1
|
Perez-Stable C, de las Pozas A, Wangpaichitr M, Sha W, Wang H, Cai R, Schally AV. Growth Hormone-Releasing Hormone (GHRH) Antagonist Peptides Combined with PI3K Isoform Inhibitors Enhance Cell Death in Prostate Cancer. Cancers (Basel) 2025; 17:1643. [PMID: 40427140 PMCID: PMC12110010 DOI: 10.3390/cancers17101643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 05/05/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Antagonists of GHRH have experimental therapeutic value, but as single agents are not likely to improve clinical outcomes, especially in advanced prostate cancer resistant to androgen deprivation therapy. Our objective is to identify anti-cancer drugs that, in combination with MIA-602 or -690 GHRH antagonists, increase cell death in all types of prostate cancer. METHODS/RESULTS We identified inhibitors of PI3Kα or PI3Kβ that consistently increased cell death when combined with MIA-602/690. The PI3K family is critical in mediating upstream signals from receptors to downstream AKT/mTOR signaling pathways and has an important role in cancer progression. The results revealed that MIA-602/690 alone decreased androgen receptors and likely enhanced PI3K (negative feedback), which was then countered by the addition of PI3K inhibitors. Furthermore, the MIA-602/690 + PI3K inhibitor combination affected multiple signaling pathways, including apoptosis (anti-apoptotic Mcl-1L switching to pro-apoptotic Mcl-1S), proliferation (E2F1, cyclin A), PI3Kα/β, AKT, and ERK. Similar results were obtained with a more clinically relevant acetate salt form of MIA-602/690. The identification of PI3K as a drug target for prostate cancer is significant because PTEN (negative regulator of PI3K) loss of function occurs in 40-50% and PIK3CA mutation/amplification occurs in 60% of prostate cancer patients, leading to a poor prognosis. CONCLUSION The ability of the MIA-602/690 + PI3K inhibitor combination to alter multiple signaling pathways may weaken the activation of adaptive mechanisms resulting from each drug and improve efficacy.
Collapse
Affiliation(s)
- Carlos Perez-Stable
- Research Service, Bruce W. Carter Veterans Affairs Medical Center, Miami, FL 33125, USA; (A.d.l.P.); (M.W.); (W.S.); (R.C.); (A.V.S.)
- Geriatric Research, Education, and Clinical Center, Bruce W. Carter Veterans Affairs Medical Center, Miami, FL 33125, USA
- Division of Gerontology & Palliative Medicine, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alicia de las Pozas
- Research Service, Bruce W. Carter Veterans Affairs Medical Center, Miami, FL 33125, USA; (A.d.l.P.); (M.W.); (W.S.); (R.C.); (A.V.S.)
| | - Medhi Wangpaichitr
- Research Service, Bruce W. Carter Veterans Affairs Medical Center, Miami, FL 33125, USA; (A.d.l.P.); (M.W.); (W.S.); (R.C.); (A.V.S.)
- Geriatric Research, Education, and Clinical Center, Bruce W. Carter Veterans Affairs Medical Center, Miami, FL 33125, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Surgery, Division of Cardiothoracic Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- South Florida VA Foundation for Research and Education, Miami, FL 33125, USA
| | - Wei Sha
- Research Service, Bruce W. Carter Veterans Affairs Medical Center, Miami, FL 33125, USA; (A.d.l.P.); (M.W.); (W.S.); (R.C.); (A.V.S.)
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- South Florida VA Foundation for Research and Education, Miami, FL 33125, USA
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Haibo Wang
- Research Service, Bruce W. Carter Veterans Affairs Medical Center, Miami, FL 33125, USA; (A.d.l.P.); (M.W.); (W.S.); (R.C.); (A.V.S.)
| | - Renzhi Cai
- Research Service, Bruce W. Carter Veterans Affairs Medical Center, Miami, FL 33125, USA; (A.d.l.P.); (M.W.); (W.S.); (R.C.); (A.V.S.)
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Andrew V. Schally
- Research Service, Bruce W. Carter Veterans Affairs Medical Center, Miami, FL 33125, USA; (A.d.l.P.); (M.W.); (W.S.); (R.C.); (A.V.S.)
- Geriatric Research, Education, and Clinical Center, Bruce W. Carter Veterans Affairs Medical Center, Miami, FL 33125, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- South Florida VA Foundation for Research and Education, Miami, FL 33125, USA
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
2
|
Li X, Han M, Zhu H, Pan Y, Su C, Liu Y, Liao Z, Zhang B, Chen X. m 6A-Mediated TMCO3 Promotes Hepatocellular Carcinoma Progression by Facilitating the Membrane Translocation and Activation of AKT. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2504187. [PMID: 40285646 DOI: 10.1002/advs.202504187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Indexed: 04/29/2025]
Abstract
The transmembrane and coiled-coil domains 3 (TMCO3) are highly expressed in many tumors. However, the underlying mechanisms governing the way in which TMCO3 affects the progression of hepatocellular carcinoma (HCC) remain unclear. This study screens out the molecule TMCO3 with high N6-methyladenosine (m6A) modification level in tumor samples compared to the adjacent non-cancerous tissues of three pairs of HCC patients through Methylated RNA Immunoprecipitation Sequencing (MeRIP-seq) and RNA sequencing (RNA-seq). Subsequently, the oncogenic effect of TMCO3 in HCC is verified through in vivo and in vitro experiments. AlkB Homolog 5 (ALKBH5), an m6A demethylase of TMCO3 is then screened out. The following experiments demonstrate that TMCO3 can activate AKT directly through the Phosphatidylinositol-3-Kinase (PI3K) pathway, thus promoting the progression of HCC. Meanwhile, the phosphorylation site on TMCO3: the 85th amino acid-serine, and mutation of this site can directly impair the activity and membrane translocation of AKT is found. Finally, the carcinogenic effect of TMCO3 is further elucidated in HCC through the orthotopic treatment model and the hydrodynamic tail vein injection treatment model. The findings can provide a potential target for targeted AKT treatment in patients with HCC and verify a possible prognostic marker in HCC.
Collapse
Affiliation(s)
- Xinxin Li
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
| | - Mengzhen Han
- Department of General Surgery, Ezhou Central Hospital, Ezhou, Hubei, 436099, China
| | - He Zhu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
| | - Yonglong Pan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
| | - Chen Su
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
| | - Yachong Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
| | - Zhibin Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei, 430030, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, Hubei, 430030, China
| |
Collapse
|
3
|
Xu G, Zhang Q, Cheng R, Qu J, Li W. Survival strategies of cancer cells: the role of macropinocytosis in nutrient acquisition, metabolic reprogramming, and therapeutic targeting. Autophagy 2025; 21:693-718. [PMID: 39817564 PMCID: PMC11925119 DOI: 10.1080/15548627.2025.2452149] [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: 10/09/2024] [Revised: 12/27/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025] Open
Abstract
Macropinocytosis is a nonselective form of endocytosis that allows cancer cells to largely take up the extracellular fluid and its contents, including nutrients, growth factors, etc. We first elaborate meticulously on the process of macropinocytosis. Only by thoroughly understanding this entire process can we devise targeted strategies against it. We then focus on the central role of the MTOR (mechanistic target of rapamycin kinase) complex 1 (MTORC1) in regulating macropinocytosis, highlighting its significance as a key signaling hub where various pathways converge to control nutrient uptake and metabolic processes. The article covers a comprehensive analysis of the literature on the molecular mechanisms governing macropinocytosis, including the initiation, maturation, and recycling of macropinosomes, with an emphasis on how these processes are hijacked by cancer cells to sustain their growth. Key discussions include the potential therapeutic strategies targeting macropinocytosis, such as enhancing drug delivery via this pathway, inhibiting macropinocytosis to starve cancer cells, blocking the degradation and recycling of macropinosomes, and inducing methuosis - a form of cell death triggered by excessive macropinocytosis. Targeting macropinocytosis represents a novel and innovative approach that could significantly advance the treatment of cancers that rely on this pathway for survival. Through continuous research and innovation, we look forward to developing more effective and safer anti-cancer therapies that will bring new hope to patients.Abbreviation: AMPK: AMP-activated protein kinase; ASOs: antisense oligonucleotides; CAD: carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase; DC: dendritic cell; EGF: epidermal growth factor; EGFR: epidermal growth factor receptor; ERBB2: erb-b2 receptor tyrosine kinase 2; ESCRT: endosomal sorting complex required for transport; GAP: GTPase-activating protein; GEF: guanine nucleotide exchange factor; GRB2: growth factor receptor bound protein 2; LPP: lipopolyplex; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin kinase complex 1; MTORC2: mechanistic target of rapamycin kinase complex 2; NSCLC: non-small cell lung cancer; PADC: pancreatic ductal adenocarcinoma; PDPK1: 3-phosphoinositide dependent protein kinase 1; PI3K: phosphoinositide 3-kinase; PIK3C3: phosphatidylinositol 3-kinase catalytic subunit type 3; PtdIns(3,4,5)P3: phosphatidylinositol-(3,4,5)-trisphosphate; PtdIns(4,5)P2: phosphatidylinositol-(4,5)-bisphosphate; PTT: photothermal therapies; RAC1: Rac family small GTPase 1; RPS6: ribosomal protein S6; RPS6KB1: ribosomal protein S6 kinase B1; RTKs: receptor tyrosine kinases; SREBF: sterol regulatory element binding transcription factor; TFEB: transcription factor EB; TNBC: triple-negative breast cancer; TSC2: TSC complex subunit 2; ULK1: unc-51 like autophagy activating kinase 1; UPS: ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Guoshuai Xu
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| | - Qinghong Zhang
- Emergency Department, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Renjia Cheng
- Department of Intensive Care Medicine, The General Hospital of the Northern Theater Command of the People’s Liberation Army of China, Shenyang, Liaoning, China
| | - Jun Qu
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| | - Wenqiang Li
- Department of General Surgery, Aerospace Center Hospital, Beijing, China
| |
Collapse
|
4
|
Zeng W, Liu F, Liu Y, Zhang Z, Hu H, Ning S, Zhang H, Chen X, Liao Z, Zhang Z. Targeting TM4SF1 promotes tumor senescence enhancing CD8+ T cell cytotoxic function in hepatocellular carcinoma. Clin Mol Hepatol 2025; 31:489-508. [PMID: 39736265 PMCID: PMC12016601 DOI: 10.3350/cmh.2024.0934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/18/2024] [Accepted: 12/26/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND/AIMS Transmembrane 4 L six family member 1 (TM4SF1) is highly expressed and contributes to the progression of various malignancies. However, how it modulates hepatocellular carcinoma (HCC) progression and senescence remains to be elucidated. METHODS TM4SF1 expression in HCC samples was evaluated using immunohistochemistry and flow cytometry. Cellular senescence was assessed through SA-β-gal activity assays and Western blot analysis. TM4SF1-related protein interactions were investigated using immunoprecipitation-mass spectrometry, co-immunoprecipitation, bimolecular fluorescence complementation, and immunofluorescence. Tumor-infiltrating immune cells were analyzed by flow cytometry. The HCC mouse model was established via hydrodynamic tail vein injection. RESULTS TM4SF1 was highly expressed in human HCC samples and murine models. Knockdown of TM4SF1 suppressed HCC proliferation both in vitro and in vivo, inducing non-secretory senescence through upregulation of p16 and p21. TM4SF1 enhanced the interaction between AKT1 and PDPK1, thereby promoting AKT phosphorylation, which subsequently downregulated p16 and p21. Meanwhile, TM4SF1-mediated AKT phosphorylation enhanced PD-L1 expression while reducing major histocompatibility complex class I level on tumor cells, leading to impaired cytotoxic function of CD8+ T cells and an increased proportion of exhausted CD8+ T cells. In clinical HCC samples, elevated TM4SF1 expression was associated with resistance to anti-PD-1 immunotherapy. Targeting TM4SF1 via adeno-associated virus induced tumor senescence, reduced tumor burden and synergistically enhanced the efficacy of anti-PD-1 therapy. CONCLUSION Our results revealed that TM4SF1 regulated tumor cell senescence and immune evasion through the AKT pathway, highlighting its potential as a therapeutic target in HCC, particularly in combination with first-line immunotherapy.
