1
|
Wang J, Yang R, Wang F, Zhang J, Dong Y, Wang J, Yu M, Xu Y, Liu L, Cheng Y, Zhang C, Yang Y, Yang W, Wang J, Chen G, Huang Y, Tian Y, Jian R, Ni B, Wu W, Ruan Y. CRISPR-Cas9 screening identifies the role of FER as a tumor suppressor. J Pathol 2024. [PMID: 39648412 DOI: 10.1002/path.6374] [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: 06/06/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 12/10/2024]
Abstract
It is important to systematically identify tumor suppressor genes (TSGs) to improve our understanding of tumorigenesis and develop strategies for early diagnosis and mitigating disease progression. In the present study, we used an in vivo genome-wide clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (Cas9) screen and identified FPS/FES-related (FER) as a TSG. Single-cell RNA sequencing (scRNA-seq) revealed that normal cells with low FER expression exhibited elevated malignant transformation potential and stemness properties. FER knockout promoted the tumorigenic transformation, characterized by high colony-forming efficiency and suspension growth ability, acquired tumorigenicity in vivo, increased metabolic activity, dedifferentiation properties, and immune evasion. Moreover, analysis revealed that low FER expression tumors share molecular phenotypes with FER knockout cells, suggesting the consistent role of FER in tumor initiation and progression. Taken together, our findings not only provide insights into the essential role of FER as a tumor suppressor in tumor initiation and progression but also highlight its potential as a target for future clinical diagnosis. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jiaqi Wang
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
| | - Ran Yang
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
- Department of Pathophysiology, College of High Altitude Military Medicine, Chongqing, PR China
| | - Fengsheng Wang
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
- State Key Laboratory of NBC Protection for Civilian, Beijing, PR China
| | - Junlei Zhang
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
| | - Yutong Dong
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
- Army Health Service Training Base, Chongqing, PR China
| | - Jiangjun Wang
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
- Clinical Laboratory and Department of Pathology, The 72nd Army Hospital of the People's Liberation Army, Zhejiang, PR China
| | - Meng Yu
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
- 927th Hospital of Joint Logistics Support Force, Yunnan, PR China
| | - Yixiao Xu
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
- The 83rd Affiliated Hospital of Xinxiang Medical University, Xinxiang, PR China
| | - Lianlian Liu
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
| | - Yuda Cheng
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
| | - Chen Zhang
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
| | - Yi Yang
- Army Medical University, Chongqing, PR China
- Experimental Center of Basic Medicine, College of Basic Medical Sciences, Chongqing, PR China
| | - Wubin Yang
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
- Department of Pathophysiology, College of High Altitude Military Medicine, Chongqing, PR China
| | - Jiali Wang
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
| | - Guangxing Chen
- Army Medical University, Chongqing, PR China
- Department of Joint Surgery, The First Affiliated Hospital, Chongqing, PR China
| | - Yi Huang
- Army Medical University, Chongqing, PR China
- Biomedical Analysis Center, Chongqing, PR China
| | - Yanping Tian
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
| | - Rui Jian
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
| | - Bing Ni
- Army Medical University, Chongqing, PR China
- Department of Pathophysiology, College of High Altitude Military Medicine, Chongqing, PR China
| | - Wei Wu
- Army Medical University, Chongqing, PR China
- Thoracic Surgery Department, Southwest Hospital, The First Affiliated Hospital, Chongqing, PR China
| | - Yan Ruan
- Army Medical University, Chongqing, PR China
- Laboratory of Stem Cell & Developmental Biology, Department of Histology and Embryology, College of Basic Medical Sciences, Chongqing, PR China
| |
Collapse
|
2
|
Tsakiroglou M, Evans A, Doce-Carracedo A, Little M, Hornby R, Roberts P, Zhang E, Miyajima F, Pirmohamed M. Gene Expression Dysregulation in Whole Blood of Patients with Clostridioides difficile Infection. Int J Mol Sci 2024; 25:12653. [PMID: 39684365 DOI: 10.3390/ijms252312653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Clostridioides difficile (C. difficile) is a global threat and has significant implications for individuals and health care systems. Little is known about host molecular mechanisms and transcriptional changes in peripheral immune cells. This is the first gene expression study in whole blood from patients with C. difficile infection. We took blood and stool samples from patients with toxigenic C. difficile infection (CDI), non-toxigenic C. difficile infection (GDH), inflammatory bowel disease (IBD), diarrhea from other causes (DC), and healthy controls (HC). We performed transcriptome-wide RNA profiling on peripheral blood to identify diarrhea common and CDI unique gene sets. Diarrhea groups upregulated innate immune responses with neutrophils at the epicenter. The common signature associated with diarrhea was non-specific and shared by various other inflammatory conditions. CDI had a unique 45 gene set reflecting the downregulation of humoral and T cell memory functions. Dysregulation of immunometabolic genes was also abundant and linked to immune cell fate during differentiation. Whole transcriptome analysis of white cells in blood from patients with toxigenic C. difficile infection showed that there is an impairment of adaptive immunity and immunometabolism.
Collapse
Affiliation(s)
- Maria Tsakiroglou
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Anthony Evans
- Computational Biology Facility, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7BE, UK
| | - Alejandra Doce-Carracedo
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
- Clinical Directorate, GCP Laboratories, University of Liverpool, Liverpool L7 8TX, UK
| | - Margaret Little
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Rachel Hornby
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Paul Roberts
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
- Faculty of Science and Engineering, School of Biomedical Science and Physiology, University of Wolverhampton, Wolverhampton WV1 1LZ, UK
| | - Eunice Zhang
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Fabio Miyajima
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
- Oswaldo Cruz Foundation (Fiocruz), Branch Ceara, Eusebio 61773-270, Brazil
| | - Munir Pirmohamed
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| |
Collapse
|
3
|
Jin X, Wang S, Luo L, Yan F, He Q. Targeting the Wnt/β-catenin signal pathway for the treatment of gastrointestinal cancer: Potential for advancement. Biochem Pharmacol 2024; 227:116463. [PMID: 39102994 DOI: 10.1016/j.bcp.2024.116463] [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: 05/07/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
Gastrointestinal cancers (GICs) are highly prevalent cancers that threaten human health worldwide. The Wnt/β-catenin signaling pathway has been reported to play a pivotal role in the carcinogenesis of GICs. Numerous interventions targeting the Wnt/β-catenin signaling in GICs are currently being tested in clinical trials with promising results. Unfortunately, there are no clinically approved drugs that effectively target this pathway. This comprehensive review aims to evaluate the impact of clinical therapies targeting the Wnt/β-catenin signaling pathway in GICs. By integrating data from bioinformatics databases and recent literature from the past five years, we examine the heterogeneous expression and regulatory mechanisms of Wnt/β-catenin pathway genes and proteins in GICs. Specifically, we focus on expression patterns, mutation frequencies, and clinical prognoses to understand their implications for treatment strategies. Additionally, we discuss recent clinical trial efforts targeting this pathway. Understanding the inhibitors currently under clinical investigation may help optimize foundational research and clinical strategies. We hope that elucidating the current status of precision therapeutic stratification for patients targeting the Wnt/β-catenin pathway will guide future innovations in precision medicine for GICs.
Collapse
Affiliation(s)
- Xizhi Jin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, Zhejiang 310018, PR China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| | - Fangjie Yan
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou, Zhejiang 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, Zhejiang 310018, PR China.
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Zhejiang University, Hangzhou, Zhejiang 310058, PR China.
| |
Collapse
|
4
|
Festa M, Coppola MA, Angeli E, Tettey-Matey A, Giusto A, Mazza I, Gatta E, Barbieri R, Picollo A, Gavazzo P, Pusch M, Picco C, Sbrana F. TMEM9B Regulates Endosomal ClC-3 and ClC-4 Transporters. Life (Basel) 2024; 14:1034. [PMID: 39202776 PMCID: PMC11355779 DOI: 10.3390/life14081034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 09/03/2024] Open
Abstract
The nine-member CLC gene family of Cl- chloride-transporting membrane proteins is divided into plasma membrane-localized Cl- channels and endo-/lysosomal Cl-/H+ antiporters. Accessory proteins have been identified for ClC-K and ClC-2 channels and for the lysosomal ClC-7, but not the other CLCs. Here, we identified TMEM9 Domain Family Member B (TMEM9B), a single-span type I transmembrane protein of unknown function, to strongly interact with the neuronal endosomal ClC-3 and ClC-4 transporters. Co-expression of TMEM9B with ClC-3 or ClC-4 dramatically reduced transporter activity in Xenopus oocytes and transfected HEK cells. For ClC-3, TMEM9B also induced a slow component in the kinetics of the activation time course, suggesting direct interaction. Currents mediated by ClC-7 were hardly affected by TMEM9B, and ClC-1 currents were only slightly reduced, demonstrating specific interaction with ClC-3 and ClC-4. We obtained strong evidence for direct interaction by detecting significant Förster Resonance Energy Transfer (FRET), exploiting fluorescence lifetime microscopy-based (FLIM-FRET) techniques between TMEM9B and ClC-3 and ClC-4, but hardly any FRET with ClC-1 or ClC-7. The discovery of TMEM9B as a novel interaction partner of ClC-3 and ClC-4 might have important implications for the physiological role of these transporters in neuronal endosomal homeostasis and for a better understanding of the pathological mechanisms in CLCN3- and CLCN4-related pathological conditions.
Collapse
Affiliation(s)
- Margherita Festa
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, 16149 Genova, Italy; (M.F.); (M.A.C.); (A.T.-M.); (A.G.); (I.M.); (R.B.); (A.P.); (P.G.)
| | - Maria Antonietta Coppola
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, 16149 Genova, Italy; (M.F.); (M.A.C.); (A.T.-M.); (A.G.); (I.M.); (R.B.); (A.P.); (P.G.)
| | - Elena Angeli
- DIFI Lab, Dipartimento di Fisica, Università di Genova, 16146 Genova, Italy; (E.A.); (E.G.)
| | - Abraham Tettey-Matey
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, 16149 Genova, Italy; (M.F.); (M.A.C.); (A.T.-M.); (A.G.); (I.M.); (R.B.); (A.P.); (P.G.)
| | - Alice Giusto
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, 16149 Genova, Italy; (M.F.); (M.A.C.); (A.T.-M.); (A.G.); (I.M.); (R.B.); (A.P.); (P.G.)
| | - Irene Mazza
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, 16149 Genova, Italy; (M.F.); (M.A.C.); (A.T.-M.); (A.G.); (I.M.); (R.B.); (A.P.); (P.G.)
| | - Elena Gatta
- DIFI Lab, Dipartimento di Fisica, Università di Genova, 16146 Genova, Italy; (E.A.); (E.G.)
| | - Raffaella Barbieri
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, 16149 Genova, Italy; (M.F.); (M.A.C.); (A.T.-M.); (A.G.); (I.M.); (R.B.); (A.P.); (P.G.)
| | - Alessandra Picollo
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, 16149 Genova, Italy; (M.F.); (M.A.C.); (A.T.-M.); (A.G.); (I.M.); (R.B.); (A.P.); (P.G.)
| | - Paola Gavazzo
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, 16149 Genova, Italy; (M.F.); (M.A.C.); (A.T.-M.); (A.G.); (I.M.); (R.B.); (A.P.); (P.G.)
| | - Michael Pusch
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, 16149 Genova, Italy; (M.F.); (M.A.C.); (A.T.-M.); (A.G.); (I.M.); (R.B.); (A.P.); (P.G.)
| | - Cristiana Picco
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, 16149 Genova, Italy; (M.F.); (M.A.C.); (A.T.-M.); (A.G.); (I.M.); (R.B.); (A.P.); (P.G.)
| | - Francesca Sbrana
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, 16149 Genova, Italy; (M.F.); (M.A.C.); (A.T.-M.); (A.G.); (I.M.); (R.B.); (A.P.); (P.G.)
| |
Collapse
|
5
|
Baek S, Chang JW, Yoo SM, Choo J, Jung S, Nah J, Jung YK. TMEM9 activates Rab9-dependent alternative autophagy through interaction with Beclin1. Cell Mol Life Sci 2024; 81:322. [PMID: 39078420 PMCID: PMC11335249 DOI: 10.1007/s00018-024-05366-1] [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: 02/28/2024] [Revised: 06/06/2024] [Accepted: 06/30/2024] [Indexed: 07/31/2024]
Abstract
Transmembrane protein 9 (TMEM9) is a transmembrane protein that regulates lysosomal acidification by interacting with the v-type ATPase complex. However, the role of TMEM9 in the lysosome-dependent autophagy machinery has yet to be identified. In this study, we demonstrate that the lysosomal protein TMEM9, which is involved in vesicle acidification, regulates Rab9-dependent alternative autophagy through its interaction with Beclin1. The cytosolic domain of TMEM9 interacts with Beclin1 via its Bcl-2-binding domain. This interaction between TMEM9 and Beclin1 dissociates Bcl-2, an autophagy-inhibiting partner, from Beclin1, thereby activating LC3-independent and Rab9-dependent alternative autophagy. Late endosomal and lysosomal TMEM9 apparently colocalizes with Rab9 but not with LC3. Furthermore, we show that multiple glycosylation of TMEM9, essential for lysosomal localization, is essential for its interaction with Beclin1 and the activation of Rab9-dependent alternative autophagy. These findings reveal that TMEM9 recruits and activates the Beclin1 complex at the site of Rab9-dependent autophagosome to induce alternative autophagy.
Collapse
Affiliation(s)
- Sohyeon Baek
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, 28644, South Korea
| | - Jae-Woong Chang
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Seung-Min Yoo
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - JeongRim Choo
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea
| | - Sunmin Jung
- Department of Biochemistry, Chungbuk National University, Cheongju, 28644, South Korea
| | - Jihoon Nah
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, 28644, South Korea.
