, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Division of Clinical Public Health, Dalla Lana School of Public Health, The University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Ellen Langille
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Susan P C Cole
- Division of Cancer Biology and Genetics, Queen's University Cancer Research Institute, Kingston, ON K7L 3N6, Canada
| | - Philippe P Monnier
- Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON M5T 3A9, Canada; Krembil Research Institute, Vision Division, Krembil Discovery Tower, Toronto, ON M5T 2S8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Benjamin H Lok
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; Department of Radiation Oncology, University of Toronto, Toronto, ON M5T 1P5, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Ming-Sound Tsao
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Nagako Akeno
- Division of Pathology & Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Daniel Schramek
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Kathryn A Wikenheiser-Brokamp
- Division of Pathology & Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; The Perinatal Institute Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pathology & Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Erik S Knudsen
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Agnieszka K Witkiewicz
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Jeffrey L Wrana
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - David W Goodrich
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Rod Bremner
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health System, Toronto, ON M5G 1X5, Canada; Department of Ophthalmology and Vision Science, University of Toronto, Toronto, ON M5T 3A9, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
49
|
Nguyen-Lefebvre AT, Selzner N, Wrana JL, Bhat M. The hippo pathway: A master regulator of liver metabolism, regeneration, and disease. FASEB J 2021; 35:e21570. [PMID: 33831275 DOI: 10.1096/fj.202002284rr] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/04/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022]
Abstract
The liver is the only visceral organ in the body with a tremendous capacity to regenerate in response to insults that induce inflammation, cell death, and injury. Liver regeneration is a complicated process involving a well-orchestrated activation of non-parenchymal cells in the injured area and proliferation of undamaged hepatocytes. Furthermore, the liver has a Hepatostat, defined as adjustment of its volume to that required for homeostasis. Understanding the mechanisms that control different steps of liver regeneration is critical to informing therapies for liver repair, to help patients with liver disease. The Hippo signaling pathway is well known for playing an essential role in the control and regulation of liver size, regeneration, stem cell self-renewal, and liver cancer. Thus, the Hippo pathway regulates dynamic cell fates in liver, and in absence of its downstream effectors YAP and TAZ, liver regeneration is severely impaired, and the proliferative expansion of liver cells blocked. We will mainly review upstream mechanisms activating the Hippo signaling pathway following partial hepatectomy in mouse model and patients, its roles during different steps of liver regeneration, metabolism, and cancer. We will also discuss how targeting the Hippo signaling cascade might improve liver regeneration and suppress liver tumorigenesis.
Collapse
Affiliation(s)
- Anh Thu Nguyen-Lefebvre
- Department of Medicine, Multi-Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada.,Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
| | - Nazia Selzner
- Department of Medicine, Multi-Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada
| | | | - Mamatha Bhat
- Department of Medicine, Multi-Organ Transplant Program, Toronto General Hospital, Toronto, ON, Canada
| |
Collapse
|
50
|
Ma J, Huang X. Research progress in role of Hippo signaling pathway in diagnosis and treatment for hepatocellular carcinoma. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2021; 46:637-643. [PMID: 34275933 PMCID: PMC10930194 DOI: 10.11817/j.issn.1672-7347.2021.200243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Indexed: 11/03/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumor worldwide, with high incidence and mortality. However, the exact mechanisms leading to HCC development remain unclear. The cores of the Hippo signaling pathway consist of a kinase cascade to transmit signals, which inhibits the transcriptional coactivator translocate into the nucleus and reduces the transcription of downstream proliferation-related genes. Hippo signaling pathway regulates liver development and regeneration after liver resection, and it is also related to the occurrence of HCC. The Hippo pathway regulates proliferation, apoptosis, metastasis, autophagy, metabolic reprogramming of HCC cells, affects the tumor immune microenvironment, and participates multiple-drug resistance. Further study on the role of Hippo signaling pathway in HCC is important to develop new therapeutic targets.
Collapse
Affiliation(s)
- Jiamei Ma
- Department of Gastroenterology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University, Haikou 570208, China.
| | - Xiaoxi Huang
- Department of Gastroenterology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University, Haikou 570208, China.
| |
Collapse
|