Collapse
Affiliation(s)
- Weifeng Zeng
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Furong Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Yachong Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Ze Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Haofan Hu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Shangwu Ning
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Hongwei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Zhibin Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Zhanguo Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
5
|
Tang Q, Li J, Zhang L, Zeng S, Bao Q, Hu W, He L, Huang G, Wang L, Liu Y, Zhao X, Yang S, Hu C. Orlistat facilitates immunotherapy via AKT-FOXO3a-FOXM1-mediated PD-L1 suppression. J Immunother Cancer 2025; 13:e008923. [PMID: 40139835 PMCID: PMC11951015 DOI: 10.1136/jitc-2024-008923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 12/02/2024] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND The immunotherapy targeting cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and programmed cell death ligand-1 (PD-L1) has achieved significant breakthroughs, but further improvements are still needed in cancer treatment. METHODS We investigated orlistat, a drug approved by the Food and Drug Administration for the treatment of obesity and found that it can enhance the efficacy of CTLA-4 blockade immunotherapy. We conducted both in vivo and in vitro experiments to explore the mechanism by which orlistat increased antitumor immunity. RESULTS Orlistat enhances the efficacy of anti-CTLA-4 immunotherapy by suppressing tumor cell PD-L1 protein expression and boosting the transcription of interferon-stimulated genes (ISGs) and MHC-I. Mechanistically, orlistat inhibits AKT activity and subsequent phosphorylation of forkhead box O3a (FOXO3a) at its threonine (T) 32, serine (S) 253, thereby downregulating Forkhead box M1 (FOXM1) expression, which ultimately suppresses PD-L1 transcription. Specifically, inhibition of FOXM1 leads to FOXO3a accumulation through impaired AKT activity. FOXM1 activates protein kinase B (AKT) via acting as a scaffold to facilitate 3-phosphoinositide-dependent protein kinase 1 (PDK1) and AKT and interaction. In addition, orlistat enhances phosphorylated signal transducer and activator of transcription 1 (p-STAT1) at tyrosine (Y) 701, resulting in upregulation of ISGs and MHC-I. CONCLUSIONS Orlistat plays a crucial role in modulating the immune response and supporting the combination with CTLA-4 blockade to promote antitumor immunotherapy.
Collapse
Affiliation(s)
- Qingyun Tang
- Department of Gastroenterology, Army Medical University Xinqiao Hospital, Chongqing, China
| | - Jie Li
- Department of Gastroenterology, Army Medical University Xinqiao Hospital, Chongqing, China
| | - Lianhua Zhang
- Department of Gastroenterology, Army Medical University Xinqiao Hospital, Chongqing, China
| | - Shuo Zeng
- Department of Gastroenterology, Army Medical University Xinqiao Hospital, Chongqing, China
| | - Qiyu Bao
- Department of Gastroenterology, Army Medical University Xinqiao Hospital, Chongqing, China
| | - Weichao Hu
- Department of Gastroenterology, Army Medical University Xinqiao Hospital, Chongqing, China
| | - Lijiao He
- Department of Gastroenterology, Army Medical University Xinqiao Hospital, Chongqing, China
| | - Guiping Huang
- Department of Gastroenterology, Army Medical University Xinqiao Hospital, Chongqing, China
| | - Liting Wang
- Army Military Medical University, Chongqing, China
| | - Yunyi Liu
- Department of Gastroenterology, Army Medical University Xinqiao Hospital, Chongqing, China
| | - Xiaoyan Zhao
- Department of Gastroenterology, Army Medical University Xinqiao Hospital, Chongqing, China
| | - Shiming Yang
- Department of Gastroenterology, Army Medical University Xinqiao Hospital, Chongqing, China
| | - Changjiang Hu
- Department of Gastroenterology, Army Medical University Xinqiao Hospital, Chongqing, China
| |
Collapse
|
6
|
Turewicz M, Skagen C, Hartwig S, Majda S, Thedinga K, Herwig R, Binsch C, Altenhofen D, Ouwens DM, Förster PM, Wachtmeister T, Köhrer K, Stermann T, Chadt A, Lehr S, Marschall T, Thoresen GH, Al-Hasani H. Temporal phosphoproteomics reveals circuitry of phased propagation in insulin signaling. Nat Commun 2025; 16:1570. [PMID: 39939313 PMCID: PMC11821911 DOI: 10.1038/s41467-025-56335-6] [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: 10/05/2023] [Accepted: 01/16/2025] [Indexed: 02/14/2025] Open
Abstract
Insulin is a pleiotropic hormone that elicits its metabolic and mitogenic actions through numerous rapid and reversible protein phosphorylations. The temporal regulation of insulin's intracellular signaling cascade is highly complex and insufficiently understood. We conduct a time-resolved analysis of the global insulin-regulated phosphoproteome of differentiated human primary myotubes derived from satellite cells of healthy donors using high-resolution mass spectrometry. Identification and tracking of ~13,000 phosphopeptides over time reveal a highly complex and coordinated network of transient phosphorylation and dephosphorylation events that can be allocated to time-phased regulation of distinct and non-overlapping subcellular pathways. Advanced network analysis combining protein-protein-interaction (PPI) resources and investigation of donor variability in relative phosphosite occupancy over time identifies novel putative candidates in non-canonical insulin signaling and key regulatory nodes that are likely essential for signal propagation. Lastly, we find that insulin-regulated phosphorylation of the pre-catalytic spliceosome complex is associated with acute alternative splicing events in the transcriptome of human skeletal muscle. Our findings highlight the temporal relevance of protein phosphorylations and suggest that synchronized contributions of multiple signaling pathways form part of the circuitry for propagating information to insulin effector sites.
Collapse
Affiliation(s)
- Michael Turewicz
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Munchen-Neuherberg, Germany
| | - Christine Skagen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Sonja Hartwig
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Munchen-Neuherberg, Germany
| | - Stephan Majda
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Munchen-Neuherberg, Germany
| | - Kristina Thedinga
- Department of Computational Molecular Biology, Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - Ralf Herwig
- Department of Computational Molecular Biology, Max-Planck-Institute for Molecular Genetics, Berlin, Germany
| | - Christian Binsch
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Munchen-Neuherberg, Germany
| | - Delsi Altenhofen
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Munchen-Neuherberg, Germany
| | - D Margriet Ouwens
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Munchen-Neuherberg, Germany
- Department of Endocrinology, Ghent University Hospital, Ghent, Belgium
| | - Pia Marlene Förster
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Munchen-Neuherberg, Germany
| | - Thorsten Wachtmeister
- Genomics and Transcriptomics Laboratory, Biologisch-Medizinisches-Forschungszentrum (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Karl Köhrer
- Genomics and Transcriptomics Laboratory, Biologisch-Medizinisches-Forschungszentrum (BMFZ), Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Torben Stermann
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Munchen-Neuherberg, Germany
| | - Alexandra Chadt
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Munchen-Neuherberg, Germany
| | - Stefan Lehr
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany
- German Center for Diabetes Research (DZD e.V.), Munchen-Neuherberg, Germany
| | - Tobias Marschall
- Institute for Medical Biometry and Bioinformatics, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Center for Digital Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - G Hege Thoresen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hadi Al-Hasani
- Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research at the Heinrich Heine University Düsseldorf, Medical Faculty, Düsseldorf, Germany.