- Department of Biochemistry, Chungbuk National University, Cheongju, 28644, South Korea.
| | - Yong-Keun Jung
- School of Biological Sciences, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
6
|
Gao H, Wei L, Indulkar S, Nguyen TTL, Liu D, Ho MF, Zhang C, Li H, Weinshilboum RM, Ingle JN, Wang L. Androgen receptor-mediated pharmacogenomic expression quantitative trait loci: implications for breast cancer response to AR-targeting therapy. Breast Cancer Res 2024; 26:111. [PMID: 38965614 PMCID: PMC11225427 DOI: 10.1186/s13058-024-01861-2] [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: 11/17/2023] [Accepted: 06/20/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Endocrine therapy is the most important treatment modality of breast cancer patients whose tumors express the estrogen receptor α (ERα). The androgen receptor (AR) is also expressed in the vast majority (80-90%) of ERα-positive tumors. AR-targeting drugs are not used in clinical practice, but have been evaluated in multiple trials and preclinical studies. METHODS We performed a genome-wide study to identify hormone/drug-induced single nucleotide polymorphism (SNP) genotype - dependent gene-expression, known as PGx-eQTL, mediated by either an AR agonist (dihydrotestosterone) or a partial antagonist (enzalutamide), utilizing a previously well characterized lymphoblastic cell line panel. The association of the identified SNPs-gene pairs with breast cancer phenotypes were then examined using three genome-wide association (GWAS) studies that we have published and other studies from the GWAS catalog. RESULTS We identified 13 DHT-mediated PGx-eQTL loci and 23 Enz-mediated PGx-eQTL loci that were associated with breast cancer outcomes post ER antagonist or aromatase inhibitors (AI) treatment, or with pharmacodynamic (PD) effects of AIs. An additional 30 loci were found to be associated with cancer risk and sex-hormone binding globulin levels. The top loci involved the genes IDH2 and TMEM9, the expression of which were suppressed by DHT in a PGx-eQTL SNP genotype-dependent manner. Both of these genes were overexpressed in breast cancer and were associated with a poorer prognosis. Therefore, suppression of these genes by AR agonists may benefit patients with minor allele genotypes for these SNPs. CONCLUSIONS We identified AR-related PGx-eQTL SNP-gene pairs that were associated with risks, outcomes and PD effects of endocrine therapy that may provide potential biomarkers for individualized treatment of breast cancer.
Collapse
Affiliation(s)
- Huanyao Gao
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Lixuan Wei
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Shreya Indulkar
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Thanh Thanh L Nguyen
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Duan Liu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Ming-Fen Ho
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - Richard M Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA
| | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, MN, USA
| | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 200 First Street Southwest, Rochester, MN, 55905, USA.
| |
Collapse
|
7
|
Zhang W, Sha Z, Tang Y, Jin C, Gao W, Chen C, Yu L, Lv N, Liu S, Xu F, Wang D, Shi L. Defective Lamtor5 Leads to Autoimmunity by Deregulating v-ATPase and Lysosomal Acidification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400446. [PMID: 38639386 PMCID: PMC11165510 DOI: 10.1002/advs.202400446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/02/2024] [Indexed: 04/20/2024]
Abstract
Despite accumulating evidence linking defective lysosome function with autoimmune diseases, how the catabolic machinery is regulated to maintain immune homeostasis remains unknown. Late endosomal/lysosomal adaptor, MAPK and mTOR activator 5 (Lamtor5) is a subunit of the Ragulator mediating mechanistic target of rapamycin complex 1 (mTORC1) activation in response to amino acids, but its action mode and physiological role are still unclear. Here it is demonstrated that Lamtor5 level is markedly decreased in peripheral blood mononuclear cells (PBMCs) of patients with systemic lupus erythematosus (SLE). In parallel, the mice with myeloid Lamtor5 ablation developed SLE-like manifestation. Impaired lysosomal function and aberrant activation of mTORC1 are evidenced in Lamtor5 deficient macrophages and PBMCs of SLE patients, accompanied by blunted autolysosomal pathway and undesirable inflammatory responses. Mechanistically, it is shown that Lamtor5 is physically associated with ATP6V1A, an essential subunit of vacuolar H+-ATPase (v-ATPase), and promoted the V0/V1 holoenzyme assembly to facilitate lysosome acidification. The binding of Lamtor5 to v-ATPase affected the lysosomal tethering of Rag GTPase and weakened its interaction with mTORC1 for activation. Overall, Lamtor5 is identified as a critical factor for immune homeostasis by intergrading v-ATPase activity, lysosome function, and mTOR pathway. The findings provide a potential therapeutic target for SLE and/or other autoimmune diseases.
Collapse
Affiliation(s)
- Wei Zhang
- School of MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Zhou Sha
- School of MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Yunzhe Tang
- School of MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Cuiyuan Jin
- Key lab of Artificial Organs and Computational MedicineInstitute of Translational MedicineZhejiang Shuren UniversityHangzhou310022China
| | - Wenhua Gao
- School of MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Changmai Chen
- School of PharmacyFujian Medical UniversityFuzhou350122China
| | - Lang Yu
- School of MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Nianyin Lv
- School of MedicineNanjing University of Chinese MedicineNanjing210046China
| | - Shijia Liu
- The Affiliated Hospital of Nanjing University of Chinese MedicineNanjing210029China
| | - Feng Xu
- Department of Infectious DiseasesThe Second Affiliated HospitalZhejiang University School of MedicineHangzhou310009China
| | - Dandan Wang
- Department of Rheumatology and ImmunologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210093China
| | - Liyun Shi
- School of MedicineNanjing University of Chinese MedicineNanjing210046China
- Key lab of Artificial Organs and Computational MedicineInstitute of Translational MedicineZhejiang Shuren UniversityHangzhou310022China
| |
Collapse
|
8
|
Xue Y, Ruan Y, Wang Y, Xiao P, Xu J. Signaling pathways in liver cancer: pathogenesis and targeted therapy. MOLECULAR BIOMEDICINE 2024; 5:20. [PMID: 38816668 PMCID: PMC11139849 DOI: 10.1186/s43556-024-00184-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/23/2024] [Indexed: 06/01/2024] Open
Abstract
Liver cancer remains one of the most prevalent malignancies worldwide with high incidence and mortality rates. Due to its subtle onset, liver cancer is commonly diagnosed at a late stage when surgical interventions are no longer feasible. This situation highlights the critical role of systemic treatments, including targeted therapies, in bettering patient outcomes. Despite numerous studies on the mechanisms underlying liver cancer, tyrosine kinase inhibitors (TKIs) are the only widely used clinical inhibitors, represented by sorafenib, whose clinical application is greatly limited by the phenomenon of drug resistance. Here we show an in-depth discussion of the signaling pathways frequently implicated in liver cancer pathogenesis and the inhibitors targeting these pathways under investigation or already in use in the management of advanced liver cancer. We elucidate the oncogenic roles of these pathways in liver cancer especially hepatocellular carcinoma (HCC), as well as the current state of research on inhibitors respectively. Given that TKIs represent the sole class of targeted therapeutics for liver cancer employed in clinical practice, we have particularly focused on TKIs and the mechanisms of the commonly encountered phenomena of its resistance during HCC treatment. This necessitates the imperative development of innovative targeted strategies and the urgency of overcoming the existing limitations. This review endeavors to shed light on the utilization of targeted therapy in advanced liver cancer, with a vision to improve the unsatisfactory prognostic outlook for those patients.
Collapse
Affiliation(s)
- Yangtao Xue
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Yeling Ruan
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Yali Wang
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Peng Xiao
- Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Junjie Xu
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China.
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China.
- Zhejiang University Cancer Center, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China.
| |
Collapse
|
9
|
Cox CM, Wu MH, Padilla-Rodriguez M, Blum I, Momtaz S, Mitchell SAT, Wilson JM. Regulation of YAP and Wnt signaling by the endosomal protein MAMDC4. PLoS One 2024; 19:e0296003. [PMID: 38787854 PMCID: PMC11125477 DOI: 10.1371/journal.pone.0296003] [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: 03/27/2023] [Accepted: 12/04/2023] [Indexed: 05/26/2024] Open
Abstract
Maintenance of the intestinal epithelium requires constant self-renewal and regeneration. Tight regulation of proliferation and differentiation of intestinal stem cells within the crypt region is critical to maintaining homeostasis. The transcriptional co-factors β-catenin and YAP are required for proliferation during normal homeostasis as well as intestinal regeneration after injury: aberrant signaling activity results in over proliferation and tumorigenesis. Although both YAP and β-catenin activity are controlled along canonical pathways, it is becoming increasingly clear that non-canonical regulation of these transcriptional regulators plays a role in fine tuning their activity. We have shown previously that MAMDC4 (Endotubin, AEGP), an integral membrane protein present in endosomes, regulates both YAP and β-catenin activity in kidney epithelial cells and in the developing intestinal epithelium. Here we show that MAMDC4 interacts with members of the signalosome and mediates cross-talk between YAP and β-catenin. Interestingly, this cross-talk occurs through a non-canonical pathway involving interactions between AMOT:YAP and AMOT:β-catenin.
Collapse
Affiliation(s)
- Christopher M. Cox
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Meng-Han Wu
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Marco Padilla-Rodriguez
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Isabella Blum
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Samina Momtaz
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Stefanie A. T. Mitchell
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Jean M. Wilson
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States of America
- The University of Arizona Cancer Center, University of Arizona, Tucson, AZ, United States of America
- Bio5 Institute, University of Arizona, Tucson, AZ, United States of America
| |
Collapse
|
10
|
Wang Z, Zhao P, Tian K, Qiao Z, Dong H, Li J, Guan Z, Su H, Song Y, Ma X. TMEM9 promotes lung adenocarcinoma progression via activating the MEK/ERK/STAT3 pathway to induce VEGF expression. Cell Death Dis 2024; 15:295. [PMID: 38664392 PMCID: PMC11045738 DOI: 10.1038/s41419-024-06669-8] [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: 08/06/2023] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024]
Abstract
Abnormal Transmembrane protein 9 (TMEM9) expression has been identified in various human tumors. However, the prognostic potential and mechanistic role of TMEM9 in lung adenocarcinoma (LUAD) remain unclear. Here, we first found a significant upregulation of TMEM9 in LUAD tissues, and TMEM9 expression was positively correlated with microvessel density (MVD), T stage, and clinical stage. Survival analysis demonstrated TMEM9 was an independent indicator of poor prognosis in LUAD patients. In addition, downregulation of TMEM9 suppressed tumor growth and metastasis in vitro and in vivo models, and reduced HUVEC proliferation, migration, and tube formation in a cancer cell/HUVEC coculture model. Furthermore, TMEM9 upregulated VEGF expression, and VEGF-neutralizing antibodies reversed HUVEC angiogenesis and cancer cell migration ability caused by overexpression of TMEM9. In contrast, recombinant VEGF (rVEGF) abolished the inhibitory effect of TMEM9-knockdown LUAD cells on HUVEC angiogenesis and tumor cell migration. Moreover, we showed that TMEM9 upregulated VEGF expression by activating the mitogen-activated protein kinase/extracellular signal-regulated kinase/STAT3 (MEK/ERK/STAT3) pathway. Together, our study provides mechanistic insights into the role of TMEM9 in LUAD and highlights the potential of targeting the TMEM9/MEK/ERK/STAT3/VEGF pathway as a novel therapy for preventing LUAD progression.
Collapse
Affiliation(s)
- Zhiqian Wang
- Department of Oncology, Medical College of Qingdao University, Qingdao, Shandong, China
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, Shandong, China
| | - Peng Zhao
- Biotherapy Center, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China
| | - Kaihua Tian
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhongshi Qiao
- Department of Oncology, Medical College of Qingdao University, Qingdao, Shandong, China
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, Shandong, China
| | - Hao Dong
- Department of Oncology, Medical College of Qingdao University, Qingdao, Shandong, China
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jie Li
- Department of Oncology, Medical College of Qingdao University, Qingdao, Shandong, China
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zitong Guan
- Department of Oncology, Medical College of Qingdao University, Qingdao, Shandong, China
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, Shandong, China
| | - Hui Su
- Department of Oncology, LiaochengPeople's Hospital, Liaocheng, Shandong, China
| | - Yang Song
- Department of Nutrition and Food Hygiene, School of Public Health, Medical College of Qingdao University, Qingdao, Shandong, China.
| | - Xuezhen Ma
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
11
|
Zhou P, Zhang J, Feng J, Wang G. Construction of an oxidative phosphorylation-related gene signature for predicting prognosis and identifying immune infiltration in osteosarcoma. Aging (Albany NY) 2024; 16:5311-5335. [PMID: 38506898 PMCID: PMC11006489 DOI: 10.18632/aging.205650] [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: 11/09/2023] [Accepted: 02/13/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND Osteosarcoma is a prevalent malignant tumor that originates from mesenchymal tissue. It typically affects children and adolescents. Although it is known that the growth of osteosarcoma relies on oxidative phosphorylation for energy production, limited attention has been paid to exploring the potential of oxidative phosphorylation-related genes in predicting the prognosis of individuals suffering from osteosarcoma. METHODS All the data were retrieved from the UCSC Xena and GEO (GENE EXPRESSION OMNIBUS). Identification of the oxidative phosphorylation genes linked to the prognosis of individuals with osteosarcoma was done by means of univariate COX and LASSO regression analyses. Following that, patients were categorized into a high-risk group and a low-risk group as per the risk score determined by the identified oxidative phosphorylation genes. Furthermore, a comparison was made in terms of the survival and immune infiltration between both groups, and the prognostic model was established. RESULTS Five oxidative phosphorylation genes (ATP6V0D1, LHPP, COX6A2, MTHFD2, NDUFB9) associated with the prognosis of individuals with osteosarcoma were identified and the risk prognostic models were constructed. In the current research, the analysis of the ROC curves indicated a superior predictive accuracy exhibited by the risk model. The prognosis was adversely affected by immune infiltration in the high-risk group in comparison with the low-risk group. The function of the oxidative phosphorylation-related prognostic gene set was verified by GO and KEGG analysis. Furthermore, the link between oxidative phosphorylation-related genes and osteosarcoma immune infiltration was examined by GSEA analysis. CONCLUSIONS In this study, a prognostic model that demonstrated good predictive performance was constructed. Additionally, this study highlighted a correlation between oxidative phosphorylation-related genes and immune infiltration.
Collapse
Affiliation(s)
- Peng Zhou
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Department of Orthopedics, Affiliated Hospital of Chifeng University, Chifeng, Inner Mongolia, China
| | - Jin Zhang
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jinyan Feng
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Guowen Wang
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
12
|
Liu L, Chen X, Wu L, Huang K, Wang Z, Zheng Y, Zheng C, Zhang Z, Chen J, Wei J, Chen S, Jin W, Chen J, Wei D, Xu Y. Ubiquitin ligase subunit FBXO9 inhibits V-ATPase assembly and impedes lung cancer metastasis. Exp Hematol Oncol 2024; 13:32. [PMID: 38486234 PMCID: PMC10938814 DOI: 10.1186/s40164-024-00497-4] [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: 11/26/2023] [Accepted: 02/29/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND The evolutionarily conserved protein FBXO9 acts as a substrate receptor for the SKP1-cullin-1-RBX1 ubiquitin ligase and is implicated in cancer, exhibiting either tumor-suppressive or oncogenic effects depending on the specific tumor type. However, their role in lung cancer metastasis remains unclear. METHODS Lentiviral vectors carrying miRNA-based shRNA sequences for gene-specific knockdown were generated, and Lenti-CRISPR-Cas9 vectors containing gene-specific sgRNA sequences were designed. Gene overexpression was achieved using doxycycline-inducible lentiviral constructs, while gene knockdown or knockout cells were generated using shRNA and CRISPR-Cas9, respectively. Functional assays included migration, clonogenic survival assays, tumor sphere assays, and protein interaction studies using mass spectrometry, immunoprecipitation, and immunoblot analysis. RESULTS This study identified FBXO9 as a crucial regulator that suppresses lung cancer cell migration, tumor sphere growth and restricts metastasis. We showed that FBXO9 facilitates the ubiquitination of the catalytic subunit A (ATP6V1A) of the Vacuolar-type H+-ATPase (V-ATPase), resulting in its interaction with the cytoplasmic chaperone HSPA8 and subsequent sequestration within the cytoplasm. This process hinders the assembly of functional V-ATPase, resulting in reduced vesicular acidification. In contrast, depletion of FBXO9 reduced ATP6V1A ubiquitination, resulting in increased V-ATPase assembly and vesicular acidification, thus promoting pro-metastatic Wnt signaling and metastasis of lung cancer cells. Furthermore, we demonstrated the effectiveness of inhibitors targeting V-ATPase in inhibiting lung cancer metastasis in a mouse model. Finally, we established a correlation between lower FBXO9 levels and poorer survival outcomes in patients with lung cancer. CONCLUSION These findings collectively elucidate the critical role of FBXO9 in regulating V-ATPase assembly and provide a molecular basis for FBXO9's function in inhibiting lung cancer metastasis. This highlights the potential therapeutic opportunities of FBXO9 supplementation.