- German Center for Diabetes Research (DZD e.V.), Munchen-Neuherberg, Germany.
| |
Collapse
|
7
|
Ghahremani PT, BaniArdalan S, Alehossein P, Parveen A, Jorjani M, Brown CM, Geldenhuys WJ, Huber JD, Ishrat T, Nasoohi S. Poststroke hyperglycemia dysregulates cap-dependent translation in neural cells. Life Sci 2025; 361:123336. [PMID: 39719167 DOI: 10.1016/j.lfs.2024.123336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/04/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
AIMS Post stroke hyperglycemia has been shown to deter functional recovery. Earlier findings have indicated the cap-dependent translation regulator 4E-BP1 is detrimentally upregulated in hyperglycemic conditions. The present study aims to test the hypothesis that hyperglycemic ischemic reperfusion injury (I/R) affects normal protein translation poststroke. METHODS Rat primary cortical neurons (PCNs) were exposed to oxygen glucose deprivation (OGD) followed by increasing glucose concentration (0, 5, 10, 25 mM) at reoxygenation. In vivo, adult rats were subjected to two hours transient distal middle cerebral artery occlusion (t-dMCAO) and hyperglycemic reperfusion. KEY FINDINGS In PCN cultures, high glucose levels impaired normal neurite growth at 24 h I/R where it drastically depressed S6 ribosomal protein phosphorylation at serine 235/236 residues in 40S ribosomal subunit. This concurred with substantial hypoxia inducible factor-1α (HIF-1α) destabilization and sustained vascular endothelial growth factor (VEGF). Our immunoblotting findings indicated HIF-1α stabilization and AMPK activation rely on glucose availability. Incremental glucose concentrations above the physiological levels, induced a shift towards 4E-BP1, eIF-4E hypo-phosphorylated forms leading to reduced eIF-4E availability and efficacy, as the key to recruit the 40S ribosomal subunit to the 5' end of mRNA. In vivo, immunostaining of t-dMCAO rat brains showed remarkable decrease in phosphorylated 4E-BP1 and particularly s6 ribosomal protein in the marginal cortical tissue of hyperglycemic compared to normoglycemic animals. SIGNIFICANCE These findings suggest a remarkable association between hyperglycemic I/R injury with dysregulated cap-dependent translation poststroke. Further loss/gain of function experiment may elucidate the potential therapeutic targets in regulation of HIF-1α/translation in hyperglycemic I/R injury.
Collapse
Affiliation(s)
| | - Soha BaniArdalan
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parsa Alehossein
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arshi Parveen
- Department of Anatomy and Neurobiology, School of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Masoumeh Jorjani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Candice M Brown
- Department of Neuroscience, School of Medicine, and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Werner J Geldenhuys
- Department of Pharmaceutical Sciences, School of Pharmacy, Morgantown, WV, United States
| | - Jason D Huber
- Department of Pharmaceutical Sciences, School of Pharmacy, Morgantown, WV, United States
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, School of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sanaz Nasoohi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Medicine, and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States; Department of Pharmaceutical Sciences, School of Pharmacy, Morgantown, WV, United States; Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
8
|
Xiang X, Shuya P, Jiamin Z, Zihan Z, Xumei Y, Jingjin L. 3-Phosphoinositide-Dependent Kinase 1 as a Therapeutic Target for Treating Diabetes. Curr Diabetes Rev 2025; 21:47-56. [PMID: 38468518 DOI: 10.2174/0115733998278669240226061329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 01/09/2024] [Accepted: 01/30/2024] [Indexed: 03/13/2024]
Abstract
The role of 3-phosphoinositide-dependent kinase 1 (PDK1) has been welldocumented in the development of diabetes. This review offers a thorough examination of its composition and associated routes, specifically focusing on insulin signaling and glucose processing. By examining the precise connection between PDK1 and diabetes, various strategies specifically targeting PDK1 were also investigated. Additionally, recent discoveries from mouse models were compiled where PDK1 was knocked out in certain tissues, which demonstrated encouraging outcomes for focused treatments despite the absence of any currently approved clinical PDK1 activators. Moreover, the dual nature of PDK1 activation was discussed, encompassing both anti-diabetic and pro-oncogenic effects. Hence, the development of a PDK1 modifier is of utmost importance, as it can activate anti-diabetic pathways while inhibiting pro-oncogenic pathways, thus aiding in the treatment of diabetes. In general, PDK1 presents a noteworthy opportunity for future therapeutic strategies in the treatment of diabetes.
Collapse
Affiliation(s)
- Xie Xiang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Childrens' Hospital of Wenzhou Medical University, Wenzhou, Zhejieng 325027, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejieng 325027, China
| | - Pan Shuya
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejieng, China
| | - Zhang Jiamin
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejieng, China
| | - Zhang Zihan
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejieng, China
| | - Yang Xumei
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Childrens' Hospital of Wenzhou Medical University, Wenzhou, Zhejieng 325027, China
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou, Zhejieng 325027, China
| | - Liu Jingjin
- Department of Cardiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| |
Collapse
|
9
|
Yao Z, Chen H. Everolimus in pituitary tumor: a review of preclinical and clinical evidence. Front Endocrinol (Lausanne) 2024; 15:1456922. [PMID: 39736867 PMCID: PMC11682973 DOI: 10.3389/fendo.2024.1456922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
Although pituitary tumors (PTs) are mostly benign, some PTs are characterized by low surgical resection rates, high recurrence rates, and poor response to conventional treatments and profoundly affect patients' quality of life. Everolimus (EVE) is the only FDA-approved mTOR inhibitor, which can be used for oral treatment. It effectively inhibits tumor cell proliferation and angiogenesis. It has been administered for various neuroendocrine tumors of the digestive tract, lungs, and pancreas. EVE not only suppresses the growth and proliferation of APT cells but also enhances their sensitivity to radiotherapy and chemotherapy. This review introduces the role of the PI3K/AKT/mTOR pathway in the development of APTs, comprehensively explores the current status of preclinical and clinical research of EVE in APTs, and discusses the blood-brain barrier permeability and safety of EVE.
Collapse
Affiliation(s)
- Zihong Yao
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
- Department of Endocrinology and Metabolism, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Hui Chen
- Department of Endocrinology and Metabolism, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
10
|
Xu L, Jang H, Nussinov R. Capturing Autoinhibited PDK1 Reveals the Linker's Regulatory Role, Informing Innovative Inhibitor Design. J Chem Inf Model 2024; 64:7709-7724. [PMID: 39348509 PMCID: PMC12101721 DOI: 10.1021/acs.jcim.4c01392] [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: 08/01/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/02/2024]
Abstract
PDK1 is crucial for PI3K/AKT/mTOR and Ras/MAPK cancer signaling. It phosphorylates AKT in a PIP3-dependent but S6K, SGK, and RSK kinases in a PIP3-independent manner. Unlike its substrates, its autoinhibited monomeric state has been unclear, likely due to its low population time, and phosphorylation in the absence of PIP3 has been puzzling too. Here, guided by experimental data, we constructed models and performed all-atom molecular dynamics simulations. In the autoinhibited PDK1 conformation that resembles autoinhibited AKT, binding of the linker between the kinase and PH domains to the PIF-binding pocket promotes the formation of the Glu130-Lys111 salt bridge and weakens the association of the kinase domain with the PH domain, shifting the population from the autoinhibited state to states accessible to the membrane and its kinase substrates. The interaction of the substrates' hydrophobic motif and the PDK1 PIF-binding pocket facilitates the release of the autoinhibition even in the absence of PIP3. Phosphorylation of the serine-rich motif within the linker further attenuates the association of the PH domain with the kinase domain. These suggest that while the monomeric autoinhibited state is relatively stable, it can readily shift to its active, catalysis-prone state to phosphorylate its diverse substrates. Our findings reveal the PDK1 activation mechanism and discover the regulatory role of PDK1's linker, which lead to two innovative linker-based inhibitor strategies: (i) locking the autoinhibited PDK1 through optimization of the interactions of AKT inhibitors with the PH domain of PDK1 and (ii) analogs (small molecules or peptidomimetics) that mimic the linker interactions with the PIF-binding pocket.
Collapse
Affiliation(s)
- Liang Xu
- Computational
Structural Biology Section, Frederick National Laboratory for Cancer
Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, Maryland21702, United States
| | - Hyunbum Jang
- Computational
Structural Biology Section, Frederick National Laboratory for Cancer
Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, Maryland21702, United States
| | - Ruth Nussinov
- Computational
Structural Biology Section, Frederick National Laboratory for Cancer
Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, Maryland21702, United States
- Department
of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv69978, Israel
| |
Collapse
|
11
|
Frederick MI, Nassef MZ, Borrelli MJ, Kuang S, Buensuceso A, More T, Cordes T, O'Donoghue P, Shepherd TG, Hiller K, Heinemann IU. Metabolic adaptation in epithelial ovarian cancer metastasis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167312. [PMID: 38901649 DOI: 10.1016/j.bbadis.2024.167312] [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: 01/16/2024] [Revised: 05/30/2024] [Accepted: 06/13/2024] [Indexed: 06/22/2024]
Abstract
Epithelial ovarian cancer (EOC) is highly lethal due to its unique metastatic characteristics. EOC spheroids enter a non-proliferative state, with hypoxic cores and reduced oncogenic signaling, all of which contribute to tumour dormancy during metastasis. We investigated the metabolomic states of EOC cells progressing through the three steps to metastasis. Metabolomes of adherent, spheroid, and re-adherent cells were validated by isotopic metabolic flux analysis and mitochondrial functional assays to identify metabolic pathways that were previously unknown to promote EOC metastasis. Although spheroids were thought to exist in a dormant state, metabolomic analysis revealed an unexpected upregulation of energy production pathways in spheroids, accompanied by increased abundance of tricarboxylic acid (TCA) cycle and electron transport chain proteins. Tracing of 13C-labelled glucose and glutamine showed increased pyruvate carboxylation and decreased glutamine anaplerosis in spheroids. Increased reductive carboxylation suggests spheroids adjust redox homeostasis by shuttling cytosolic NADPH into mitochondria via isocitrate dehydrogenase. Indeed, we observed spheroids have increased respiratory capacity and mitochondrial ATP production. Relative to adherent cells, spheroids reduced serine consumption and metabolism, processes which were reversed upon spheroid re-adherence. The data reveal a distinct metabolism in EOC spheroids that enhances energy production by the mitochondria while maintaining a dormant state with respect to growth and proliferation. The findings advance our understanding of EOC metastasis and identify the TCA cycle and mitochondrional activity as novel targets to disrupt EOC metastasis, providing new approaches to treat advanced disease.