Collapse
Affiliation(s)
- Liang Liu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
- Institute of Clinical Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaodong Chen
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, Zhejiang Province, China
| | - Leilei Wu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Kaizong Huang
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Zhenyi Wang
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, Zhejiang Province, China
| | - Yaolin Zheng
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Cheng Zheng
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, Zhejiang Province, China
| | - Zhenshan Zhang
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Cancer Center, Shanghai, 200032, China
| | - Jiayan Chen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jiaming Wei
- Institute of Medicinal Biotechnology, Jiangsu College of Nursing, Huai'an, 223300, Jiangsu, China
| | - Song Chen
- Institute of Medicinal Biotechnology, Jiangsu College of Nursing, Huai'an, 223300, Jiangsu, China
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450053, Henan, China
| | - Weilin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Jinfei Chen
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 730000, Zhejiang Province, China
| | - Dongping Wei
- Medical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, Zhejiang Province, China.
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Yaping Xu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| |
Collapse
|
13
|
Billing AM, Kim YC, Gullaksen S, Schrage B, Raabe J, Hutzfeldt A, Demir F, Kovalenko E, Lassé M, Dugourd A, Fallegger R, Klampe B, Jaegers J, Li Q, Kravtsova O, Crespo-Masip M, Palermo A, Fenton RA, Hoxha E, Blankenberg S, Kirchhof P, Huber TB, Laugesen E, Zeller T, Chrysopoulou M, Saez-Rodriguez J, Magnussen C, Eschenhagen T, Staruschenko A, Siuzdak G, Poulsen PL, Schwab C, Cuello F, Vallon V, Rinschen MM. Metabolic Communication by SGLT2 Inhibition. Circulation 2024; 149:860-884. [PMID: 38152989 PMCID: PMC10922673 DOI: 10.1161/circulationaha.123.065517] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 11/22/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND SGLT2 (sodium-glucose cotransporter 2) inhibitors (SGLT2i) can protect the kidneys and heart, but the underlying mechanism remains poorly understood. METHODS To gain insights on primary effects of SGLT2i that are not confounded by pathophysiologic processes or are secondary to improvement by SGLT2i, we performed an in-depth proteomics, phosphoproteomics, and metabolomics analysis by integrating signatures from multiple metabolic organs and body fluids after 1 week of SGLT2i treatment of nondiabetic as well as diabetic mice with early and uncomplicated hyperglycemia. RESULTS Kidneys of nondiabetic mice reacted most strongly to SGLT2i in terms of proteomic reconfiguration, including evidence for less early proximal tubule glucotoxicity and a broad downregulation of the apical uptake transport machinery (including sodium, glucose, urate, purine bases, and amino acids), supported by mouse and human SGLT2 interactome studies. SGLT2i affected heart and liver signaling, but more reactive organs included the white adipose tissue, showing more lipolysis, and, particularly, the gut microbiome, with a lower relative abundance of bacteria taxa capable of fermenting phenylalanine and tryptophan to cardiovascular uremic toxins, resulting in lower plasma levels of these compounds (including p-cresol sulfate). SGLT2i was detectable in murine stool samples and its addition to human stool microbiota fermentation recapitulated some murine microbiome findings, suggesting direct inhibition of fermentation of aromatic amino acids and tryptophan. In mice lacking SGLT2 and in patients with decompensated heart failure or diabetes, the SGLT2i likewise reduced circulating p-cresol sulfate, and p-cresol impaired contractility and rhythm in human induced pluripotent stem cell-derived engineered heart tissue. CONCLUSIONS SGLT2i reduced microbiome formation of uremic toxins such as p-cresol sulfate and thereby their body exposure and need for renal detoxification, which, combined with direct kidney effects of SGLT2i, including less proximal tubule glucotoxicity and a broad downregulation of apical transporters (including sodium, amino acid, and urate uptake), provides a metabolic foundation for kidney and cardiovascular protection.
Collapse
Affiliation(s)
- Anja M. Billing
- Departments of Biomedicine (A.M.B., F.D., E.K., J.J., R.A.F., M.C., M.M.R.), Aarhus University, Denmark
| | - Young Chul Kim
- Departments of Medicine and Pharmacology, University of California San Diego, La Jolla (Y.C.K., M.C.-M., V.V.)
- VA San Diego Healthcare System, CA (Y.C.K., M.C.-M., V.V.)
| | - Søren Gullaksen
- Clinical Medicine (S.G., P.L.P.), Aarhus University, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Denmark (S.G., E.L.)
| | - Benedikt Schrage
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany (B.S., S.B., P.K., T.Z., C.M.)
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (B.S., J.R., S.B., P.K., T.Z., C.M., T.E., F.C.)
| | - Janice Raabe
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (B.S., J.R., S.B., P.K., T.Z., C.M., T.E., F.C.)
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.R., B.K., T.E., F.C.)
| | - Arvid Hutzfeldt
- III Department of Medicine and Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.H., M.L., E.H., T.B.H., M.M.R.)
| | - Fatih Demir
- Departments of Biomedicine (A.M.B., F.D., E.K., J.J., R.A.F., M.C., M.M.R.), Aarhus University, Denmark
| | - Elina Kovalenko
- Departments of Biomedicine (A.M.B., F.D., E.K., J.J., R.A.F., M.C., M.M.R.), Aarhus University, Denmark
| | - Moritz Lassé
- III Department of Medicine and Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.H., M.L., E.H., T.B.H., M.M.R.)
| | - Aurelien Dugourd
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, BioQuant, Heidelberg, Germany (A.D., R.F., J.S.-R.)
| | - Robin Fallegger
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, BioQuant, Heidelberg, Germany (A.D., R.F., J.S.-R.)
| | - Birgit Klampe
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.R., B.K., T.E., F.C.)
| | - Johannes Jaegers
- Departments of Biomedicine (A.M.B., F.D., E.K., J.J., R.A.F., M.C., M.M.R.), Aarhus University, Denmark
| | - Qing Li
- Engineering (Q.L., C.S.), Aarhus University, Denmark
| | - Olha Kravtsova
- Departments of Biomedicine (A.M.B., F.D., E.K., J.J., R.A.F., M.C., M.M.R.), Aarhus University, Denmark
| | - Maria Crespo-Masip
- Departments of Medicine and Pharmacology, University of California San Diego, La Jolla (Y.C.K., M.C.-M., V.V.)
- VA San Diego Healthcare System, CA (Y.C.K., M.C.-M., V.V.)
| | - Amelia Palermo
- Scripps Research, Center for Metabolomics, San Diego, CA (A.P., G.S., M.M.R.)
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles (A.P.)
| | - Robert A. Fenton
- Departments of Biomedicine (A.M.B., F.D., E.K., J.J., R.A.F., M.C., M.M.R.), Aarhus University, Denmark
| | - Elion Hoxha
- III Department of Medicine and Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.H., M.L., E.H., T.B.H., M.M.R.)
| | - Stefan Blankenberg
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany (B.S., S.B., P.K., T.Z., C.M.)
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (B.S., J.R., S.B., P.K., T.Z., C.M., T.E., F.C.)
| | - Paulus Kirchhof
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany (B.S., S.B., P.K., T.Z., C.M.)
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (B.S., J.R., S.B., P.K., T.Z., C.M., T.E., F.C.)
- Institute of Cardiovascular Sciences, University of Birmingham, United Kingdom (P.K.)
| | - Tobias B. Huber
- III Department of Medicine and Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.H., M.L., E.H., T.B.H., M.M.R.)
| | - Esben Laugesen
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Denmark (S.G., E.L.)
- Diagnostic Centre, Silkeborg Regional Hospital, Denmark (E.L.)
| | - Tanja Zeller
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany (B.S., S.B., P.K., T.Z., C.M.)
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (B.S., J.R., S.B., P.K., T.Z., C.M., T.E., F.C.)
| | - Maria Chrysopoulou
- Departments of Biomedicine (A.M.B., F.D., E.K., J.J., R.A.F., M.C., M.M.R.), Aarhus University, Denmark
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, BioQuant, Heidelberg, Germany (A.D., R.F., J.S.-R.)
| | - Christina Magnussen
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany (B.S., S.B., P.K., T.Z., C.M.)
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (B.S., J.R., S.B., P.K., T.Z., C.M., T.E., F.C.)
| | - Thomas Eschenhagen
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (B.S., J.R., S.B., P.K., T.Z., C.M., T.E., F.C.)
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.R., B.K., T.E., F.C.)
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa (O.K., A.S.)
| | - Gary Siuzdak
- Scripps Research, Center for Metabolomics, San Diego, CA (A.P., G.S., M.M.R.)
| | - Per L. Poulsen
- Clinical Medicine (S.G., P.L.P.), Aarhus University, Denmark
- Steno Diabetes Center (P.L.P.), Aarhus University, Denmark
| | | | - Friederike Cuello
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany (B.S., J.R., S.B., P.K., T.Z., C.M., T.E., F.C.)
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (J.R., B.K., T.E., F.C.)
| | - Volker Vallon
- Departments of Medicine and Pharmacology, University of California San Diego, La Jolla (Y.C.K., M.C.-M., V.V.)
- VA San Diego Healthcare System, CA (Y.C.K., M.C.-M., V.V.)
| | - Markus M. Rinschen
- Departments of Biomedicine (A.M.B., F.D., E.K., J.J., R.A.F., M.C., M.M.R.), Aarhus University, Denmark
- Aarhus Institute of Advanced Studies (M.M.R.), Aarhus University, Denmark
- III Department of Medicine and Hamburg Center for Kidney Health, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (A.H., M.L., E.H., T.B.H., M.M.R.)
- Scripps Research, Center for Metabolomics, San Diego, CA (A.P., G.S., M.M.R.)
| |
Collapse
|
14
|
Zhang D, Ma Y, Liu J, Wang D, Geng Z, Wen D, Chen H, Wang H, Li L, Zhu X, Wang X, Huang M, Zou C, Chen Y, Ma L. Fenofibrate improves hepatic steatosis, insulin resistance, and shapes the gut microbiome via TFEB-autophagy in NAFLD mice. Eur J Pharmacol 2023; 960:176159. [PMID: 37898287 DOI: 10.1016/j.ejphar.2023.176159] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 10/30/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major liver disease subtype worldwide, is commonly associated with insulin resistance and obesity. NAFLD is characterized by an excessive hepatic lipid accumulation, as well as hepatic steatosis. Fenofibrate is a peroxisome proliferator-activated receptor α agonist widely used in clinical therapy to effectively ameliorate the development of NAFLD, but its mechanism of action is incompletely understood. Here, we found that fenofibrate dramatically modulate the gut microbiota composition of high-fat diet (HFD)-induced NAFLD mouse model, and the change of gut microbiota composition is dependent on TFEB-autophagy axis. Furthermore, we also found that fenofibrate improved hepatic steatosis, and increased the activation of TFEB, which severed as a regulator of autophagy, thus, the protective effects of fenofibrate against NAFLD are depended on TFEB-autophagy axis. Our study demonstrates the host gene may influence the gut microbiota and highlights the role of TFEB and autophagy in the protective effect of NAFLD. This work expands our understanding of the regulatory interactions between the host and gut microbiota and provides novel strategies for alleviating obesity.
Collapse
Affiliation(s)
- Dan Zhang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Yicheng Ma
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, 650091, PR China
| | - Jianjun Liu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Biomedical Engineering, Kunming Medical University, Kunming, 650500, PR China
| | - Da Wang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Zuotao Geng
- Department of Pediatrics, Women and Children's Hospital of Lijiang, Lijiang, 674100, PR China
| | - Daiyan Wen
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Hang Chen
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Hui Wang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Lanyi Li
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Xiaotong Zhu
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Xuemin Wang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Minshan Huang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China
| | - Chenggang Zou
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, 650091, PR China.
| | - Yuanli Chen
- Faculty of Basic Medicine, Kunming Medical University, Kunming, 650500, PR China.
| | - Lanqing Ma
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, 650032, PR China.
| |
Collapse
|
15
|
Li W, Kawaguchi K, Tanaka S, He C, Maeshima Y, Suzuki E, Toi M. Cellular senescence triggers intracellular acidification and lysosomal pH alkalinized via ATP6AP2 attenuation in breast cancer cells. Commun Biol 2023; 6:1147. [PMID: 37993606 PMCID: PMC10665353 DOI: 10.1038/s42003-023-05433-6] [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: 03/22/2023] [Accepted: 10/09/2023] [Indexed: 11/24/2023] Open
Abstract
Several chemotherapeutic drugs induce senescence in cancer cells; however, the mechanisms underlying intracellular pH dysregulation in senescent cells remain unclear. Adenosine triphosphatase H+ transporting accessory protein 2 (ATP6AP2) plays a critical role in maintaining pH homeostasis in cellular compartments. Herein, we report the regulatory role of ATP6AP2 in senescent breast cancer cells treated with doxorubicin (Doxo) and abemaciclib (Abe). A decline in ATP6AP2 triggers aberrant pH levels that impair lysosomal function and cause immune profile changes in senescent breast cancer cells. Doxo and Abe elicited a stable senescent phenotype and altered the expression of senescence-related genes. Additionally, senescent cells show altered inflammatory and immune transcriptional profiles due to reprogramming of the senescence-associated secretory phenotype. These findings elucidate ATP6AP2-mediated cellular pH regulation and suggest a potential link in immune profile alteration during therapy-induced senescence in breast cancer cells, providing insights into the mechanisms involved in the senescence response to anticancer therapy.