Collapse
Affiliation(s)
- Mallory I Frederick
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Mohamed Z Nassef
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Matthew J Borrelli
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Siyun Kuang
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Adrian Buensuceso
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Tushar More
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Thekla Cordes
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Patrick O'Donoghue
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; Department of Chemistry, Western University, London, ON N6A 5C1, Canada
| | - Trevor G Shepherd
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada; Department of Obstetrics & Gynaecology, Western University, London, ON N6A 5C1, Canada; London Regional Cancer Program, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany.
| | - Ilka U Heinemann
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada.
| |
Collapse
|
12
|
Pan J, Yao Q, Wang Y, Chang S, Li C, Wu Y, Shen J, Yang R. The role of PI3K signaling pathway in Alzheimer's disease. Front Aging Neurosci 2024; 16:1459025. [PMID: 39399315 PMCID: PMC11466886 DOI: 10.3389/fnagi.2024.1459025] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
Alzheimer's disease (AD) is a debilitating progressively neurodegenerative disease. The best-characterized hallmark of AD, which is marked by behavioral alterations and cognitive deficits, is the aggregation of deposition of amyloid-beta (Aβ) and hyper-phosphorylated microtubule-associated protein Tau. Despite decades of experimental progress, the control rate of AD remains poor, and more precise deciphering is needed for potential therapeutic targets and signaling pathways involved. In recent years, phosphoinositide 3-kinase (PI3K) and Akt have been recognized for their role in the neuroprotective effect of various agents, and glycogen synthase kinase 3 (GSK3), a downstream enzyme, is also crucial in the tau phosphorylation and Aβ deposition. An overview of the function of PI3K/Akt pathway in the pathophysiology of AD is provided in this review, along with a discussion of recent developments in the pharmaceuticals and herbal remedies that target the PI3K/Akt signaling pathway. In conclusion, despite the challenges and hurdles, cumulative findings of novel targets and agents in the PI3K/Akt signaling axis are expected to hold promise for advancing AD prevention and treatment.
Collapse
Affiliation(s)
- Jingying Pan
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Qi Yao
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yankai Wang
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Suyan Chang
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Chenlong Li
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yongjiang Wu
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| | - Jianhong Shen
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Riyun Yang
- Department of Histology and Embryology, Medical School of Nantong University, Nantong, China
| |
Collapse
|
13
|
Kang N, Kim J, Kwon M, Son Y, Eo SK, Baryawno N, Kim BS, Yoon S, Oh SO, Lee D, Kim K. Blockade of mTORC1 via Rapamycin Suppresses 27-Hydroxycholestrol-Induced Inflammatory Responses. Int J Mol Sci 2024; 25:10381. [PMID: 39408711 PMCID: PMC11477202 DOI: 10.3390/ijms251910381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/15/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Atherosclerosis is characterized by the deposition and accumulation of extracellular cholesterol and inflammatory cells in the arterial blood vessel walls, and 27-hydroxycholesterol (27OHChol) is the most abundant cholesterol metabolite. 27OHChol is an oxysterol that induces immune responses, including immune cell activation and chemokine secretion, although the underlying mechanisms are not fully understood. In this study, we investigated the roles of the mechanistic target of rapamycin (mTOR) in 27HChol-induced inflammation using rapamycin. Treating monocytic cells with rapamycin effectively reduced the expression of CCL2 and CD14, which was involved with the increased immune response by 27OHChol. Rapamycin also suppressed the phosphorylation of S6 and 4EBP1, which are downstream of mTORC1. Additionally, it also alleviates the increase in differentiation markers into macrophage. These results suggest that 27OHChol induces inflammation by activating the mTORC1 signaling pathway, and rapamycin may be useful for the treatment of atherosclerosis-related inflammation involving 27OHchol.
Collapse
Affiliation(s)
- Nakyung Kang
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| | - Jaesung Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| | - Munju Kwon
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Yonghae Son
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| | - Seong-Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea;
| | - Ninib Baryawno
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institute, 17177 Stockholm, Sweden;
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Sik Yoon
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.Y.); (S.-O.O.)
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.Y.); (S.-O.O.)
| | - Dongjun Lee
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
- Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| |
Collapse
|
14
|
Hua R, Zhao K, Xu Z, Zheng Y, Wu C, Zhang L, Teng Y, Wang J, Wang M, Hu J, Chen L, Yuan D, Dong W, Cheng X, Xia Y. Stratifin-mediated activation of AKT signaling and therapeutic targetability in hepatocellular carcinoma progression. CELL INSIGHT 2024; 3:100178. [PMID: 39027058 PMCID: PMC11254524 DOI: 10.1016/j.cellin.2024.100178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 07/20/2024]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related deaths worldwide and presents a significant threat to human health. Despite its prevalence, the underlying regulatory mechanisms of HCC remain unclear. In this study, we integrated RNA-seq datasets, proteome dataset and survival analysis and unveiled Stratifin (SFN) as a potential prognostic biomarker for HCC. SFN knockdown inhibited HCC progression in cell cultures and mouse models. Conversely, ectopic expression of Sfn in primary mouse HCC model accelerated HCC progression. Mechanistically, SFN acted as an adaptor protein, activating AKT1 signaling by fostering the interaction between PDK1 and AKT1, with the R56 and R129 sites on SFN proving to be crucial for this binding. In the syngeneic implantation model, the R56A/R129A mutant of SFN inhibited Akt signaling activation and impeded HCC growth. Additionally, peptide inhibitors designed based on the binding motif of AKT1 to SFN significantly inhibited HCC progression. In summary, our findings establish that SFN promotes HCC progression by activating AKT signaling through the R56 and R129 binding sites. This discovery opens new avenues for a promising therapeutic strategy for the treatment of HCC.
Collapse
Affiliation(s)
- Rong Hua
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Kaitao Zhao
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Zaichao Xu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Yingcheng Zheng
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Chuanjian Wu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Lu Zhang
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Yan Teng
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Jingjing Wang
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Mengfei Wang
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Jiayu Hu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Lang Chen
- Department of Immunology, TaiKang Medical School, Wuhan University, Wuhan, China
| | - Detian Yuan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, Hubei, China
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Wuhan, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Xiaoming Cheng
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
- Wuhan University Center for Pathology and Molecular Diagnostics, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
| | - Yuchen Xia
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
- Pingyuan Laboratory, Henan, China
| |
Collapse
|
15
|
Li H, Sun J, Hu H, Wang Y. Transcription Factor E2F8 Activates PDK1-Mediated DNA Damage Repair to Enhance Cisplatin Resistance in Lung Adenocarcinoma. Pharmacology 2024; 109:341-356. [PMID: 38810606 DOI: 10.1159/000537819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 02/12/2024] [Indexed: 05/31/2024]
Abstract
INTRODUCTION Cisplatin (DDP) is the commonest chemo drug in lung adenocarcinoma (LUAD) treatment, and DDP resistance is a significant barrier to therapeutic therapy. This study attempted to elucidate the impact of PDK1 on DDP resistance in LUAD and its mechanism. METHODS Bioinformatics analysis was used to determine the expression and enriched pathways of PDK1 in LUAD tissue. Subsequently, E2F8, the upstream transcription factor of PDK1, was predicted, and the binding relationship between the two was analyzed using dual-luciferase and ChIP experiments. PDK1 and E2F8 levels in LUAD tissues and cells were detected via qRT-PCR. Cell viability, proliferation, and apoptosis levels were assayed by CCK-8, EdU, and flow cytometry experiments, respectively. Comet assay was used to assess DNA damage, and immunofluorescence was used to assess the expression of γ-H2AX. NHEJ reporter assay was to assess DNA repair efficiency. Western blot tested levels of DNA damage repair (DDR)-related proteins. Immunohistochemistry assessed the expression of relevant genes. Finally, an animal model was constructed to investigate the influence of PDK1 expression on LUAD growth. RESULTS PDK1 was found to be upregulated in LUAD and enhanced DDP resistance by mediating DDR. E2F8 was identified as an upstream transcription factor of PDK1 and was highly expressed in LUAD. Rescue experiments presented that knocking down E2F8 could weaken the promotion of PDK1 overexpression on DDR-mediated DDP resistance in LUAD. In vivo experiments showed that knocking down PDK1 plus DDP significantly reduced the growth of xenograft tumors. CONCLUSION Our results indicated that the E2F8/PDK1 axis mediated DDR to promote DDP resistance in LUAD. Our findings lead to an improved treatment strategy after drug resistance.
Collapse
Affiliation(s)
- Hongliang Li
- Department of Radiotherapy, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huaian, China
| | - Junxia Sun
- Department of Radiotherapy, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huaian, China
| | - Haibo Hu
- Department of Respiratory Diseases, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huaian, China
| | - Yi Wang
- Department of Cardiothoracic Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huaian, China
| |
Collapse
|
16
|
Varlı M, Bhosle SR, Kim E, Yang Y, Taş İ, Zhou R, Pulat S, Gamage CDB, Park SY, Ha HH, Kim H. Usnic Acid Targets 14-3-3 Proteins and Suppresses Cancer Progression by Blocking Substrate Interaction. JACS AU 2024; 4:1521-1537. [PMID: 38665668 PMCID: PMC11040559 DOI: 10.1021/jacsau.3c00774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024]
Abstract
The anticancer therapeutic effects of usnic acid (UA), a lichen secondary metabolite, have been demonstrated in vitro and in vivo. However, the mechanism underlying the anticancer effect of UA remains to be clarified. In this study, the target protein of UA was identified using a UA-linker-Affi-Gel molecule, which showed that UA binds to the 14-3-3 protein. UA binds to 14-3-3, causing the degradation of proteasomal and autophagosomal proteins. The interaction of UA with 14-3-3 isoforms modulated cell invasion, cell cycle progression, aerobic glycolysis, mitochondrial biogenesis, and the Akt/mTOR, JNK, STAT3, NF-κB, and AP-1 signaling pathways in colorectal cancer. A peptide inhibitor of 14-3-3 blocked or regressed the activity of UA and inhibited its effects. The results suggest that UA binds to 14-3-3 isoforms and suppresses cancer progression by affecting 14-3-3 targets and phosphorylated proteins.
Collapse
Affiliation(s)
- Mücahit Varlı
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Suresh R. Bhosle
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Eunae Kim
- College
of Pharmacy, Chosun University, 146 Chosundae-gil, Gwangju 61452, Republic of Korea
| | - Yi Yang
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - İsa Taş
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Rui Zhou
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Sultan Pulat
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Chathurika D. B. Gamage
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - So-Yeon Park
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Hyung-Ho Ha
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Hangun Kim
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| |
Collapse
|
17
|
Zhu X, Zhou Y, Yan S, Qian S, Wang Y, Ju E, Zhang C. Herbal Medicine-Inspired Carbon Quantum Dots with Antibiosis and Hemostasis Effects for Promoting Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2024; 16:8527-8537. [PMID: 38329426 DOI: 10.1021/acsami.3c18418] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Bleeding and bacterial infections are crucial factors affecting wound healing. The usage of herbal medicine-derived materials holds great potential for promoting wound healing. However, the uncertain intrinsic effective ingredients and unclear mechanism of action remain great concerns. Herein, inspired by the herbal medicine Ligusticum wallichii, we reported the synthesis of tetramethylpyrazine-derived carbon quantum dots (TMP-CQDs) for promoting wound healing. Of note, the use of TMP as the precursor instead of L. wallichii ensured the repeatability and homogeneity of the obtained products. Furthermore, TMP-CQDs exhibited high antibacterial activity. Mechanically, TMP-CQDs inhibited the DNA repair, biosynthesis, and quorum sensing of the bacteria and induced intracellular reactive oxygen species (ROS). Moreover, TMP-CQDs could accelerate blood coagulation through activating factor VIII and promoting platelet aggregation. Effective wound healing was achieved by using TMP-CQDs in the Staphylococcus aureus-infected mouse skin wound model. This study sheds light on the development of herbal medicine-inspired materials as effective therapeutic drugs.
Collapse
Affiliation(s)
- Xiaofei Zhu
- Department of Laboratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Yu Zhou
- College of First Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Shihai Yan
- Department of Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Shining Qian
- Department of Laboratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Yaohui Wang
- Department of Pathology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| | - Enguo Ju
- Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Chunbing Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, China
| |
Collapse
|
18
|
Gao B, Wu X, Bu L, Jiang Q, Wang L, Liu H, Zhang X, Wu Y, Li X, Li J, Liang Y, Xu L, Xie W, Guo J. Atypical inflammatory kinase IKBKE phosphorylates and inactivates FoxA1 to promote liver tumorigenesis. SCIENCE ADVANCES 2024; 10:eadk2285. [PMID: 38324694 PMCID: PMC10849599 DOI: 10.1126/sciadv.adk2285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/08/2024] [Indexed: 02/09/2024]
Abstract
Physiologically, FoxA1 plays a key role in liver differentiation and development, and pathologically exhibits an oncogenic role in prostate and breast cancers. However, its role and upstream regulation in liver tumorigenesis remain unclear. Here, we demonstrate that FoxA1 acts as a tumor suppressor in liver cancer. Using a CRISPR-based kinome screening approach, noncanonical inflammatory kinase IKBKE has been identified to inhibit FoxA1 transcriptional activity. Notably, IKBKE directly binds to and phosphorylates FoxA1 to reduce its complex formation and DNA interaction, leading to elevated hepatocellular malignancies. Nonphosphorylated mimic Foxa1 knock-in mice markedly delay liver tumorigenesis in hydrodynamic transfection murine models, while phospho-mimic Foxa1 knock-in phenocopy Foxa1 knockout mice to exhibit developmental defects and liver inflammation. Notably, Ikbke knockout delays diethylnitrosamine (DEN)-induced mouse liver tumor development. Together, our findings not only reveal FoxA1 as a bona fide substrate and negative nuclear effector of IKBKE in hepatocellular carcinioma (HCC) but also provide a promising strategy to target IKBEK for HCC therapy.
Collapse
Affiliation(s)
- Bing Gao
- Center of Hepato-Pancreate-Biliary Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Xueji Wu
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Lang Bu
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Qiwei Jiang
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Lei Wang
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Haining Liu
- Center of Hepato-Pancreate-Biliary Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Xiaomei Zhang
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Yuanzhong Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Xiaoxing Li
- Center of Hepato-Pancreate-Biliary Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Jingting Li
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Ying Liang
- Department of Nephrology, Guangzhou Eighth People′s Hospital, Guangzhou Medical University, Guangdong 510060, China
| | - Lixia Xu
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
- Department of Oncology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Wei Xie
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Jianping Guo
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| |
Collapse
|
19
|
Hu YJ, Zhang X, Lv HM, Liu Y, Li SZ. Protein O-GlcNAcylation: The sweet hub in liver metabolic flexibility from a (patho)physiological perspective. Liver Int 2024; 44:293-315. [PMID: 38110988 DOI: 10.1111/liv.15812] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/20/2023]
Abstract
O-GlcNAcylation is a dynamic, reversible and atypical O-glycosylation that regulates various cellular physiological processes via conformation, stabilisation, localisation, chaperone interaction or activity of target proteins. The O-GlcNAcylation cycle is precisely controlled by collaboration between O-GlcNAc transferase and O-GlcNAcase. Uridine-diphosphate-N-acetylglucosamine, the sole donor of O-GlcNAcylation produced by the hexosamine biosynthesis pathway, is controlled by the input of glucose, glutamine, acetyl coenzyme A and uridine triphosphate, making it a sensor of the fluctuation of molecules, making O-GlcNAcylation a pivotal nutrient sensor for the metabolism of carbohydrates, amino acids, lipids and nucleotides. O-GlcNAcylation, particularly prevalent in liver, is the core hub for controlling systemic glucose homeostasis due to its nutritional sensitivity and precise spatiotemporal regulation of insulin signal transduction. The pathology of various liver diseases has highlighted hepatic metabolic disorder and dysfunction, and abnormal O-GlcNAcylation also plays a specific pathological role in these processes. Therefore, this review describes the unique features of O-GlcNAcylation and its dynamic homeostasis maintenance. Additionally, it explains the underlying nutritional sensitivity of O-GlcNAcylation and discusses its mechanism of spatiotemporal modulation of insulin signal transduction and liver metabolic homeostasis during the fasting and feeding cycle. This review emphasises the pathophysiological implications of O-GlcNAcylation in nonalcoholic fatty liver disease, nonalcoholic steatohepatitis and hepatic fibrosis, and focuses on the adverse effects of hyper O-GlcNAcylation on liver cancer progression and metabolic reprogramming.
Collapse
Affiliation(s)
- Ya-Jie Hu
- Key Laboratory of Bovine Disease Control in Northeast China of Ministry of Agriculture and Rural affairs of the People's Republic of China, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Xu Zhang
- Key Laboratory of Bovine Disease Control in Northeast China of Ministry of Agriculture and Rural affairs of the People's Republic of China, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Hong-Ming Lv
- Key Laboratory of Bovine Disease Control in Northeast China of Ministry of Agriculture and Rural affairs of the People's Republic of China, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Yang Liu
- Key Laboratory of Bovine Disease Control in Northeast China of Ministry of Agriculture and Rural affairs of the People's Republic of China, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shi-Ze Li
- Key Laboratory of Bovine Disease Control in Northeast China of Ministry of Agriculture and Rural affairs of the People's Republic of China, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| |
Collapse
|
20
|
DA Costa Machado AK, Machado CB, DE Pinho Pessoa FMC, Barreto IV, Gadelha RB, DE Sousa Oliveira D, Ribeiro RM, Lopes GS, DE Moraesfilho MO, DE Moraes MEA, Khayat AS, Moreira-Nunes CA. Development and Clinical Applications of PI3K/AKT/mTOR Pathway Inhibitors as a Therapeutic Option for Leukemias. CANCER DIAGNOSIS & PROGNOSIS 2024; 4:9-24. [PMID: 38173664 PMCID: PMC10758851 DOI: 10.21873/cdp.10279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 11/15/2023] [Indexed: 01/05/2024]
Abstract
Leukemias are hematological neoplasms characterized by dysregulations in several cellular signaling pathways, prominently including the PI3K/AKT/mTOR pathway. Since this pathway is associated with several important cellular mechanisms, such as proliferation, metabolism, survival, and cell death, its hyperactivation significantly contributes to the development of leukemias. In addition, it is a crucial prognostic factor, often correlated with therapeutic resistance. Changes in the PI3K/AKT/mTOR pathway are identified in more than 50% of cases of acute leukemia, especially in myeloid lineages. Furthermore, these changes are highly frequent in cases of chronic lymphocytic leukemia, especially those with a B cell phenotype, due to the correlation between the hyperactivation of B cell receptors and the abnormal activation of PI3Kδ. Thus, the search for new therapies that inhibit the activity of the PI3K/AKT/mTOR pathway has become the objective of several clinical studies that aim to replace conventional oncological treatments that have high rates of toxicities and low specificity with target-specific therapies offering improved patient quality of life. In this review we describe the PI3K/AKT/mTOR signal transduction pathway and its implications in leukemogenesis. Furthermore, we provide an overview of clinical trials that employed PI3K/AKT/mTOR inhibitors either as monotherapy or in combination with other cytotoxic agents for treating patients with various types of leukemias. The varying degrees of treatment efficacy are also reported.