Collapse
Affiliation(s)
- Wei Li
- Department of Breast Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kosuke Kawaguchi
- Department of Breast Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Sunao Tanaka
- Department of Breast Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Chenfeng He
- Department of Breast Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yurina Maeshima
- Department of Breast Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Eiji Suzuki
- Kobe City Medical Center General Hospital, 2-1-1 Minatojimaminami-cho, Chuo-ku, Kobe, 650-0047, Japan
| | - Masakazu Toi
- Department of Breast Surgery, Kyoto University Graduate School of Medicine, 54 Shogoin-kawaharacho, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
16
|
Herrera-Quiterio GA, Encarnación-Guevara S. The transmembrane proteins (TMEM) and their role in cell proliferation, migration, invasion, and epithelial-mesenchymal transition in cancer. Front Oncol 2023; 13:1244740. [PMID: 37936608 PMCID: PMC10627164 DOI: 10.3389/fonc.2023.1244740] [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: 06/23/2023] [Accepted: 09/11/2023] [Indexed: 11/09/2023] Open
Abstract
Transmembrane proteins (TMEM) are located in the different biological membranes of the cell and have at least one passage through these cellular compartments. TMEM proteins carry out a wide variety of functions necessary to maintain cell homeostasis TMEM165 participates in glycosylation protein, TMEM88 in the development of cardiomyocytes, TMEM45A in epidermal keratinization, and TMEM74 regulating autophagy. However, for many TMEM proteins, their physiological function remains unknown. The role of these proteins is being recently investigated in cancer since transcriptomic and proteomic studies have revealed that exits differential expression of TMEM proteins in different neoplasms concerning cancer-free tissues. Among the cellular processes in which TMEM proteins have been involved in cancer are the promotion or suppression of cell proliferation, epithelial-mesenchymal transition, invasion, migration, intravasation/extravasation, metastasis, modulation of the immune response, and response to antineoplastic drugs. Inclusive data suggests that the participation of TMEM proteins in these cellular events could be carried out through involvement in different cell signaling pathways. However, the exact mechanisms not clear. This review shows a description of the involvement of TMEM proteins that promote or decrease cell proliferation, migration, and invasion in cancer cells, describes those TMEM proteins for which both a tumor suppressor and a tumor promoter role have been identified, depending on the type of cancer in which the protein is expressed. As well as some TMEM proteins involved in chemoresistance. A better characterization of these proteins is required to improve the understanding of the tumors in which their expression and function are altered; in addition to improving the understanding of the role of these proteins in cancer will show those TMEM proteins be potential candidates as biomarkers of response to chemotherapy or prognostic biomarkers or as potential therapeutic targets in cancer.
Collapse
Affiliation(s)
| | - Sergio Encarnación-Guevara
- Laboratorio de Proteómica, Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| |
Collapse
|
17
|
Ge J, Yu YJ, Li JY, Li MY, Xia SM, Xue K, Wang SY, Yang C. Activating Wnt/β-catenin signaling by autophagic degradation of APC contributes to the osteoblast differentiation effect of soy isoflavone on osteoporotic mesenchymal stem cells. Acta Pharmacol Sin 2023; 44:1841-1855. [PMID: 36973541 PMCID: PMC10462682 DOI: 10.1038/s41401-023-01066-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 02/17/2023] [Indexed: 03/29/2023] Open
Abstract
The functional role of autophagy in regulating differentiation of bone marrow mesenchymal stem cells (MSCs) has been studied extensively, but the underlying mechanism remains largely unknown. The Wnt/β-catenin signaling pathway plays a pivotal role in the initiation of osteoblast differentiation of mesenchymal progenitor cells, and the stability of core protein β-catenin is tightly controlled by the APC/Axin/GSK-3β/Ck1α complex. Here we showed that genistein, a predominant soy isoflavone, stimulated osteoblast differentiation of MSCs in vivo and in vitro. Female rats were subjected to bilateral ovariectomy (OVX); four weeks after surgery the rats were orally administered genistein (50 mg·kg-1·d-1) for 8 weeks. The results showed that genistein administration significantly suppressed the bone loss and bone-fat imbalance, and stimulated bone formation in OVX rats. In vitro, genistein (10 nM) markedly activated autophagy and Wnt/β-catenin signaling pathway, and stimulated osteoblast differentiation in OVX-MSCs. Furthermore, we found that genistein promoted autophagic degradation of adenomatous polyposis coli (APC), thus initiated β-catenin-driven osteoblast differentiation. Notably, genistein activated autophagy through transcription factor EB (TFEB) rather than mammalian target of rapamycin (mTOR). These findings unveil the mechanism of how autophagy regulates osteogenesis in OVX-MSCs, which expands our understanding that such interplay could be employed as a useful therapeutic strategy for treating postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Jing Ge
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200001, China
| | - Ye-Jia Yu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200001, China
| | - Jia-Yi Li
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200001, China
| | - Meng-Yu Li
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200001, China
| | - Si-Mo Xia
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200001, China
| | - Ke Xue
- Department of Pastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Shao-Yi Wang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200001, China.
| | - Chi Yang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology, Shanghai, 200001, China.
| |
Collapse
|
18
|
Yaghoobi A, Seyedmirzaei H, Ala M. Genome- and Exome-Wide Association Studies Revealed Candidate Genes Associated with DaTscan Imaging Features. PARKINSON'S DISEASE 2023; 2023:2893662. [PMID: 37664790 PMCID: PMC10468272 DOI: 10.1155/2023/2893662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/02/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023]
Abstract
Introduction Despite remarkable progress in identifying Parkinson's disease (PD) genetic risk loci, the genetic basis of PD remains largely unknown. With the help of the endophenotype approach and using data from dopamine transporter single-photon emission computerized tomography (DaTscan), we identified potentially involved genes in PD. Method We conducted an imaging genetic study by performing exome-wide association study (EWAS) and genome-wide association study (GWAS) on the specific binding ratio (SBR) of six DaTscan anatomical areas between 489 and 559 subjects of Parkinson's progression markers initiative (PPMI) cohort and 83,623 and 36,845 single-nucleotide polymorphisms (SNPs)/insertion-deletion mutations (INDELs). We also investigated the association of cerebrospinal fluid (CSF) protein concentration of our significant genes with PD progression using PPMI CSF proteome data. Results Among 83,623 SNPs/INDELs in EWAS, one SNP (rs201465075) on 1 q32.1 locus was significantly (P value = 4.03 × 10-7) associated with left caudate DaTscan SBR, and 33 SNPs were suggestive. Among 36,845 SNPs in GWAS, one SNP (rs12450112) on 17 p.12 locus was significantly (P value = 1.34 × 10-6) associated with right anterior putamen DaTscan SBR, and 39 SNPs were suggestive among which 8 SNPs were intergenic. We found that rs201465075 and rs12450112 are most likely related to IGFN1 and MAP2K4 genes. The protein level of MAP2K4 in the CSF was significantly associated with PD progression in the PPMI cohort; however, proteomic data were not available for the IGFN1 gene. Conclusion We have shown that particular variants of IGFN1 and MAP2K4 genes may be associated with PD. Since DaTscan imaging could be positive in other Parkinsonian syndromes, caution should be taken when interpreting our results. Future experimental studies are also needed to verify these findings.
Collapse
Affiliation(s)
- Arash Yaghoobi
- Institute for Research in Fundamental Sciences (IPM), School of Biological Sciences, Tehran, Iran
| | - Homa Seyedmirzaei
- Interdisciplinary Neuroscience Research Program (INRP), Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Moein Ala
- Experimental Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Im E, Jiang Y, Stavrides PH, Darji S, Erdjument-Bromage H, Neubert TA, Choi JY, Wegiel J, Lee JH, Nixon RA. Lysosomal dysfunction in Down syndrome and Alzheimer mouse models is caused by v-ATPase inhibition by Tyr 682-phosphorylated APP βCTF. SCIENCE ADVANCES 2023; 9:eadg1925. [PMID: 37494443 PMCID: PMC10371027 DOI: 10.1126/sciadv.adg1925] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/23/2023] [Indexed: 07/28/2023]
Abstract
Lysosome dysfunction arises early and propels Alzheimer's disease (AD). Herein, we show that amyloid precursor protein (APP), linked to early-onset AD in Down syndrome (DS), acts directly via its β-C-terminal fragment (βCTF) to disrupt lysosomal vacuolar (H+)-adenosine triphosphatase (v-ATPase) and acidification. In human DS fibroblasts, the phosphorylated 682YENPTY internalization motif of APP-βCTF binds selectively within a pocket of the v-ATPase V0a1 subunit cytoplasmic domain and competitively inhibits association of the V1 subcomplex of v-ATPase, thereby reducing its activity. Lowering APP-βCTF Tyr682 phosphorylation restores v-ATPase and lysosome function in DS fibroblasts and in vivo in brains of DS model mice. Notably, lowering APP-βCTF Tyr682 phosphorylation below normal constitutive levels boosts v-ATPase assembly and activity, suggesting that v-ATPase may also be modulated tonically by phospho-APP-βCTF. Elevated APP-βCTF Tyr682 phosphorylation in two mouse AD models similarly disrupts v-ATPase function. These findings offer previously unknown insight into the pathogenic mechanism underlying faulty lysosomes in all forms of AD.
Collapse
Affiliation(s)
- Eunju Im
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ying Jiang
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Philip H. Stavrides
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Sandipkumar Darji
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
| | - Hediye Erdjument-Bromage
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Thomas A. Neubert
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Jun Yong Choi
- Department of Chemistry and Biochemistry, Queens College, Queens, NY 11367, USA
- Ph.D. Programs in Chemistry and Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, USA
| | - Jerzy Wegiel
- Department of Developmental Neurobiology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
| | - Ju-Hyun Lee
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ralph A. Nixon
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, NY 10962, USA
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
- NYU Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
20
|
Park WJ, Kim MJ. A New Wave of Targeting 'Undruggable' Wnt Signaling for Cancer Therapy: Challenges and Opportunities. Cells 2023; 12:cells12081110. [PMID: 37190019 DOI: 10.3390/cells12081110] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Aberrant Wnt signaling activation is frequently observed in many cancers. The mutation acquisition of Wnt signaling leads to tumorigenesis, whereas the inhibition of Wnt signaling robustly suppresses tumor development in various in vivo models. Based on the excellent preclinical effect of targeting Wnt signaling, over the past 40 years, numerous Wnt-targeted therapies have been investigated for cancer treatment. However, Wnt signaling-targeting drugs are still not clinically available. A major obstacle to Wnt targeting is the concomitant side effects during treatment due to the pleiotropic role of Wnt signaling in development, tissue homeostasis, and stem cells. Additionally, the complexity of the Wnt signaling cascades across different cancer contexts hinders the development of optimized targeted therapies. Although the therapeutic targeting of Wnt signaling remains challenging, alternative strategies have been continuously developed alongside technological advances. In this review, we give an overview of current Wnt targeting strategies and discuss recent promising trials that have the potential to be clinically realized based on their mechanism of action. Furthermore, we highlight new waves of Wnt targeting that combine recently developed technologies such as PROTAC/molecular glue, antibody-drug conjugates (ADC), and anti-sense oligonucleotides (ASO), which may provide us with new opportunities to target 'undruggable' Wnt signaling.
Collapse
Affiliation(s)
- Woo-Jung Park
- Department of Life Science, Gachon University, Seongnam 13120, Republic of Korea
| | - Moon Jong Kim
- Department of Life Science, Gachon University, Seongnam 13120, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Lee Gil Ya Cancer and Diabetes Institute, Incheon 21999, Republic of Korea
| |
Collapse
|
21
|
Chen Z, Zhou X, Zhou X, Tang Y, Lu M, Zhao J, Tian C, Wu M, Liu Y, Prochownik EV, Wang F, Li Y. Phosphomevalonate Kinase Controls β-Catenin Signaling via the Metabolite 5-Diphosphomevalonate. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204909. [PMID: 36808719 PMCID: PMC10131864 DOI: 10.1002/advs.202204909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 02/03/2023] [Indexed: 06/18/2023]
Abstract
β-catenin signaling is abnormally activated in cancer. Here, this work screens the mevalonate metabolic pathway enzyme PMVK to stabilize β-catenin signaling using a human genome-wide library. On the one hand, PMVK-produced MVA-5PP competitively binds to CKIα to prevent β-catenin Ser45 phosphorylation and degradation. On the other hand, PMVK functions as a protein kinase to directly phosphorylate β-catenin Ser184 to increase its protein nuclear localization. This synergistic effect of PMVK and MVA-5PP together promotes β-catenin signaling. In addition, PMVK deletion impairs mouse embryonic development and causes embryonic lethal. PMVK deficiency in liver tissue alleviates DEN/CCl4 -induced hepatocarcinogenesis. Finally, the small molecule inhibitor of PMVK, PMVKi5, is developed and PMVKi5 inhibits carcinogenesis of liver and colorectal tissues. These findings reveal a non-canonical function of a key metabolic enzyme PMVK and a novel link between the mevalonate pathway and β-catenin signaling in carcinogenesis providing a new target for clinical cancer therapy.
Collapse
Affiliation(s)
- Zhiqiang Chen
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesFrontier Science Center for Immunology and MetabolismTaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430072P. R. China
- Medical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan UniversityWuhan430071P. R. China
| | - Xinyi Zhou
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesFrontier Science Center for Immunology and MetabolismTaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430072P. R. China
- Medical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan UniversityWuhan430071P. R. China
| | - Xiaojun Zhou
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesFrontier Science Center for Immunology and MetabolismTaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430072P. R. China
- Medical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan UniversityWuhan430071P. R. China
| | - Yi Tang
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesFrontier Science Center for Immunology and MetabolismTaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430072P. R. China
- Medical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan UniversityWuhan430071P. R. China
| | - Mingzhu Lu
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesFrontier Science Center for Immunology and MetabolismTaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430072P. R. China
- Medical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan UniversityWuhan430071P. R. China
| | - Jianhong Zhao
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesFrontier Science Center for Immunology and MetabolismTaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430072P. R. China
- Medical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan UniversityWuhan430071P. R. China
| | - Chenhui Tian
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesFrontier Science Center for Immunology and MetabolismTaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430072P. R. China
- Medical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan UniversityWuhan430071P. R. China
| | - Mingzhi Wu
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesFrontier Science Center for Immunology and MetabolismTaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430072P. R. China
- Medical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan UniversityWuhan430071P. R. China
| | - Yanliang Liu
- Department of Gastrointestinal SurgeryRenmin Hospital of Wuhan UniversityWuhan430060P. R. China
| | - Edward V. Prochownik
- Division of Hematology/OncologyChildren's Hospital of Pittsburgh of UPMCDepartment of Microbiology and Molecular GeneticsPittsburgh Liver Research Center and Hillman Cancer Center of UPMCUniversity of Pittsburgh Medical CenterPittsburghPA15224USA
| | - Fubing Wang
- Department of Laboratory Medicine and Center for Single‐Cell Omics and Tumor Liquid BiopsyZhongnan Hospital of Wuhan UniversityWuhan430071P. R. China
- Wuhan Research Center for Infectious Diseases and CancerChinese Academy of Medical SciencesWuhan430071P. R. China
| | - Youjun Li
- Hubei Key Laboratory of Cell HomeostasisCollege of Life SciencesFrontier Science Center for Immunology and MetabolismTaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430072P. R. China
- Medical Research InstituteZhongnan Hospital of Wuhan UniversityWuhan UniversityWuhan430071P. R. China
| |
Collapse
|
22
|
Reprogramming of palmitic acid induced by dephosphorylation of ACOX1 promotes β-catenin palmitoylation to drive colorectal cancer progression. Cell Discov 2023; 9:26. [PMID: 36878899 PMCID: PMC9988979 DOI: 10.1038/s41421-022-00515-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 12/30/2022] [Indexed: 03/08/2023] Open
Abstract
Metabolic reprogramming is a hallmark of cancer. However, it is not well known how metabolism affects cancer progression. We identified that metabolic enzyme acyl-CoA oxidase 1 (ACOX1) suppresses colorectal cancer (CRC) progression by regulating palmitic acid (PA) reprogramming. ACOX1 is highly downregulated in CRC, which predicts poor clinical outcome in CRC patients. Functionally, ACOX1 depletion promotes CRC cell proliferation in vitro and colorectal tumorigenesis in mouse models, whereas ACOX1 overexpression inhibits patient-derived xenograft growth. Mechanistically, DUSP14 dephosphorylates ACOX1 at serine 26, promoting its polyubiquitination and proteasomal degradation, thereby leading to an increase of the ACOX1 substrate PA. Accumulated PA promotes β-catenin cysteine 466 palmitoylation, which inhibits CK1- and GSK3-directed phosphorylation of β-catenin and subsequent β-Trcp-mediated proteasomal degradation. In return, stabilized β-catenin directly represses ACOX1 transcription and indirectly activates DUSP14 transcription by upregulating c-Myc, a typical target of β-catenin. Finally, we confirmed that the DUSP14-ACOX1-PA-β-catenin axis is dysregulated in clinical CRC samples. Together, these results identify ACOX1 as a tumor suppressor, the downregulation of which increases PA-mediated β-catenin palmitoylation and stabilization and hyperactivates β-catenin signaling thus promoting CRC progression. Particularly, targeting β-catenin palmitoylation by 2-bromopalmitate (2-BP) can efficiently inhibit β-catenin-dependent tumor growth in vivo, and pharmacological inhibition of DUSP14-ACOX1-β-catenin axis by Nu-7441 reduced the viability of CRC cells. Our results reveal an unexpected role of PA reprogramming induced by dephosphorylation of ACOX1 in activating β-catenin signaling and promoting cancer progression, and propose the inhibition of the dephosphorylation of ACOX1 by DUSP14 or β-catenin palmitoylation as a viable option for CRC treatment.
Collapse
|
23
|
Ko KP, Zhang S, Huang Y, Kim B, Zou G, Jun S, Zhang J, Martin C, Dunbar KJ, Efe G, Rustgi AK, Zhang H, Nakagawa H, Park JI. Tumor Niche Network-Defined Subtypes Predict Immunotherapy Response of Esophageal Squamous Cell Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528539. [PMID: 36824935 PMCID: PMC9949073 DOI: 10.1101/2023.02.15.528539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Despite the promising outcomes of immune checkpoint blockade (ICB), resistance to ICB presents a new challenge. Therefore, selecting patients for specific ICB applications is crucial for maximizing therapeutic efficacy. Herein we curated 69 human esophageal squamous cell cancer (ESCC) patients' tumor microenvironment (TME) single-cell transcriptomic datasets to subtype ESCC. Integrative analyses of the cellular network transcriptional signatures of T cells, myeloid cells, and fibroblasts define distinct ESCC subtypes characterized by T cell exhaustion, Interferon (IFN) a/b signaling, TIGIT enrichment, and specific marker genes. Furthermore, this approach classifies ESCC patients into ICB responders and non-responders, as validated by liquid biopsy single-cell transcriptomics. Our study stratifies ESCC patients based on TME transcriptional network, providing novel insights into tumor niche remodeling and predicting ICB responses in ESCC patients.
Collapse
Affiliation(s)
- Kyung-Pil Ko
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shengzhe Zhang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yuanjian Huang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bongjun Kim
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gengyi Zou
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sohee Jun
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jie Zhang
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cecilia Martin
- Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Karen J. Dunbar
- Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Gizem Efe
- Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Anil K. Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Haiyang Zhang
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hiroshi Nakagawa
- Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
24
|
Misawa T, Hitomi K, Miyata K, Tanaka Y, Fujii R, Chiba M, Loo TM, Hanyu A, Kawasaki H, Kato H, Maezawa Y, Yokote K, Nakamura AJ, Ueda K, Yaegashi N, Takahashi A. Identification of Novel Senescent Markers in Small Extracellular Vesicles. Int J Mol Sci 2023; 24:ijms24032421. [PMID: 36768745 PMCID: PMC9916821 DOI: 10.3390/ijms24032421] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Senescent cells exhibit several typical features, including the senescence-associated secretory phenotype (SASP), promoting the secretion of various inflammatory proteins and small extracellular vesicles (EVs). SASP factors cause chronic inflammation, leading to age-related diseases. Recently, therapeutic strategies targeting senescent cells, known as senolytics, have gained attention; however, noninvasive methods to detect senescent cells in living organisms have not been established. Therefore, the goal of this study was to identify novel senescent markers using small EVs (sEVs). sEVs were isolated from young and senescent fibroblasts using three different methods, including size-exclusion chromatography, affinity column for phosphatidylserine, and immunoprecipitation using antibodies against tetraspanin proteins, followed by mass spectrometry. Principal component analysis revealed that the protein composition of sEVs released from senescent cells was significantly different from that of young cells. Importantly, we identified ATP6V0D1 and RTN4 as novel markers that are frequently upregulated in sEVs from senescent and progeria cells derived from patients with Werner syndrome. Furthermore, these two proteins were significantly enriched in sEVs from the serum of aged mice. This study supports the potential use of senescent markers from sEVs to detect the presence of senescent cells in vivo.
Collapse
Affiliation(s)
- Tomoka Misawa
- Division of Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi 980-8575, Japan
| | - Kazuhiro Hitomi
- Division of Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
- Graduate School of Science and Engineering, Ibaraki University, Ibaraki 310-8512, Japan
| | - Kenichi Miyata
- Division of Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Yoko Tanaka
- Division of Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Risa Fujii
- Project for Personalized Cancer Medicine, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Masatomo Chiba
- Division of Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Tze Mun Loo
- Division of Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Aki Hanyu
- Division of Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Hiroko Kawasaki
- Division of Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Hisaya Kato
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba 260-0856, Japan
| | - Yoshiro Maezawa
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba 260-0856, Japan
| | - Koutaro Yokote
- Department of Endocrinology, Hematology and Gerontology, Graduate School of Medicine, Chiba University, Chiba 260-0856, Japan
| | - Asako J. Nakamura
- Graduate School of Science and Engineering, Ibaraki University, Ibaraki 310-8512, Japan
| | - Koji Ueda
- Project for Personalized Cancer Medicine, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Miyagi 980-8575, Japan
| | - Akiko Takahashi
- Division of Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
- Advanced Research & Development Programs for Medical Innovation (PRIME), Japan Agency for Medical Research and Development (AMED), Tokyo 104-0004, Japan
- Cancer Cell Communication Project, NEXT-Ganken Program, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
- Correspondence:
| |
Collapse
|
25
|
Zou G, Park JI. Wnt signaling in liver regeneration, disease, and cancer. Clin Mol Hepatol 2023; 29:33-50. [PMID: 35785913 PMCID: PMC9845677 DOI: 10.3350/cmh.2022.0058] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/30/2022] [Indexed: 02/02/2023] Open
Abstract
The liver exhibits the highest recovery rate from acute injuries. However, in chronic liver disease, the long-term loss of hepatocytes often leads to adverse consequences such as fibrosis, cirrhosis, and liver cancer. The Wnt signaling plays a pivotal role in both liver regeneration and tumorigenesis. Therefore, manipulating the Wnt signaling has become an attractive approach to treating liver disease, including cancer. Nonetheless, given the crucial roles of Wnt signaling in physiological processes, blocking Wnt signaling can also cause several adverse effects. Recent studies have identified cancer-specific regulators of Wnt signaling, which would overcome the limitation of Wnt signaling target approaches. In this review, we discussed the role of Wnt signaling in liver regeneration, precancerous lesion, and liver cancer. Furthermore, we summarized the basic and clinical approaches of Wnt signaling blockade and proposed the therapeutic prospects of cancer-specific Wnt signaling blockade for liver cancer treatment.
Collapse
Affiliation(s)
- Gengyi Zou
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Corresponding author : Gengyi Zou Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd Unit 1054, Houston, TX 77030, USA Tel: +1-713-792-3659, Fax: +1-713-794-5369, E-mail:
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA,Genetics and Epigenetics Program, The University of Texas MD Anderson Cancer Center Graduate School of Biomedical Sciences, Houston, TX, USA,Jae-Il Park Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Blvd. Unit 1052, Houston, TX 77030, USA Tel: +1-713-792-3659, Fax: +1-713-794-5369, E-mail:
| |
Collapse
|
26
|
Zhang S, Lee SH, Nie L, Huang Y, Zou G, Jung Y, Jun S, Zhang J, Lien EM, Chen J, Park J. Lysosomal TMEM9-LAMTOR4-controlled mTOR signaling integrity is required for mammary tumorigenesis. CANCER COMMUNICATIONS (LONDON, ENGLAND) 2022; 43:159-163. [PMID: 36336962 PMCID: PMC9859727 DOI: 10.1002/cac2.12382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 09/16/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Shengzhe Zhang
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Sung Ho Lee
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Litong Nie
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Yuanjian Huang
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Gengyi Zou
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Youn‐Sang Jung
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Sohee Jun
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Jie Zhang
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Esther M. Lien
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Junjie Chen
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Jae‐Il Park
- Department of Experimental Radiation OncologyDivision of Radiation Oncologythe University of Texas MD Anderson Cancer CenterHoustonTX77030USA,Graduate School of Biomedical Sciencesthe University of Texas MD Anderson Cancer CenterHoustonTX77030USA,Program in Genetics and Epigeneticsthe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| |
Collapse
|
27
|
Castroflorio E, Pérez Berná AJ, López-Márquez A, Badosa C, Loza-Alvarez P, Roldán M, Jiménez-Mallebrera C. The Capillary Morphogenesis Gene 2 Triggers the Intracellular Hallmarks of Collagen VI-Related Muscular Dystrophy. Int J Mol Sci 2022; 23:ijms23147651. [PMID: 35886995 PMCID: PMC9322809 DOI: 10.3390/ijms23147651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022] Open
Abstract
Collagen VI-related disorders (COL6-RD) represent a severe form of congenital disease for which there is no treatment. Dominant-negative pathogenic variants in the genes encoding α chains of collagen VI are the main cause of COL6-RD. Here we report that patient-derived fibroblasts carrying a common single nucleotide variant mutation are unable to build the extracellular collagen VI network. This correlates with the intracellular accumulation of endosomes and lysosomes triggered by the increased phosphorylation of the collagen VI receptor CMG2. Notably, using a CRISPR-Cas9 gene-editing tool to silence the dominant-negative mutation in patients’ cells, we rescued the normal extracellular collagen VI network, CMG2 phosphorylation levels, and the accumulation of endosomes and lysosomes. Our findings reveal an unanticipated role of CMG2 in regulating endosomal and lysosomal homeostasis and suggest that mutated collagen VI dysregulates the intracellular environment in fibroblasts in collagen VI-related muscular dystrophy.
Collapse
Affiliation(s)
- Enrico Castroflorio
- ICFO-The Institute of Photonic Sciences, The Barcelona Institute of Science and Technology, 08860 Castelldefels, Spain;
- Correspondence: (E.C.); (C.J.-M.)
| | | | - Arístides López-Márquez
- Laboratorio de Investigación Aplicada en Enfermedades Neuromusculares, Unidad de Patología Neuromuscular, Servicio de Neuropediatría, Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain; (A.L.-M.); (C.B.)
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain;
- Centro de Investigaciones Biomédicas en Red de Enfermedades Rara (CIBERER), 28029 Madrid, Spain
| | - Carmen Badosa
- Laboratorio de Investigación Aplicada en Enfermedades Neuromusculares, Unidad de Patología Neuromuscular, Servicio de Neuropediatría, Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain; (A.L.-M.); (C.B.)
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain;
| | - Pablo Loza-Alvarez
- ICFO-The Institute of Photonic Sciences, The Barcelona Institute of Science and Technology, 08860 Castelldefels, Spain;
| | - Mónica Roldán
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain;
- Unitat de Microscòpia Confocal i Imatge Cellular, Servei de Medicina Genètica i Molecular, Institut Pediàtric de Malaties Rares (IPER), Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain
| | - Cecilia Jiménez-Mallebrera
- Laboratorio de Investigación Aplicada en Enfermedades Neuromusculares, Unidad de Patología Neuromuscular, Servicio de Neuropediatría, Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain; (A.L.-M.); (C.B.)
- Institut de Recerca Sant Joan de Déu, 08950 Esplugues de Llobregat, Spain;
- Centro de Investigaciones Biomédicas en Red de Enfermedades Rara (CIBERER), 28029 Madrid, Spain
- Department of Genetics, University of Barcelona, 08028 Barcelona, Spain
- Correspondence: (E.C.); (C.J.-M.)
| |
Collapse
|
28
|
Montalvo-Martínez L, Cruz-Carrillo G, Maldonado-Ruiz R, Trujillo-Villarreal LA, Cardenas-Tueme M, Viveros-Contreras R, Ortiz-López R, Camacho-Morales A. Transgenerational Susceptibility to Food Addiction-Like Behavior in Rats Associates to a Decrease of the Anti-Inflammatory IL-10 in Plasma. Neurochem Res 2022; 47:3093-3103. [PMID: 35767136 DOI: 10.1007/s11064-022-03660-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 11/25/2022]
Abstract
Maternal nutritional programming by energy-dense foods leads to the transgenerational heritance of addiction-like behavior. Exposure to energy-dense foods also activates systemic and central inflammation in the offspring. This study aimed to characterize pro- and anti-inflammatory cytokine profiles in blood and their correlation to the transgenerational heritance of the addiction-like behavior in rats. F1 offspring of male Wistar diagnosed with addiction-like behavior were mated with virgin females to generate the F2 and the F3 offspring, respectively. Diagnosis of addiction-like behavior was performed by the operant training schedule (FR1, FR5 and PR) and pro- and anti-inflammatory cytokine profiles in blood were measured by multiplex platform. Multiple linear models between behavior, fetal programming by diet and pro- and anti-inflammatory cytokine profiles were performed. We found that the addiction-like behavior found in the F1 male offspring exposed to energy-dense food (cafeteria, CAF) diet during fetal programing is transgenerational inherited to the F2 and F3 generations. Blood from addiction-like behavior subjects of F2 and F3 generations exposed to CAF diet during maternal programming showed decrease in the anti-inflammatory IL-10 in the plasma. Conversely, decreased levels of the pro-inflammatory MCP-1 was identified in non-addiction-like subjects. No changes were found in plasmatic TNF-α levels in the F2 and F3 offspring of non-addiction-like and addiction-like subjects. Finally, biological modeling between IL-10 or MCP-1 plasma levels and prenatal diet exposure on operant training responses confirmed an association of decreased IL-10 levels on addiction-like behavior in the F2 and F3 generations. Globally, we identified decreased anti-inflammatory IL-10 cytokine in the blood of F2 and F3 offspring subjects diagnosed with addiction-like behavior for food rewards.