Collapse
Affiliation(s)
- Anna Karolyna DA Costa Machado
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Caio Bezerra Machado
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Flávia Melo Cunha DE Pinho Pessoa
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Igor Valentim Barreto
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Renan Brito Gadelha
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | | | | | | | - Manoel Odorico DE Moraesfilho
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | - Maria Elisabete Amaral DE Moraes
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
| | - André Salim Khayat
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém, PA, Brazil
| | - Caroline Aquino Moreira-Nunes
- Department of Medicine, Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, Brazil
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém, PA, Brazil
- Clementino Fraga Group, Central Unity, Molecular Biology Laboratory, Fortaleza, CE, Brazil
| |
Collapse
|
21
|
Keshava S, Owens S, Qin W, Jeffers A, Kyei P, Komatsu S, Kleam J, Ikebe M, Idell S, Tucker TA. The mTORC2/SGK1/NDRG1 Signaling Axis Is Critical for the Mesomesenchymal Transition of Pleural Mesothelial Cells and the Progression of Pleural Fibrosis. Am J Respir Cell Mol Biol 2024; 70:50-62. [PMID: 37607215 PMCID: PMC10768834 DOI: 10.1165/rcmb.2023-0131oc] [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: 04/06/2023] [Accepted: 08/22/2023] [Indexed: 08/24/2023] Open
Abstract
Progressive lung scarring because of persistent pleural organization often results in pleural fibrosis (PF). This process affects patients with complicated parapneumonic pleural effusions, empyema, and other pleural diseases prone to loculation. In PF, pleural mesothelial cells undergo mesomesenchymal transition (MesoMT) to become profibrotic, characterized by increased expression of α-smooth muscle actin and matrix proteins, including collagen-1. In our previous study, we showed that blocking PI3K/Akt signaling inhibits MesoMT induction in human pleural mesothelial cells (HPMCs) (1). However, the downstream signaling pathways leading to MesoMT induction remain obscure. Here, we investigated the role of mTOR complexes (mTORC1/2) in MesoMT induction. Our studies show that activation of the downstream mediator mTORC1/2 complex is, likewise, a critical component of MesoMT. Specific targeting of mTORC1/2 complex using pharmacological inhibitors such as INK128 and AZD8055 significantly inhibited transforming growth factor β (TGF-β)-induced MesoMT markers in HPMCs. We further identified the mTORC2/Rictor complex as the principal contributor to MesoMT progression induced by TGF-β. Knockdown of Rictor, but not Raptor, attenuated TGF-β-induced MesoMT in these cells. In these studies, we further show that concomitant activation of the SGK1/NDRG1 signaling cascade is essential for inducing MesoMT. Targeting SGK1 and NDRG1 with siRNA and small molecular inhibitors attenuated TGF-β-induced MesoMT in HPMCs. Additionally, preclinical studies in our Streptococcus pneumoniae-mediated mouse model of PF showed that inhibition of mTORC1/2 with INK128 significantly attenuated the progression of PF in subacute and chronic injury. In conclusion, our studies demonstrate that mTORC2/Rictor-mediated activation of SGK1/NDRG1 is critical for MesoMT induction and that targeting this pathway could inhibit or even reverse the progression of MesoMT and PF.
Collapse
Affiliation(s)
| | - Shuzi Owens
- Department of Cellular and Molecular Biology, and
| | - Wenyi Qin
- Department of Cellular and Molecular Biology, and
| | | | - Perpetual Kyei
- Biotechnology Graduate Program, The University of Texas Health Science Center at Tyler, Tyler, Texas
| | | | - Joshua Kleam
- Department of Cellular and Molecular Biology, and
| | - Mitsuo Ikebe
- Department of Cellular and Molecular Biology, and
| | - Steven Idell
- Texas Lung Injury Institute
- Department of Cellular and Molecular Biology, and
| | - Torry A. Tucker
- Texas Lung Injury Institute
- Department of Cellular and Molecular Biology, and
| |
Collapse
|
22
|
Jin J, He J, Li X, Ni X, Jin X. The role of ubiquitination and deubiquitination in PI3K/AKT/mTOR pathway: A potential target for cancer therapy. Gene 2023; 889:147807. [PMID: 37722609 DOI: 10.1016/j.gene.2023.147807] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
The PI3K/AKT/mTOR pathway controls key cellular processes, including proliferation and tumor progression, and abnormally high activation of this pathway is a hallmark in human cancers. The post-translational modification, such as Ubiquitination and deubiquitination, fine-tuning the protein level and the activity of members in this pathway play a pivotal role in maintaining normal physiological process. Emerging evidence show that the unbalanced ubiquitination/deubiquitination modification leads to human diseases via PI3K/AKT/mTOR pathway. Therefore, a comprehensive understanding of the ubiquitination/deubiquitination regulation of PI3K/AKT/mTOR pathway may be helpful to uncover the underlying mechanism and improve the potential treatment of cancer via targeting this pathway. Herein, we summarize the latest research progress of ubiquitination and deubiquitination of PI3K/AKT/mTOR pathway, systematically discuss the associated crosstalk between them, as well as focus the clinical transformation via targeting ubiquitination process.
Collapse
Affiliation(s)
- Jiabei Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Jian He
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Xinming Li
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Xiaoqi Ni
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China.
| |
Collapse
|
23
|
Zheng N, Wei J, Wu D, Xu Y, Guo J. Master kinase PDK1 in tumorigenesis. Biochim Biophys Acta Rev Cancer 2023; 1878:188971. [PMID: 37640147 DOI: 10.1016/j.bbcan.2023.188971] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/13/2023] [Accepted: 08/05/2023] [Indexed: 08/31/2023]
Abstract
3-phosphoinositide-dependent protein kinase 1 (PDK1) is considered as master kinase regulating AGC kinase family members such as AKT, SGK, PLK, S6K and RSK. Although autophosphorylation regulates PDK1 activity, accumulating evidence suggests that PDK1 is manipulated by many other mechanisms, including S6K-mediated phosphorylation, and the E3 ligase SPOP-mediated ubiquitination and degradation. Dysregulation of these upstream regulators or downstream signals involves in cancer development, as PDK1 regulating cell growth, metastasis, invasion, apoptosis and survival time. Meanwhile, overexpression of PDK1 is also exposed in a plethora of cancers, whereas inhibition of PDK1 reduces cell size and inhibits tumor growth and progression. More importantly, PDK1 also modulates the tumor microenvironments and markedly influences tumor immunotherapies. In summary, we comprehensively summarize the downstream signals, upstream regulators, mouse models, inhibitors, tumor microenvironment and clinical treatments for PDK1, and highlight PDK1 as a potential cancer therapeutic target.
Collapse
Affiliation(s)
- Nana Zheng
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, China
| | - Jiaqi Wei
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, China.
| | - Yang Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, China.
| | - Jianping Guo
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510275, China.
| |
Collapse
|
24
|
Leroux AE, Biondi RM. The choreography of protein kinase PDK1 and its diverse substrate dance partners. Biochem J 2023; 480:1503-1532. [PMID: 37792325 DOI: 10.1042/bcj20220396] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/24/2023] [Accepted: 08/31/2023] [Indexed: 10/05/2023]
Abstract
The protein kinase PDK1 phosphorylates at least 24 distinct substrates, all of which belong to the AGC protein kinase group. Some substrates, such as conventional PKCs, undergo phosphorylation by PDK1 during their synthesis and subsequently get activated by DAG and Calcium. On the other hand, other substrates, including members of the Akt/PKB, S6K, SGK, and RSK families, undergo phosphorylation and activation downstream of PI3-kinase signaling. This review presents two accepted molecular mechanisms that determine the precise and timely phosphorylation of different substrates by PDK1. The first mechanism involves the colocalization of PDK1 with Akt/PKB in the presence of PIP3. The second mechanism involves the regulated docking interaction between the hydrophobic motif (HM) of substrates and the PIF-pocket of PDK1. This interaction, in trans, is equivalent to the molecular mechanism that governs the activity of AGC kinases through their HMs intramolecularly. PDK1 has been instrumental in illustrating the bi-directional allosteric communication between the PIF-pocket and the ATP-binding site and the potential of the system for drug discovery. PDK1's interaction with substrates is not solely regulated by the substrates themselves. Recent research indicates that full-length PDK1 can adopt various conformations based on the positioning of the PH domain relative to the catalytic domain. These distinct conformations of full-length PDK1 can influence the interaction and phosphorylation of substrates. Finally, we critically discuss recent findings proposing that PIP3 can directly regulate the activity of PDK1, which contradicts extensive in vitro and in vivo studies conducted over the years.
Collapse
Affiliation(s)
- Alejandro E Leroux
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Ricardo M Biondi
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| |
Collapse
|
25
|
Liu L, Zhu L, Cheng Z, Sun Y, Zhou Y, Cao J. Aberrant expression of AKR1B1 indicates poor prognosis and promotes gastric cancer progression by regulating the AKT-mTOR pathway. Aging (Albany NY) 2023; 15:9661-9675. [PMID: 37751590 PMCID: PMC10564407 DOI: 10.18632/aging.205041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023]
Abstract
Gastric cancer (GC) is a common malignant tumor in the digestive tract and a major cause of global cancer death. Due to the limited access to early screening, many patients are diagnosed with advanced GC. Therefore, postoperative radiotherapy and chemotherapy possess limited efficacy in treating GC. AKR1B1 has been associated with tumorigenesis and metastasis across various tumors, becoming a potential therapeutic target for GC. However, its role and mechanism in GC remain unclear. In this study, AKR1B1 was elevated in GC tissue, depicting a poor prognosis. AKR1B1 is closely related to age, vascular and neural invasion, lymph node metastasis, and the TNM stage of GC. The developed survival prediction model suggested that AKR1B1 expression level is crucial in the prognosis of GC patients. Moreover, the expression level of AKR1B1 in GC tissues is closely associated with the AKT-mTOR pathway. In vitro and in vivo assays functional assays helped determine the oncogenic role of AKR1B1. Additionally, the knockdown of AKR1B1 expression level in GC cell lines could effectively suppress the AKT-mTOR pathway and inhibit the proliferation and migration of tumor cells. In conclusion, this study provides a theoretical basis to establish the potential association and regulatory mechanism of AKR1B1 while offering a new strategy for GC-targeted therapy.