Collapse
Affiliation(s)
- Larisa Montalvo-Martínez
- Department of Biochemistry, College of Medicine, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Monterrey, NL, Mexico
- Neurometabolism Unit, Center for Research and Development in Health Sciences, Universidad Autónoma de Nuevo León, Monterrey, NL, Mexico
| | - Gabriela Cruz-Carrillo
- Department of Biochemistry, College of Medicine, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Monterrey, NL, Mexico
- Neurometabolism Unit, Center for Research and Development in Health Sciences, Universidad Autónoma de Nuevo León, Monterrey, NL, Mexico
| | - Roger Maldonado-Ruiz
- Department of Biochemistry, College of Medicine, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Monterrey, NL, Mexico
- Neurometabolism Unit, Center for Research and Development in Health Sciences, Universidad Autónoma de Nuevo León, Monterrey, NL, Mexico
| | - Luis A Trujillo-Villarreal
- Department of Biochemistry, College of Medicine, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Monterrey, NL, Mexico
- Neurometabolism Unit, Center for Research and Development in Health Sciences, Universidad Autónoma de Nuevo León, Monterrey, NL, Mexico
| | - Marcela Cardenas-Tueme
- Institute for Obesity Research. Escuela de Medicina y Ciencias de la Salud, Instituto Tecnológico de Estudios Superiores Monterrey, Monterrey, NL, Mexico
| | | | - Rocío Ortiz-López
- Institute for Obesity Research. Escuela de Medicina y Ciencias de la Salud, Instituto Tecnológico de Estudios Superiores Monterrey, Monterrey, NL, Mexico
| | - Alberto Camacho-Morales
- Department of Biochemistry, College of Medicine, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Monterrey, NL, Mexico.
- Neurometabolism Unit, Center for Research and Development in Health Sciences, Universidad Autónoma de Nuevo León, Monterrey, NL, Mexico.
- Departamento de Bioquímica, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Madero y Dr. Aguirre Pequeño. Col. Mitras Centro, S/N, C.P. 64460, Monterrey, NL, Mexico.
| |
Collapse
|
29
|
Murphy WA, Lin N, Damask A, Schwartz GG, Steg PG, Szarek M, Banerjee P, Fazio S, Manvelian G, Pordy R, Shuldiner AR, Paulding C. Pharmacogenomic Study of Statin-Associated Muscle Symptoms in the ODYSSEY OUTCOMES Trial. Circ Genom Precis Med 2022; 15:e003503. [PMID: 35543701 PMCID: PMC9213083 DOI: 10.1161/circgen.121.003503] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Statin-associated muscle symptoms (SAMS) are the most frequently reported adverse events for statin therapies. Previous studies have reported an association between the p.Val174Ala missense variant in SLCO1B1 and SAMS in simvastatin-treated subjects; however, evidence for genetic predictors of SAMS in atorvastatin- or rosuvastatin-treated subjects is currently lacking.
Collapse
Affiliation(s)
- William A. Murphy
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill‚ Chapel Hill‚ NC (W.A.M.)
| | - Nan Lin
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY (N.L., A.D., P.B., S.F., G.M., R.P., A.R.S., C.P.)
| | - Amy Damask
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill‚ Chapel Hill‚ NC (W.A.M.)
| | | | - P. Gabriel Steg
- Université de Paris, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Paris‚ INSERM U1148, France (P.G.S.)
| | - Michael Szarek
- University of Colorado School of Medicine, Aurora‚ CO (G.G.S., M.S.)
- Department of Biostatistics and Epidemiology, SUNY Downstate School of Public Health, Brooklyn, NY (M.S.)
- CPC Clinical Research, Aurora, CO (M.S.)
| | - Poulabi Banerjee
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY (N.L., A.D., P.B., S.F., G.M., R.P., A.R.S., C.P.)
| | - Sergio Fazio
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY (N.L., A.D., P.B., S.F., G.M., R.P., A.R.S., C.P.)
| | - Garen Manvelian
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY (N.L., A.D., P.B., S.F., G.M., R.P., A.R.S., C.P.)
| | - Robert Pordy
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY (N.L., A.D., P.B., S.F., G.M., R.P., A.R.S., C.P.)
| | - Alan R. Shuldiner
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY (N.L., A.D., P.B., S.F., G.M., R.P., A.R.S., C.P.)
| | - Charles Paulding
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc, Tarrytown, NY (N.L., A.D., P.B., S.F., G.M., R.P., A.R.S., C.P.)
| |
Collapse
|
30
|
Lu Y, Zhong L, Luo X, Liu C, Dan W, Chu X, Wan P, Zhang Z, Wang X, Liu Z, Liu B. MiRNA-301b-3p induces proliferation and inhibits apoptosis in AML cells by targeting FOXF2 and regulating Wnt/β-catenin axis. Mol Cell Probes 2022; 63:101805. [PMID: 35259424 DOI: 10.1016/j.mcp.2022.101805] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/10/2022] [Accepted: 02/26/2022] [Indexed: 01/15/2023]
Abstract
BACKGROUND MiRNA-301b-3p functions as an oncomiRNA or tumor suppressor, and has been reported in various cancer types, including pancreatic, colorectal, oral, hepatocellular and lung cancers. Although the expression of miRNA-301b-3p is upregulated in acute myeloid leukemia (AML), its biological function and precise mechanisms remain unclarified. This study explores the roles of miRNA-301b-3p in AML, with the aim of ascertaining its regulatory action on Wnt/β-catenin axis by targeting Forkhead box F2 (FOXF2). METHODS The expression levels of miRNA-301b-3p and FOXF2 were measured by quantitative real-time PCR. The effects of miRNA-301b-3p knockdown and overexpression on cell proliferation were evaluated by CCK8 and cell counting assays, while cell apoptosis was analyzed by flow cytometry. The expression levels of apoptosis-related proteins, including FOXF2, and other targets in Wnt/β-catenin axis were determined by immunoblotting. Possible interaction between miRNA-301-3p and FOXF2 in AML cells was examined by luciferase reporter assays. RESULTS MiRNA-301b-3p was dramatically upregulated in AML cells, and showed a negative correlation with FOXF2 expression. Downregulation of miRNA-301b-3p suppressed proliferation and promoted apoptosis in AML cells. MiRNA-301b targeted FOXF2 to regulate Wnt/β-catenin axis. In the rescue experiments, FOXF2 overexpression partly reversed the effect of miRNA-301b-3p mimic in AML cells. CONCLUSION The current findings demonstrate that miRNA-301b-3p targets FOXF2 to induce proliferation and inhibit apoptosis in AML cells via regulation of Wnt/β-catenin axis.
Collapse
Affiliation(s)
- Yang Lu
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160, China
| | - Liang Zhong
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Xu Luo
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160, China
| | - Chen Liu
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Wenran Dan
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160, China
| | - Xuan Chu
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160, China
| | - Peng Wan
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160, China
| | - Zhonghui Zhang
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160, China
| | - Xiao Wang
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160, China
| | - Zhenyan Liu
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160, China
| | - Beizhong Liu
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160, China; Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
31
|
Tejeda-Muñoz N, Mei KC, Sheladiya P, Monka J. Targeting Membrane Trafficking as a Strategy for Cancer Treatment. Vaccines (Basel) 2022; 10:vaccines10050790. [PMID: 35632546 PMCID: PMC9144176 DOI: 10.3390/vaccines10050790] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/07/2022] [Accepted: 05/15/2022] [Indexed: 12/17/2022] Open
Abstract
Membrane trafficking is emerging as an attractive therapeutic strategy for cancer. Recent reports have found a connection between Wnt signaling, receptor-mediated endocytosis, V-ATPase, lysosomal activity, and macropinocytosis through the canonical Wnt pathway. In macropinocytic cells, a massive internalization of the plasma membrane can lead to the loss of cell-surface cadherins, integrins, and other antigens that mediate cell–cell adhesion, favoring an invasive phenotype. V-ATPase is a key regulator in maintaining proper membrane trafficking, homeostasis, and the earliest developmental decisions in the Xenopus vertebrate development model system. Here, we review how the interference of membrane trafficking with membrane trafficking inhibitors might be clinically relevant in humans.
Collapse
Affiliation(s)
- Nydia Tejeda-Muñoz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA; (P.S.); (J.M.)
- Correspondence:
| | - Kuo-Ching Mei
- Division of Pharmacoengineering and Molecular School Pharmaceutics, Eshelman of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Pooja Sheladiya
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA; (P.S.); (J.M.)
| | - Julia Monka
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1662, USA; (P.S.); (J.M.)
| |
Collapse
|
32
|
Nazarian A, Arbeev KG, Yashkin AP, Kulminski AM. Genome-wide analysis of genetic predisposition to common polygenic cancers. J Appl Genet 2022; 63:315-325. [PMID: 34981446 PMCID: PMC8983541 DOI: 10.1007/s13353-021-00679-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 12/13/2021] [Accepted: 12/23/2021] [Indexed: 12/16/2022]
Abstract
Lung, breast, prostate, and colorectal cancers are among the most common and fatal malignancies worldwide. They are mainly caused by multifactorial mechanisms and are genetically heterogeneous. We investigated the genetic architecture of these cancers through genome-wide association, pathway-based, and summary-based transcriptome-/methylome-wide association analyses using three independent cohorts. Our genome-wide association analyses identified the associations of 33 single-nucleotide polymorphisms (SNPs) at P < 5E - 06, of which 32 SNPs were not previously reported and did not have proxy variants within their ± 1 Mb flanking regions. Moreover, other polymorphisms mapped to their closest genes were not previously associated with the same cancers at P < 5E - 06. Our pathway enrichment analyses revealed associations of 32 pathways; mainly related to the immune system, DNA replication/transcription, and chromosomal organization; with the studied cancers. Also, 60 probes were associated with these cancers in our transcriptome-wide and methylome-wide analyses. The ± 1 Mb flanking regions of most probes had not attained P < 5E - 06 in genome-wide association studies. The genes corresponding to the significant probes can be considered as potential targets for further functional studies. Two genes (i.e., CDC14A and PMEL) demonstrated stronger evidence of associations with lung cancer as they had significant probes in both transcriptome-wide and methylome-wide association analyses. The novel cancer-associated SNPs and genes identified here would advance our understanding of the genetic heterogeneity of the common cancers.
Collapse
Affiliation(s)
- Alireza Nazarian
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Erwin Mill Building, 2024 W. Main St., Durham, NC, 27705, USA.
| | - Konstantin G Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Erwin Mill Building, 2024 W. Main St., Durham, NC, 27705, USA
| | - Arseniy P Yashkin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Erwin Mill Building, 2024 W. Main St., Durham, NC, 27705, USA
| | - Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Erwin Mill Building, 2024 W. Main St., Durham, NC, 27705, USA.
| |
Collapse
|
33
|
Abstract
Lysosomes are the digestive center of the cell and play important roles in human diseases, including cancer. Previous work has suggested that late endosomes, also known as multivesicular bodies (MVBs), and lysosomes are essential for canonical Wnt pathway signaling. Sequestration of Glycogen Synthase 3 (GSK3) and of β‐catenin destruction complex components in MVBs is required for sustained canonical Wnt signaling. Little is known about the role of lysosomes during early development. In the Xenopus egg, a Wnt-like cytoplasmic determinant signal initiates formation of the body axis following a cortical rotation triggered by sperm entry. Here we report that cathepsin D was activated in lysosomes specifically on the dorsal marginal zone of the embryo at the 64-cell stage, long before zygotic transcription starts. Expansion of the MVB compartment with low-dose hydroxychloroquine (HCQ) greatly potentiated the dorsalizing effects of the Wnt agonist lithium chloride (LiCl) in embryos, and this effect required macropinocytosis. Formation of the dorsal axis required lysosomes, as indicated by brief treatments with the vacuolar ATPase (V-ATPase) inhibitors Bafilomycin A1 or Concanamycin A at the 32-cell stage. Inhibiting the MVB-forming machinery with a dominant-negative point mutation in Vacuolar Protein Sorting 4 (Vps4-EQ) interfered with the endogenous dorsal axis. The Wnt-like activity of the dorsal cytoplasmic determinant Huluwa (Hwa), and that of microinjected xWnt8 messenger RNA, also required lysosome acidification and the MVB-forming machinery. We conclude that lysosome function is required for early dorsal axis development in Xenopus. The results highlight the intertwining between membrane trafficking, lysosomes, and vertebrate axis formation.
Collapse
|
34
|
Zhang W, Bai J, Hang K, Xu J, Zhou C, Li L, Wang Z, Wang Y, Wang K, Xue D. Role of Lysosomal Acidification Dysfunction in Mesenchymal Stem Cell Senescence. Front Cell Dev Biol 2022; 10:817877. [PMID: 35198560 PMCID: PMC8858834 DOI: 10.3389/fcell.2022.817877] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/14/2022] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation has been widely used as a potential treatment for a variety of diseases. However, the contradiction between the low survival rate of transplanted cells and the beneficial therapeutic effects has affected its clinical use. Lysosomes as organelles at the center of cellular recycling and metabolic signaling, play essential roles in MSC homeostasis. In the first part of this review, we summarize the role of lysosomal acidification dysfunction in MSC senescence. In the second part, we summarize some of the potential strategies targeting lysosomal proteins to enhance the therapeutic effect of MSCs.
Collapse
Affiliation(s)
- Weijun Zhang
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jinwu Bai
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kai Hang
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianxiang Xu
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chengwei Zhou
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lijun Li
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhongxiang Wang
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yibo Wang
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Kanbin Wang
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
| | - Deting Xue
- Department of Orthopaedics, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Institute of Orthopaedics, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Deting Xue,
| |
Collapse
|
35
|
Yan Y, Ding X, Han C, Gao J, Liu Z, Liu Y, Wang K. Involvement of TMEM16A/ANO1 upregulation in the oncogenesis of colorectal cancer. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166370. [PMID: 35231545 DOI: 10.1016/j.bbadis.2022.166370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 12/11/2022]
|
36
|
Zhu Y, Gu L, Lin X, Zhang J, Tang Y, Zhou X, Lu B, Lin X, Liu C, Prochownik EV, Li Y. Ceramide-mediated gut dysbiosis enhances cholesterol esterification and promotes colorectal tumorigenesis in mice. JCI Insight 2021; 7:150607. [PMID: 34914638 PMCID: PMC8855812 DOI: 10.1172/jci.insight.150607] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 12/15/2021] [Indexed: 11/18/2022] Open
Abstract
Colorectal cancer (CRC) severely threatens human health and life span. An effective therapeutic strategy has not been established because we do not clearly know its pathogenesis. Here, we report that ceramide and sterol O-acyltransferase 1 (SOAT1) have roles in both spontaneous and chemical-induced intestinal cancers. We first found that miRNA-148a deficiency dramatically increased mouse gut dysbiosis through upregulating ceramide synthase 5 (Cers5) expression, which promoted ceramide synthesis afterward. The newly generated ceramide further promoted both azoxymethane/dextran sodium sulfate–induced (AOM/DSS-induced) and ApcMin/+ spontaneous intestinal tumorigenesis via increasing mouse gut dysbiosis. Meanwhile, increased level of ceramide correlated with the significant enhancements of both β-catenin activity and colorectal tumorigenesis in a TLR4-dependent fashion. Next, we found a direct binding of β-catenin to SOAT1 promoter to activate transcriptional expression of SOAT1, which further induced cholesterol esterification and colorectal tumorigenesis. In human patients with CRC, the same CERS5/TLR4/β-catenin/SOAT1 axis was also found to be dysregulated. Finally, the SOAT1 inhibitor (avasimibe) showed significant levels of therapeutic effects on both AOM/DSS-induced and ApcMin/+ spontaneous intestinal cancer. Our study clarified that ceramide promoted CRC development through increasing gut dysbiosis, further resulting in the increase of cholesterol esterification in a SOAT1-dependent way. Treatment with avasimibe to specifically decrease cholesterol esterification could be considered as a clinical strategy for effective CRC therapy in a future study.
Collapse
Affiliation(s)
- Yahui Zhu
- Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Li Gu
- Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xi Lin
- Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Jinmiao Zhang
- Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yi Tang
- Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xinyi Zhou
- Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Bingjun Lu
- Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xingrong Lin
- Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Cheng Liu
- Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Edward V Prochownik
- Division of Hematology/Oncology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, United States of America
| | - Youjun Li
- College of Life Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
37
|
ATP1A1 Mutant in Aldosterone-Producing Adenoma Leads to Cell Proliferation. Int J Mol Sci 2021; 22:ijms222010981. [PMID: 34681640 PMCID: PMC8537586 DOI: 10.3390/ijms222010981] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/04/2021] [Accepted: 10/09/2021] [Indexed: 12/14/2022] Open
Abstract
The molecular mechanisms by which ATP1A1 mutation-mediated cell proliferation or tumorigenesis in aldosterone-producing adenomas (APAs) have not been elucidated. First, we investigated whether the APA-associated ATP1A1 L104R mutation stimulated cell proliferation. Second, we aimed to clarify the molecular mechanisms by which the ATP1A1 mutation-mediated cell proliferated. We performed transcriptome analysis in APAs with ATP1A1 mutation. ATP1A1 L104R mutation were modulated in human adrenocortical carcinoma (HAC15) cells (ATP1A1-mutant cells), and we evaluated cell proliferation and molecular signaling events. Transcriptome and immunohistochemical analysis showed that Na/K-ATPase (NKA) expressions in ATP1A1 mutated APA were more abundant than those in non-functioning adrenocortical adenoma or KCNJ5 mutated APAs. The significant increase of number of cells, amount of DNA and S-phase population were shown in ATP1A1-mutant cells. Fluo-4 in ATP1A1-mutant cells were significantly increased. Low concentration of ouabain stimulated cell proliferation in ATP1A1-mutant cells. ATP1A1-mutant cells induced Src phosphorylation, and low concentration of ouabain supplementation showed further Src phosphorylation. We demonstrated that NKAs were highly expressed in ATP1A1 mutant APA, and the mutant stimulated cell proliferation and Src phosphorylation in ATP1A1-mutant cells. NKA stimulations would be a risk factor for the progression and development to an ATP1A1 mutant APA.
Collapse
|
38
|
Zhang H, Lin M, Dong C, Tang Y, An L, Ju J, Wen F, Chen F, Wang M, Wang W, Chen M, Zhao Y, Li J, Hou SX, Lin X, Hu L, Bu W, Wu D, Li L, Jiao S, Zhou Z. An MST4-pβ-Catenin Thr40 Signaling Axis Controls Intestinal Stem Cell and Tumorigenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004850. [PMID: 34240584 PMCID: PMC8425901 DOI: 10.1002/advs.202004850] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 05/01/2021] [Indexed: 06/04/2023]
Abstract
Elevated Wnt/β-catenin signaling has been commonly associated with tumorigenesis especially colorectal cancer (CRC). Here, an MST4-pβ-cateninThr40 signaling axis essential for intestinal stem cell (ISC) homeostasis and CRC development is uncovered. In response to Wnt3a stimulation, the kinase MST4 directly phosphorylates β-catenin at Thr40 to block its Ser33 phosphorylation by GSK3β. Thus, MST4 mediates an active process that prevents β-catenin from binding to and being degraded by β-TrCP, leading to accumulation and full activation of β-catenin. Depletion of MST4 causes loss of ISCs and inhibits CRC growth. Mice bearing either MST4T178E mutation with constitutive kinase activity or β-cateninT40D mutation mimicking MST4-mediated phosphorylation show overly increased ISCs/CSCs and exacerbates CRC. Furthermore, the MST4-pβ-cateninThr40 axis is upregulated and correlated with poor prognosis of human CRC. Collectively, this work establishes a previously undefined machinery for β-catenin activation, and further reveals its function in stem cell and tumor biology, opening new opportunities for targeted therapy of CRC.
Collapse
Affiliation(s)
- Hui Zhang
- State Key Laboratory of Molecular BiologyCAS Center for Excellence in Molecular Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai200031China
- State Key Laboratory of Genetic EngineeringDepartment of Cell and Developmental BiologySchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Moubin Lin
- Department of General SurgeryYangpu HospitalTongji University School of MedicineShanghai200090China
| | - Chao Dong
- Department of the Second Medical OncologyThe 3rd Affiliated Hospital of Kunming Medical UniversityYunnan Tumor HospitalKunming650118China
| | - Yang Tang
- Department of Medical UltrasoundTongji University Cancer CenterShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Liwei An
- Department of Medical UltrasoundTongji University Cancer CenterShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Junyi Ju
- Department of Medical UltrasoundTongji University Cancer CenterShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200072China
| | - Fuping Wen
- State Key Laboratory of Molecular BiologyCAS Center for Excellence in Molecular Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai200031China
| | - Fan Chen
- State Key Laboratory of Molecular BiologyCAS Center for Excellence in Molecular Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai200031China
| | - Meng Wang
- State Key Laboratory of Genetic EngineeringDepartment of Cell and Developmental BiologySchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Wenjia Wang
- State Key Laboratory of Genetic EngineeringDepartment of Cell and Developmental BiologySchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Min Chen
- State Key Laboratory of Molecular BiologyCAS Center for Excellence in Molecular Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai200031China
| | - Yun Zhao
- State Key Laboratory of Molecular BiologyCAS Center for Excellence in Molecular Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai200031China
| | - Jixi Li
- State Key Laboratory of Genetic EngineeringDepartment of Cell and Developmental BiologySchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Steven X. Hou
- State Key Laboratory of Genetic EngineeringDepartment of Cell and Developmental BiologySchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Xinhua Lin
- State Key Laboratory of Genetic EngineeringDepartment of Cell and Developmental BiologySchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Lulu Hu
- Fudan University Shanghai Cancer CenterInstitutes of Biomedical SciencesState Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical EpigeneticsShanghai Medical College of Fudan UniversityShanghai200032China
| | - Wenbo Bu
- Department of Materials ScienceFudan UniversityShanghai200433China
| | - Dianqing Wu
- Department of PharmacologyYale School of MedicineNew HavenCT06520USA
| | - Lin Li
- State Key Laboratory of Molecular BiologyCAS Center for Excellence in Molecular Cell ScienceShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai200031China
| | - Shi Jiao
- State Key Laboratory of Genetic EngineeringDepartment of Cell and Developmental BiologySchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| | - Zhaocai Zhou
- State Key Laboratory of Genetic EngineeringDepartment of Cell and Developmental BiologySchool of Life SciencesZhongshan HospitalFudan UniversityShanghai200438China
| |
Collapse
|
39
|
Wu MH, Padilla-Rodriguez M, Blum I, Camenisch A, Figliuolo da Paz V, Ollerton M, Muller J, Momtaz S, Mitchell SAT, Kiela P, Thorne C, Wilson JM, Cox CM. Proliferation in the developing intestine is regulated by the endosomal protein Endotubin. Dev Biol 2021; 480:50-61. [PMID: 34411593 DOI: 10.1016/j.ydbio.2021.08.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/05/2021] [Accepted: 08/14/2021] [Indexed: 11/19/2022]
Abstract
During postnatal intestinal development, the intestinal epithelium is highly proliferative, and this proliferation is regulated by signaling in the intervillous and crypt regions. This signaling is primarily mediated by Wnt, and requires membrane trafficking. However, the mechanisms by which membrane trafficking regulates signaling during this developmental phase are largely unknown. Endotubin (EDTB, MAMDC4) is an endosomal protein that is highly expressed in the apical endocytic complex (AEC) of villus enterocytes during fetal and postnatal development, and knockout of EDTB results in defective formation of the AEC and giant lysosome. Further, knockout of EDTB in cell lines results in decreased proliferation. However, the role of EDTB in proliferation during the development of the intestine is unknown. Using Villin-CreERT2 in EDTBfl/fl mice, we deleted EDTB in the intestine in the early postnatal period, or in enteroids in vitro after isolation of intervillous cells. Loss of EDTB results in decreased proliferation in the developing intestinal epithelium and decreased ability to form enteroids. EDTB is present in cells that contain the stem cell markers LGR5 and OLFM4, indicating that it is expressed in the proliferative compartment. Further, using immunoblot analysis and TCF/LEF-GFP mice as a reporter of Wnt activity, we find that knockout of EDTB results in decreased Wnt signaling. Our results show that EDTB is essential for normal proliferation during the early stages of intestinal development and suggest that this effect is through modulation of Wnt signaling.
Collapse
Affiliation(s)
- Meng-Han Wu
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| | | | - Isabella Blum
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| | - Abigail Camenisch
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| | | | | | - John Muller
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| | - Samina Momtaz
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| | - Stefanie A T Mitchell
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| | - Pawel Kiela
- Departments of Pediatrics and Immunobiology, University of Arizona, Tucson, AZ, USA; Steele Children's Research Center, University of Arizona, Tucson, AZ, USA.
| | - Curtis Thorne
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA; The University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA; Bio5 Institute, University of Arizona, Tucson, AZ, USA; Steele Children's Research Center, University of Arizona, Tucson, AZ, USA.
| | - Jean M Wilson
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA; The University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA; Bio5 Institute, University of Arizona, Tucson, AZ, USA.
| | - Christopher M Cox
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
40
|
Abstract
Mucins are high molecular-weight epithelial glycoproteins and are implicated in many physiological processes, including epithelial cell protection, signaling transduction, and tissue homeostasis. Abnormality of mucus expression and structure contributes to biological properties related to human cancer progression. Tumor growth sites induce inhospitable conditions. Many kinds of research suggest that mucins provide a microenvironment to avoid hypoxia, acidic, and other biological conditions that promote cancer progression. Given that the mucus layer captures growth factors or cytokines, we propose that mucin helps to ameliorate inhospitable conditions in tumor-growing sites. Additionally, the composition and structure of mucins enable them to mimic the surface of normal epithelial cells, allowing tumor cells to escape from immune surveillance. Indeed, human cancers such as mucinous carcinoma, show a higher incidence of invasion to adjacent organs and lymph node metastasis than do non-mucinous carcinoma. In this mini-review, we discuss how mucin provides a tumor-friendly environment and contributes to increased cancer malignancy in mucinous carcinoma.
Collapse
Affiliation(s)
- Dong-Han Wi
- Department of Life Science, Chung-Ang University, Seoul, 06974, Korea
| | - Jong-Ho Cha
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Korea
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Graduate school, Inha University, Incheon 22212, Korea
| | - Youn-Sang Jung
- Department of Life Science, Chung-Ang University, Seoul, 06974, Korea
| |
Collapse
|
41
|
Zhang D, Ma Y, Liu J, Deng Y, Zhou B, Wen Y, Li M, Wen D, Ying Y, Luo S, Shi C, Pu G, Miao Y, Zou C, Chen Y, Ma L. Metformin Alleviates Hepatic Steatosis and Insulin Resistance in a Mouse Model of High-Fat Diet-Induced Nonalcoholic Fatty Liver Disease by Promoting Transcription Factor EB-Dependent Autophagy. Front Pharmacol 2021; 12:689111. [PMID: 34366846 PMCID: PMC8346235 DOI: 10.3389/fphar.2021.689111] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/13/2021] [Indexed: 12/31/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) results from an abnormal accumulation of lipids within hepatocytes, and is commonly associated with obesity, insulin resistance, and hyperlipidemia. Metformin is commonly used to treat type 2 diabetes mellitus and, in recent years, it was found to play a potential role in the amelioration of NAFLD. However, the mechanisms underlying the protective effect of metformin against NAFLD remain largely unknown. Transcription factor EB (TFEB) is a master transcriptional regulator of lysosomal biogenesis and autophagy and, when activated, is effective against disorders of lipid metabolism. However, the role of TFEB in hepatic steatosis is not well understood. In this report, we demonstrate that the activity of TFEB is reduced in the liver of mice fed a high-fat diet. Metformin treatment significantly reverses the activity of TFEB, and the protective effect of metformin against hepatic steatosis and insulin resistance is dependent on TFEB. We show that metformin-induced autophagy is regulated by TFEB, and our findings reveal that TFEB acts as a mediator, linking metformin with autophagy to reverse NAFLD, and highlight that TFEB may be a promising molecular target for the treatment of NAFLD.