Collapse
Affiliation(s)
- Luojie Liu
- Department of Gastroenterology, Changshu Hospital Affiliated to Soochow University, Suzhou 215501, China
| | - Lihua Zhu
- Department of Gastroenterology, Changshu Hospital Affiliated to Soochow University, Suzhou 215501, China
| | - Zhengwu Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu 241000, China
| | - Yibin Sun
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215005, China
| | - Yuqing Zhou
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215005, China
| | - Jiwei Cao
- Department of General Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215005, China
| |
Collapse
|
26
|
Wang J, Cheng Z, Dai D, Li H, Shao X, Xu M. Overexpression of TYRO3 indicates poor prognosis and induces gastric cancer progression via AKT-mTOR pathway. Mol Carcinog 2023; 62:1325-1337. [PMID: 37212497 DOI: 10.1002/mc.23566] [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: 10/22/2022] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/23/2023]
Abstract
Gastric cancer (GC) is among of the leading causes of cancer mortality worldwide. This is because many patients are diagnosed with advanced GC and postoperative radiotherapy and chemotherapy have also exhibited limited effects on GC. TYRO3 has been considered carcinogenic and a potential therapeutic target for GC. However, TYRO3 function and mechanism in GC remains elusive. The study results indicated that TYRO3 was aberrantly elevated in GC tissues and predicted poor prognosis. TYRO3 is closely associated with clinicopathological indicators in GC tissues such as lymph node metastasis, venous invasion, neural invasion, and the tumor-node-metastasis stage. In addition, TYRO3 expression levels are closely related to the AKT-mTOR pathway in GC tissues. Moreover, the oncogenic role of TYRO3 was determined through in vitro and in vivo functional assays, and knockdown of the TYRO3 expression level in GC cell lines can effectively suppress the AKT-mTOR pathway and inhibit tumor cell proliferation and migration. In conclusion, this study provides a theoretical basis for establishing the potential association and regulatory mechanism between TYRO3 and AKT-mTOR and offers a new strategy for GC-targeted therapy.
Collapse
Affiliation(s)
- Junfeng Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Zhengwu Cheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Dafei Dai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Hao Li
- Department of Gastrointestinal Surgery, Suzhou Municipal Hospital, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Xinyu Shao
- Department of Gastroenterology, Suzhou Municipal Hospital, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Menglin Xu
- Department of Oncology, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|
27
|
Zhang J, Zhang Y, Lin X, Han X, Meredith KL, Li Z. The effects of the tumor suppressor gene PTEN on the proliferation and apoptosis of breast cancer cells via AKT phosphorylation. Transl Cancer Res 2023; 12:1863-1872. [PMID: 37588750 PMCID: PMC10425639 DOI: 10.21037/tcr-23-826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/28/2023] [Indexed: 08/18/2023]
Abstract
Background The proliferation and apoptosis of cancer cells play important roles in breast carcinomas. However, to date, there have been few reports on the correlation between the expression of PTEN and AKT phosphorylation in breast cancer. This present study investigated the effects of the phosphatase and tensin homology deleted from chromosome 10 (PTEN) gene on the proliferation and apoptosis of breast cancer cells through protein kinase B (AKT) phosphorylation. Methods Human breast cancer MDA-MB-231 cells were transfected with the pcDNA3.0 control vector or the pcDNA3.0-PTEN vector for 48 hours. The Cell Counting Kit 8 (CCK-8) was used to detect cell survival rates, double staining was performed to detect apoptosis, and Western blot (WB) analysis was conducted to detect protein expression. The effects of PTEN expression on the cell cycle and apoptosis of human breast cancer cell line MDA-MB-231, and on the levels of phosphorylated AKT protein were further analyzed. Moreover, the relationship between the PTEN gene and clinical features were also analyzed. Results The cell survival rate of cells transfected with pcDNA3.0-PTEN was significantly lower than that of cells transfected with the control pcDNA3.0 vector (55.65%±12.18% vs. 97.32%±12.45%, P=0.004). Compared with the pcDNA3.0 group, the apoptosis rate of the pcDNA3.0-PTEN group was significantly increased (20.65±2.18 vs. 2.32±0.45, P=0.001). The expression of PTEN protein in pcDNA3.0-PTEN group was higher than that in the pcDNA3.0 group, and the expression of the AKT and mTOR proteins was significantly lower than that in pcDNA3.0 group (P<0.05). The expression of PTEN in the lymph node metastasis positive group was significantly higher than that in the lymph node metastasis negative group (P<0.05). The expression of the AKT protein in breast cancer was higher than that in normal breast tissue, and the difference was statistically significant (P<0.01). Conclusions Overexpression of the PTEN gene can promote AKT phosphorylation, increase the apoptotic index of breast cancer cells, and reduce the proliferative activity of breast cancer cells. This provided a new direction for the next treatment of breast cancer, but further clinical research is needed.
Collapse
Affiliation(s)
- Junhua Zhang
- Department of Breast Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Ying Zhang
- Department of Breast Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Xiaomeng Lin
- Department of Breast Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Xiaoxu Han
- Department of Breast Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | | | - Zhong Li
- Department of Breast Surgery, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
28
|
Lin XT, Zhang J, Liu ZY, Wu D, Fang L, Li CM, Yu HQ, Xie CM. Elevated FBXW10 drives hepatocellular carcinoma tumorigenesis via AR-VRK2 phosphorylation-dependent GAPDH ubiquitination in male transgenic mice. Cell Rep 2023; 42:112812. [PMID: 37450367 DOI: 10.1016/j.celrep.2023.112812] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/24/2023] [Accepted: 06/29/2023] [Indexed: 07/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC), the most common liver cancer, occurs mainly in men, but the underlying mechanism remains to be further explored. Here, we report that ubiquitinated glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is responsible for HCC tumorigenesis in males. Mechanistically, FBXW10 promotes GAPDH polyubiquitination and activation; VRK2-dependent phosphorylation of GAPDH Ser151 residue is critical for GAPDH ubiquitination and activation. Activated GAPDH interacts with TRAF2, leading to upregulation of the canonical and noncanonical NF-κB pathways, and increases PD-L1 and AR-VRK2 expression, followed by induction of immune evasion, HCC tumorigenesis, and metastasis. Notably, the GAPDH inhibitor koningic acid (KA) activates immune response and protects against FBXW10-driven HCC in vivo. In HCC clinical samples, the expression of active GAPDH is positively correlated with that of FBXW10 and VRK2. We propose that the FBXW10/AR/VRK2/GAPDH/NF-κB axis is critical for HCC tumorigenesis in males. Targeting this axis with KA is a potential therapeutic strategy for male HCC patients.
Collapse
Affiliation(s)
- Xiao-Tong Lin
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jie Zhang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ze-Yu Liu
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Di Wu
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lei Fang
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chun-Ming Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Hong-Qiang Yu
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chuan-Ming Xie
- Key Laboratory of Hepatobiliary and Pancreatic Surgery, Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
29
|
You L, Dou Y, Zhang Y, Xiao H, Lv H, Wei GH, Xu D. SDC2 Stabilization by USP14 Promotes Gastric Cancer Progression through Co-option of PDK1. Int J Biol Sci 2023; 19:3483-3498. [PMID: 37496999 PMCID: PMC10367555 DOI: 10.7150/ijbs.84331] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/26/2023] [Indexed: 07/28/2023] Open
Abstract
Gastric cancer (GC) is a common malignancy and remains the fourth-leading cause of cancer-related deaths worldwide. Oncogenic potential of SDC2 has been implicated in multiple types of cancers, yet its role and underlying molecular mechanisms in GC remain unknown. Here, we found that SDC2 was highly expressed in GC and its upregulation correlated with poor prognosis in GC patients. Depletion of SDC2 significantly suppressed the growth and invasive capability of GC cells, while overexpressing SDC2 exerts opposite effects. Combined bioinformatics and experimental analyses substantiated that overexpression of SDC2 activated the AKT signaling pathway in GC, mechanistically through the interaction between SDC2 and PDK1-PH domain, thereby facilitating PDK1 membrane translocation to promote AKT activation. Moreover, SDC2 could also function as a co-receptor for FGF2 and was profoundly involved in the FGF2-AKT signaling axis in GC. Lastly, we revealed a mechanism on the USP14-mediated stabilization of SDC2 that is likely to contribute to SDC2 upregulation in GC tissues. Furthermore, we showed that IU1, a potent USP14 inhibitor, decreased the abundance of SDC2 in GC cells. Our findings indicate that SDC2 functions as a novel GC oncogene and has potential utility as a diagnostic marker and therapeutic target for GC.
Collapse
Affiliation(s)
- Li You
- Department of Gastric Surgery, Fudan University Shanghai Cancer, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi Dou
- Department of Gastric Surgery, Fudan University Shanghai Cancer, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yu Zhang
- Department of Gastric Surgery, Fudan University Shanghai Cancer, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Hongwei Xiao
- Key Laboratory of Animal Embryo Engineering and Molecular Breeding of Hubei province, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Science, Wuhan 430064, China
| | - Hong Lv
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Gong-Hong Wei
- Department of Gastric Surgery, Fudan University Shanghai Cancer, Shanghai 200032, China
- MOE Key Laboratory of Metabolism and Molecular Medicine and Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Fudan University Shanghai Cancer Center, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Dazhi Xu
- Department of Gastric Surgery, Fudan University Shanghai Cancer, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
30
|
Marechal E, Poliard A, Henry K, Moreno M, Legrix M, Macagno N, Mondielli G, Fauquier T, Barlier A, Etchevers HC. Multiple congenital malformations arise from somatic mosaicism for constitutively active Pik3ca signaling. Front Cell Dev Biol 2022; 10:1013001. [PMID: 36353506 PMCID: PMC9637999 DOI: 10.3389/fcell.2022.1013001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022] Open
Abstract
Recurrent missense mutations of the PIK3CA oncogene are among the most frequent drivers of human cancers. These often lead to constitutive activation of its product p110α, a phosphatidylinositol 3-kinase (PI3K) catalytic subunit. In addition to causing a broad range of cancers, the H1047R mutation is also found in affected tissues of a distinct set of congenital tumors and malformations. Collectively termed PIK3CA-related disorders (PRDs), these lead to overgrowth of brain, adipose, connective and musculoskeletal tissues and/or blood and lymphatic vessel components. Vascular malformations are frequently observed in PRD, due to cell-autonomous activation of PI3K signaling within endothelial cells. These, like most muscle, connective tissue and bone, are derived from the embryonic mesoderm. However, important organ systems affected in PRDs are neuroectodermal derivatives. To further examine their development, we drove the most common post-zygotic activating mutation of Pik3ca in neural crest and related embryonic lineages. Outcomes included macrocephaly, cleft secondary palate and more subtle skull anomalies. Surprisingly, Pik3ca-mutant subpopulations of neural crest origin were also associated with widespread cephalic vascular anomalies. Mesectodermal neural crest is a major source of non-endothelial connective tissue in the head, but not the body. To examine the response of vascular connective tissues of the body to constitutive Pik3ca activity during development, we expressed the mutation by way of an Egr2 (Krox20) Cre driver. Lineage tracing led us to observe new lineages that had normally once expressed Krox20 and that may be co-opted in pathogenesis, including vascular pericytes and perimysial fibroblasts. Finally, Schwann cell precursors having transcribed either Krox20 or Sox10 and induced to express constitutively active PI3K were associated with vascular and other tumors. These murine phenotypes may aid discovery of new candidate human PRDs affecting craniofacial and vascular smooth muscle development as well as the reciprocal paracrine signaling mechanisms leading to tissue overgrowth.