Collapse
Affiliation(s)
- Dan Zhang
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Yicheng Ma
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Jianjun Liu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Research Center of Biomedical Engineering, Kunming Medical University, Kunming, China
| | - Yi Deng
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Bo Zhou
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Yu Wen
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Mingke Li
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Daiyan Wen
- Faculty of Basic Medicine, Kunming Medical University, Kunming, China
| | - Yunyan Ying
- Faculty of Basic Medicine, Kunming Medical University, Kunming, China
| | - Sufeng Luo
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Chunjing Shi
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Guangyu Pu
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Yinglei Miao
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| | - Chenggang Zou
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China
| | - Yuanli Chen
- Faculty of Basic Medicine, Kunming Medical University, Kunming, China
| | - Lanqing Ma
- The First Affiliated Hospital, Yunnan Institute of Digestive Disease, Yunnan Clinical Research Center for Digestive Diseases, Kunming Medical University, Kunming, China
| |
Collapse
|
42
|
Chatterjee A, Paul S, Bisht B, Bhattacharya S, Sivasubramaniam S, Paul MK. Advances in targeting the WNT/β-catenin signaling pathway in cancer. Drug Discov Today 2021; 27:82-101. [PMID: 34252612 DOI: 10.1016/j.drudis.2021.07.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/27/2021] [Accepted: 07/06/2021] [Indexed: 01/05/2023]
Abstract
WNT/β-catenin signaling orchestrates various physiological processes, including embryonic development, growth, tissue homeostasis, and regeneration. Abnormal WNT/β-catenin signaling is associated with various cancers and its inhibition has shown effective antitumor responses. In this review, we discuss the pathway, potential targets for the development of WNT/β-catenin inhibitors, available inhibitors, and their specific molecular interactions with the target proteins. We also discuss inhibitors that are in clinical trials and describe potential new avenues for therapeutically targeting the WNT/β-catenin pathway. Furthermore, we introduce emerging strategies, including artificial intelligence (AI)-assisted tools and technology-based actionable approaches, to translate WNT/β-catenin inhibitors to the clinic for cancer therapy.
Collapse
Affiliation(s)
- Avradip Chatterjee
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sayan Paul
- Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu 627012, India; Centre for Cardiovascular Biology and Disease, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore 560065, India
| | - Bharti Bisht
- Department of Thoracic Surgery, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Shelley Bhattacharya
- Environmental Toxicology Laboratory, Department of Zoology (Centre for Advanced Studies), Visva Bharati (A Central University), Santiniketan 731235, India
| | - Sudhakar Sivasubramaniam
- Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu 627012, India
| | - Manash K Paul
- Department of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
43
|
Abstract
Wnt signaling has multiple functions beyond the transcriptional effects of β-catenin stabilization. We review recent investigations that uncover new cell physiological effects through the regulation of Wnt receptor endocytosis, Wnt-induced stabilization of proteins (Wnt-STOP), macropinocytosis, increase in lysosomal activity, and metabolic changes. Many of these growth-promoting effects of canonical Wnt occur within minutes and are independent of new protein synthesis. A key element is the sequestration of glycogen synthase kinase 3 (GSK3) inside multivesicular bodies and lysosomes. Twenty percent of human proteins contain consecutive GSK3 phosphorylation motifs, which in the absence of Wnt can form phosphodegrons for polyubiquitination and proteasomal degradation. Wnt signaling by either the pharmacological inhibition of GSK3 or the loss of tumor-suppressor proteins, such as adenomatous polyposis coli (APC) and Axin1, increases lysosomal acidification, anabolic metabolites, and macropinocytosis, which is normally repressed by the GSK3-Axin1-APC destruction complex. The combination of these cell physiological effects drives cell growth. Expected final online publication date for the Annual Review of Cell and Developmental Biology, Volume 37 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Lauren V Albrecht
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California 90095-1662, USA;
| | - Nydia Tejeda-Muñoz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California 90095-1662, USA;
| | - Edward M De Robertis
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California 90095-1662, USA;
| |
Collapse
|
44
|
Chu A, Zirngibl RA, Manolson MF. The V-ATPase a3 Subunit: Structure, Function and Therapeutic Potential of an Essential Biomolecule in Osteoclastic Bone Resorption. Int J Mol Sci 2021; 22:ijms22136934. [PMID: 34203247 PMCID: PMC8269383 DOI: 10.3390/ijms22136934] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/29/2022] Open
Abstract
This review focuses on one of the 16 proteins composing the V-ATPase complex responsible for resorbing bone: the a3 subunit. The rationale for focusing on this biomolecule is that mutations in this one protein account for over 50% of osteopetrosis cases, highlighting its critical role in bone physiology. Despite its essential role in bone remodeling and its involvement in bone diseases, little is known about the way in which this subunit is targeted and regulated within osteoclasts. To this end, this review is broadened to include the three other mammalian paralogues (a1, a2 and a4) and the two yeast orthologs (Vph1p and Stv1p). By examining the literature on all of the paralogues/orthologs of the V-ATPase a subunit, we hope to provide insight into the molecular mechanisms and future research directions specific to a3. This review starts with an overview on bone, highlighting the role of V-ATPases in osteoclastic bone resorption. We then cover V-ATPases in other location/functions, highlighting the roles which the four mammalian a subunit paralogues might play in differential targeting and/or regulation. We review the ways in which the energy of ATP hydrolysis is converted into proton translocation, and go in depth into the diverse role of the a subunit, not only in proton translocation but also in lipid binding, cell signaling and human diseases. Finally, the therapeutic implication of targeting a3 specifically for bone diseases and cancer is discussed, with concluding remarks on future directions.
Collapse
|
45
|
Abstract
Colorectal cancer has served as a genetic and biological paradigm for the evolution of solid tumors, and these insights have illuminated early detection, risk stratification, prevention, and treatment principles. Employing the hallmarks of cancer framework, we provide a conceptual framework to understand how genetic alterations in colorectal cancer drive cancer cell biology properties and shape the heterotypic interactions across cells in the tumor microenvironment. This review details research advances pertaining to the genetics and biology of colorectal cancer, emerging concepts gleaned from immune and single-cell profiling, and critical advances and remaining knowledge gaps influencing the development of effective therapies for this cancer that remains a major public health burden.
Collapse
Affiliation(s)
- Jiexi Li
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Xingdi Ma
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Deepavali Chakravarti
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Shabnam Shalapour
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
46
|
Colozza G, Koo BK. Wnt/β-catenin signaling: Structure, assembly and endocytosis of the signalosome. Dev Growth Differ 2021; 63:199-218. [PMID: 33619734 PMCID: PMC8251975 DOI: 10.1111/dgd.12718] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022]
Abstract
Wnt/β‐catenin signaling is an ancient pathway that regulates key aspects of embryonic development, cell differentiation, proliferation, and adult stem cell homeostasis. Work from different laboratories has shed light on the molecular mechanisms underlying the Wnt pathway, including structural details of ligand–receptor interactions. One key aspect that has emerged from multiple studies is that endocytosis of the receptor complex plays a crucial role in fine‐tuning Wnt/β‐catenin signaling. Endocytosis is a key process involved in both activation as well as attenuation of Wnt signaling, but how this is regulated is still poorly understood. Importantly, recent findings show that Wnt also regulates central metabolic pathways such as the acquisition of nutrients through actin‐driven endocytic mechanisms. In this review, we propose that the Wnt pathway displays diverse characteristics that go beyond the regulation of gene expression, through a connection with the endocytic machinery.
Collapse
Affiliation(s)
- Gabriele Colozza
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - Bon-Kyoung Koo
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| |
Collapse
|
47
|
Edwards CM, Johnson RW. From Good to Bad: The Opposing Effects of PTHrP on Tumor Growth, Dormancy, and Metastasis Throughout Cancer Progression. Front Oncol 2021; 11:644303. [PMID: 33828987 PMCID: PMC8019909 DOI: 10.3389/fonc.2021.644303] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 02/22/2021] [Indexed: 11/13/2022] Open
Abstract
Parathyroid hormone related protein (PTHrP) is a multifaceted protein with several biologically active domains that regulate its many roles in normal physiology and human disease. PTHrP causes humoral hypercalcemia of malignancy (HHM) through its endocrine actions and tumor-induced bone destruction through its paracrine actions. PTHrP has more recently been investigated as a regulator of tumor dormancy owing to its roles in regulating tumor cell proliferation, apoptosis, and survival through autocrine/paracrine and intracrine signaling. Tumor expression of PTHrP in late stages of cancer progression has been shown to promote distant metastasis formation, especially in bone by promoting tumor-induced osteolysis and exit from dormancy. In contrast, PTHrP may protect against further tumor progression and improve patient survival in early disease stages. This review highlights current knowledge from preclinical and clinical studies examining the role of PTHrP in promoting tumor progression as well as skeletal and soft tissue metastasis, especially with regards to the protein as a regulator of tumor dormancy. The discussion will also provide perspectives on PTHrP as a prognostic factor and therapeutic target to inhibit tumor progression, prevent tumor recurrence, and improve patient survival.
Collapse
Affiliation(s)
- Courtney M. Edwards
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Center for Bone Biology, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Rachelle W. Johnson
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Center for Bone Biology, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
48
|
Krishna S, Raghavan S, DasGupta R, Palakodeti D. tRNA-derived fragments (tRFs): establishing their turf in post-transcriptional gene regulation. Cell Mol Life Sci 2021; 78:2607-2619. [PMID: 33388834 PMCID: PMC11073306 DOI: 10.1007/s00018-020-03720-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/02/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023]
Abstract
Transfer RNA (tRNA)-derived fragments (tRFs) are an emerging class of conserved small non-coding RNAs that play important roles in post-transcriptional gene regulation. High-throughput sequencing of multiple biological samples have identified heterogeneous species of tRFs with distinct functionalities. These small RNAs have garnered a lot of scientific attention due to their ubiquitous expression and versatility in regulating various biological processes. In this review, we highlight our current understanding of tRF biogenesis and their regulatory functions. We summarize the diverse modes of biogenesis through which tRFs are generated and discuss the mechanism through which different tRF species regulate gene expression and the biological implications. Finally, we conceptualize research areas that require focus to strengthen our understanding of the biogenesis and function of tRFs.
Collapse
Affiliation(s)
- Srikar Krishna
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
- SASTRA University, Thirumalaisamudram, Thanjavur, India
| | - Srikala Raghavan
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India.
| | - Ramanuj DasGupta
- Precision Oncology, Genome Institute of Singapore, Singapore City, Singapore.
| | | |
Collapse
|
49
|
Kim MJ, Cervantes C, Jung YS, Zhang X, Zhang J, Lee SH, Jun S, Litovchick L, Wang W, Chen J, Fang B, Park JI. PAF remodels the DREAM complex to bypass cell quiescence and promote lung tumorigenesis. Mol Cell 2021; 81:1698-1714.e6. [PMID: 33626321 PMCID: PMC8052288 DOI: 10.1016/j.molcel.2021.02.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 12/15/2020] [Accepted: 01/29/2021] [Indexed: 01/01/2023]
Abstract
The DREAM complex orchestrates cell quiescence and the cell cycle. However, how the DREAM complex is deregulated in cancer remains elusive. Here, we report that PAF (PCLAF/KIAA0101) drives cell quiescence exit to promote lung tumorigenesis by remodeling the DREAM complex. PAF is highly expressed in lung adenocarcinoma (LUAD) and is associated with poor prognosis. Importantly, Paf knockout markedly suppressed LUAD development in mouse models. PAF depletion induced LUAD cell quiescence and growth arrest. PAF is required for the global expression of cell-cycle genes controlled by the repressive DREAM complex. Mechanistically, PAF inhibits DREAM complex formation by binding to RBBP4, a core DREAM subunit, leading to transactivation of DREAM target genes. Furthermore, pharmacological mimicking of PAF-depleted transcriptomes inhibited LUAD tumor growth. Our results unveil how the PAF-remodeled DREAM complex bypasses cell quiescence to promote lung tumorigenesis and suggest that the PAF-DREAM axis may be a therapeutic vulnerability in lung cancer.
Collapse
Affiliation(s)
- Moon Jong Kim
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christopher Cervantes
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Youn-Sang Jung
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Life Science, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Xiaoshan Zhang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jie Zhang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sung Ho Lee
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sohee Jun
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Larisa Litovchick
- Department of Internal Medicine and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Junjie Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jae-Il Park
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Program in Genetics and Epigenetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
50
|
Jung YS, Stratton SA, Lee SH, Kim MJ, Jun S, Zhang J, Zheng B, Cervantes CL, Cha JH, Barton MC, Park JI. TMEM9-v-ATPase Activates Wnt/β-Catenin Signaling Via APC Lysosomal Degradation for Liver Regeneration and Tumorigenesis. Hepatology 2021; 73:776-794. [PMID: 32380568 PMCID: PMC7647947 DOI: 10.1002/hep.31305] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND AIMS How Wnt signaling is orchestrated in liver regeneration and tumorigenesis remains elusive. Recently, we identified transmembrane protein 9 (TMEM9) as a Wnt signaling amplifier. APPROACH AND RESULTS TMEM9 facilitates v-ATPase assembly for vesicular acidification and lysosomal protein degradation. TMEM9 is highly expressed in regenerating liver and hepatocellular carcinoma (HCC) cells. TMEM9 expression is enriched in the hepatocytes around the central vein and acutely induced by injury. In mice, Tmem9 knockout impairs hepatic regeneration with aberrantly increased adenomatosis polyposis coli (Apc) and reduced Wnt signaling. Mechanistically, TMEM9 down-regulates APC through lysosomal protein degradation through v-ATPase. In HCC, TMEM9 is overexpressed and necessary to maintain β-catenin hyperactivation. TMEM9-up-regulated APC binds to and inhibits nuclear translocation of β-catenin, independent of HCC-associated β-catenin mutations. Pharmacological blockade of TMEM9-v-ATPase or lysosomal degradation suppresses Wnt/β-catenin through APC stabilization and β-catenin cytosolic retention. CONCLUSIONS Our results reveal that TMEM9 hyperactivates Wnt signaling for liver regeneration and tumorigenesis through lysosomal degradation of APC.
Collapse
Affiliation(s)
- Youn-Sang Jung
- Department of Experimental Radiation OncologyDivision of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTX.,Department of Life ScienceChung-Ang UniversitySeoulSouth Korea
| | - Sabrina A Stratton
- Department of Epigenetics and Molecular CarcinogenesisThe University of Texas MD Anderson Cancer CenterHoustonTX
| | - Sung Ho Lee
- Department of Experimental Radiation OncologyDivision of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTX
| | - Moon-Jong Kim
- Department of Experimental Radiation OncologyDivision of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTX
| | - Sohee Jun
- Department of Experimental Radiation OncologyDivision of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTX
| | - Jie Zhang
- Department of Experimental Radiation OncologyDivision of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTX
| | - Biyun Zheng
- Department of Experimental Radiation OncologyDivision of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTX
| | - Christopher L Cervantes
- Department of Experimental Radiation OncologyDivision of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTX
| | - Jong-Ho Cha
- Department of Biomedical SciencesCollege of MedicineInha UniversityIncheonSouth Korea
| | - Michelle C Barton
- Department of Epigenetics and Molecular CarcinogenesisThe University of Texas MD Anderson Cancer CenterHoustonTX.,Graduate School of Biomedical SciencesThe University of Texas MD Anderson Cancer CenterHoustonTX
| | - Jae-Il Park
- Department of Experimental Radiation OncologyDivision of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTX.,Graduate School of Biomedical SciencesThe University of Texas MD Anderson Cancer CenterHoustonTX.,Program in Genetics and EpigeneticsThe University of Texas MD Anderson Cancer CenterHoustonTX
| |
Collapse
|