Collapse
Affiliation(s)
- Elise Marechal
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Anne Poliard
- URP 2496 Orofacial Pathologies, Imagery and Biotherapies, CNRS, GDR 2031 CREST-NET, Université Paris Cité, Montrouge, France
- School of Dentistry, Université Paris Cité, Montrouge, France
| | - Kilian Henry
- School of Dentistry, Université Paris Cité, Montrouge, France
| | - Mathias Moreno
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Mathilde Legrix
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Nicolas Macagno
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Grégoire Mondielli
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Teddy Fauquier
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Anne Barlier
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
- AP-HM, MMG, MarMaRa Institute, La Conception Hospital Laboratory of Molecular Biology, Marseille, France
| | - Heather C. Etchevers
- INSERM, MMG, U1251, CNRS, GDR 2031 CREST-NET, MarMaRa Institute, Aix Marseille University, Marseille, France
- *Correspondence: Heather C. Etchevers,
| |
Collapse
|
31
|
Kotzampasi DM, Premeti K, Papafotika A, Syropoulou V, Christoforidis S, Cournia Z, Leondaritis G. The orchestrated signaling by PI3Kα and PTEN at the membrane interface. Comput Struct Biotechnol J 2022; 20:5607-5621. [PMID: 36284707 PMCID: PMC9578963 DOI: 10.1016/j.csbj.2022.10.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/03/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
The oncogene PI3Kα and the tumor suppressor PTEN represent two antagonistic enzymatic activities that regulate the interconversion of the phosphoinositide lipids PI(4,5)P2 and PI(3,4,5)P3 in membranes. As such, they are defining components of phosphoinositide-based cellular signaling and membrane trafficking pathways that regulate cell survival, growth, and proliferation, and are often deregulated in cancer. In this review, we highlight aspects of PI3Kα and PTEN interplay at the intersection of signaling and membrane trafficking. We also discuss the mechanisms of PI3Kα- and PTEN- membrane interaction and catalytic activation, which are fundamental for our understanding of the structural and allosteric implications on signaling at the membrane interface and may aid current efforts in pharmacological targeting of these proteins.
Collapse
Affiliation(s)
- Danai Maria Kotzampasi
- Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
- Department of Biology, University of Crete, Heraklion 71500, Greece
| | - Kyriaki Premeti
- Laboratory of Pharmacology, Faculty of Medicine, University of Ioannina, Ioannina 45110, Greece
| | - Alexandra Papafotika
- Laboratory of Biological Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina 45110, Greece
- Biomedical Research Institute, Foundation for Research and Technology, Ioannina 45110, Greece
| | - Vasiliki Syropoulou
- Laboratory of Pharmacology, Faculty of Medicine, University of Ioannina, Ioannina 45110, Greece
| | - Savvas Christoforidis
- Laboratory of Biological Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina 45110, Greece
- Biomedical Research Institute, Foundation for Research and Technology, Ioannina 45110, Greece
| | - Zoe Cournia
- Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| | - George Leondaritis
- Laboratory of Pharmacology, Faculty of Medicine, University of Ioannina, Ioannina 45110, Greece
- Institute of Biosciences, University Research Center of Ioannina, Ioannina 45110, Greece
| |
Collapse
|
32
|
Fumagalli S, Pende M. S6 kinase 1 at the central node of cell size and ageing. Front Cell Dev Biol 2022; 10:949196. [PMID: 36036012 PMCID: PMC9417411 DOI: 10.3389/fcell.2022.949196] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Genetic evidence in living organisms from yeast to plants and animals, including humans, unquestionably identifies the Target Of Rapamycin kinase (TOR or mTOR for mammalian/mechanistic) signal transduction pathway as a master regulator of growth through the control of cell size and cell number. Among the mTOR targets, the activation of p70 S6 kinase 1 (S6K1) is exquisitely sensitive to nutrient availability and rapamycin inhibition. Of note, in vivo analysis of mutant flies and mice reveals that S6K1 predominantly regulates cell size versus cell proliferation. Here we review the putative mechanisms of S6K1 action on cell size by considering the main functional categories of S6K1 targets: substrates involved in nucleic acid and protein synthesis, fat mass accumulation, retrograde control of insulin action, senescence program and cytoskeleton organization. We discuss how S6K1 may be involved in the observed interconnection between cell size, regenerative and ageing responses.
Collapse
Affiliation(s)
| | - Mario Pende
- *Correspondence: Stefano Fumagalli, ; Mario Pende,
| |
Collapse
|
33
|
PEBP4 Directs the Malignant Behavior of Hepatocellular Carcinoma Cells via Regulating mTORC1 and mTORC2. Int J Mol Sci 2022; 23:ijms23158798. [PMID: 35955931 PMCID: PMC9369291 DOI: 10.3390/ijms23158798] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/05/2022] [Accepted: 08/06/2022] [Indexed: 12/03/2022] Open
Abstract
Phosphatidylethanolamine binding protein 4 (PEBP4) is an understudied multifunctional small protein. Previous studies have shown that the expression of PEBP4 is increased in many cancer specimens, which correlates to cancer progression. The present study explored the mechanism by which PEBP4 regulates the growth and progression of hepatocellular carcinoma cells. Thus, we showed that knockdown of PEBP4 in MHCC97H cells, where its expression was relatively high, diminished activities of serine/threonine protein kinase B (PKB, also known as Akt), mammalian target of rapamycin complex 1(mTORC1), and mTORC2, events that were not restored by insulin-like growth factor 1 (IGF-1). Conversely, overexpression of PEBP4 in MHCC97L cells with the low endogenous level yielded opposite effects. Furthermore, physical association of PEBP4 with Akt, mTORC1, and mTORC2 was observed. Interestingly, introduction of AktS473D mutant, bypassing phosphorylation by mTORC2, rescued mTORC1 activity, but without effects on mTORC2 signaling. In contrast, the effect of PEBP4 overexpression on the activity of mTORC1 but not that of mTORC2 was suppressed by MK2206, a specific inhibitor of Akt. In conjunction, PEBP4 knockdown-engendered reduction of cell proliferation, migration and invasion was partially rescued by Akt S473D while increases in these parameters induced by overexpression of PEBP4 were completely abolished by MK2206, although the expression of epithelial mesenchymal transition (EMT) markers appeared to be fully regulated by the active mutant of Akt. Finally, knockdown of PEBP4 diminished the growth of tumor and metastasis, whereas they were enhanced by overexpression of PEBP4. Altogether, our study suggests that increased expression of PEBP4 exacerbates malignant behaviors of hepatocellular cancer cells through cooperative participation of mTORC1 and mTORC2.
Collapse
|
34
|
Beyond controlling cell size: functional analyses of S6K in tumorigenesis. Cell Death Dis 2022; 13:646. [PMID: 35879299 PMCID: PMC9314331 DOI: 10.1038/s41419-022-05081-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 01/21/2023]
Abstract
As a substrate and major effector of the mammalian target of rapamycin complex 1 (mTORC1), the biological functions of ribosomal protein S6 kinase (S6K) have been canonically assigned for cell size control by facilitating mRNA transcription, splicing, and protein synthesis. However, accumulating evidence implies that diverse stimuli and upstream regulators modulate S6K kinase activity, leading to the activation of a plethora of downstream substrates for distinct pathobiological functions. Beyond controlling cell size, S6K simultaneously plays crucial roles in directing cell apoptosis, metabolism, and feedback regulation of its upstream signals. Thus, we comprehensively summarize the emerging upstream regulators, downstream substrates, mouse models, clinical relevance, and candidate inhibitors for S6K and shed light on S6K as a potential therapeutic target for cancers.
Collapse
|
35
|
Tian W, Yuan H, Qin S, Liu W, Zhang B, Gu L, Zhou J, Deng D. Kaiso phosphorylation at threonine 606 leads to its accumulation in the cytoplasm, reducing its transcriptional repression of the tumor suppressor
CDH1
. Mol Oncol 2022; 16:3192-3209. [PMID: 35851744 PMCID: PMC9441001 DOI: 10.1002/1878-0261.13292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 06/09/2022] [Accepted: 07/18/2022] [Indexed: 11/11/2022] Open
Affiliation(s)
- Wei Tian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Cancer Etiology Peking University Cancer Hospital and Institute China
| | - Hongfan Yuan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Cancer Etiology Peking University Cancer Hospital and Institute China
| | - Sisi Qin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Cancer Etiology Peking University Cancer Hospital and Institute China
| | - Wensu Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Cancer Etiology Peking University Cancer Hospital and Institute China
| | - Baozhen Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Cancer Etiology Peking University Cancer Hospital and Institute China
| | - Liankun Gu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Cancer Etiology Peking University Cancer Hospital and Institute China
| | - Jing Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Cancer Etiology Peking University Cancer Hospital and Institute China
| | - Dajun Deng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Cancer Etiology Peking University Cancer Hospital and Institute China
| |
Collapse
|