1
|
Naseri B, Alipour S, Masoumi J, Hatami-Sadr A, Vaysi E, Hemmat N, Alizadeh N, Baradaran B. RAD001-mediated mTOR targeting in human monocyte-derived dendritic cells shifts them toward an immunogenic phenotype. Immunol Res 2024; 73:21. [PMID: 39699830 DOI: 10.1007/s12026-024-09572-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/21/2024] [Indexed: 12/20/2024]
Abstract
Dendritic cells (DCs) are essential for promoting T lymphocyte responses since they are specialist antigen-presenting cells. In order to maintain tolerance or initiate immune responses, DCs must be activated in a balanced and regulated manner via diverse signaling pathways. By using a variety of pharmacological components, we can interfere with their different signaling pathways such as the mammalian target of rapamycin (mTOR) to appropriately modulate DC activity. In the current study, we administered RAD001 to DCs to examine the impact of mTOR inhibition on both the maturation stage and the expression of inflammatory and anti-inflammatory molecules in DCs. Pure monocytes were cultivated and stimulated with GM-CSF and IL-4 to generate immature DCs, which were then treated with RAD001. The phenotype of the DCs was determined by labeling surface markers and analyzing them using flow cytometry. Afterward, real-time PCR was carried out to evaluate the expression of inflammatory and anti-inflammatory genes. The administration of RAD001 to DCs led to a significant upregulation in the gene expression of inflammatory molecules such as IL-12, IL-1β, tumor necrosis factor (TNF)-α, and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-KB). Conversely, RAD001 treatment resulted in a decrease in the gene expression of anti-inflammatory factors IL-10 and indoleamine 2,3-dioxygenase (IDO). However, the expression of differentiation and antigen presentation-related markers CD11c and human leukocyte antigens (HLA)-DR in RAD001-treated DCs was lower and higher compared to the control group that did not receive the treatment, respectively. Taken together, our findings indicated that RAD001 treatment of DCs can be a promising therapeutic approach for the generation of immunogenic DCs in order to barricade tumor growth. However, there is a need for further investigation to evaluate the impacts of mTOR inhibition by RAD001 in DCs on cellular immune responses in vitro and in vivo.
Collapse
Affiliation(s)
- Bahar Naseri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Alipour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Masoumi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Hatami-Sadr
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Edris Vaysi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Shi Y, Liu Z, Ma M, Zhao G, Zhu Y, Wang J, Yu Y, Huang X, Ye J, Li F, Wang X, Xu Q, Yin X. Platelet-Derived Growth Factor C Facilitates Malignant Behavior of Pancreatic Ductal Adenocarcinoma by Regulating SREBP1 Mediated Lipid Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407069. [PMID: 39225567 PMCID: PMC11516052 DOI: 10.1002/advs.202407069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/05/2024] [Indexed: 09/04/2024]
Abstract
Lipid metabolism reprogramming stands as a fundamental hallmark of cancer cells. Unraveling the core regulators of lipid biosynthesis holds the potential to find promising therapeutic targets in pancreatic ductal adenocarcinoma (PDAC). Here, it is demonstrated that platelet-derived growth factor C (PDGFC) orchestrated lipid metabolism, thereby facilitated the malignant progression of PDAC. Expression of PDGFC is upregulated in PDAC cohorts and is corelated with a poor prognosis. Aberrantly high expression of PDGFC promoted proliferation and metastasis of PDAC both in vitro and in vivo. Mechanistically, PDGFC accelerated the malignant progression of PDAC by upregulating fatty acid accumulation through sterol regulatory element-binding protein 1 (SREBP1), a key transcription factor in lipid metabolism. Remarkably, Betulin, an inhibitor of SREBP1, demonstrated the capability to inhibit proliferation and metastasis of PDAC cell lines, along with attenuating the process of liver metastasis in vivo. Overall, the study underscores the pivotal role of PDGFC-mediated lipid metabolism in PDAC progression, suggesting PDGFC as a potential biomarker for PDAC metastasis. Targeting PDGFC-induced lipid metabolism emerges as a promising therapeutic strategy for metastatic PDAC, with the potential to improve clinical outcomes.
Collapse
Affiliation(s)
- Yin‐Hao Shi
- Department of Pancreato‐Biliary Surgerythe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080China
| | - Zhi‐De Liu
- Department of Pancreato‐Biliary Surgerythe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080China
| | - Ming‐Jian Ma
- Department of Pancreato‐Biliary Surgerythe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080China
| | - Guang‐Yin Zhao
- Animal Experiment Center of the First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080China
| | - Ying‐Qin Zhu
- Department of Pancreato‐Biliary Surgerythe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080China
| | - Jie‐Qin Wang
- Department of Pediatric SurgeryGuangzhou Women and Children's Medical CenterGuangzhou Medical UniversityGuangzhou510623China
| | - Yang‐Yin‐Hui Yu
- Department of Pancreato‐Biliary Surgerythe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080China
| | - Xi‐Tai Huang
- Department of Pancreato‐Biliary Surgerythe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080China
| | - Jing‐Yuan Ye
- Department of Pancreato‐Biliary Surgerythe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080China
| | - Fu‐Xi Li
- Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
| | - Xi‐Yu Wang
- Department of Pancreato‐Biliary Surgerythe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080China
| | - Qiong‐Cong Xu
- Department of Pancreato‐Biliary Surgerythe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080China
| | - Xiao‐Yu Yin
- Department of Pancreato‐Biliary Surgerythe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510080China
| |
Collapse
|
3
|
Kim KQ, Nanjaraj Urs AN, Lasehinde V, Greenlaw AC, Hudson BH, Zaher HS. eIF4F complex dynamics are important for the activation of the integrated stress response. Mol Cell 2024; 84:2135-2151.e7. [PMID: 38848692 PMCID: PMC11189614 DOI: 10.1016/j.molcel.2024.04.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 12/08/2023] [Accepted: 04/19/2024] [Indexed: 06/09/2024]
Abstract
In response to stress, eukaryotes activate the integrated stress response (ISR) via phosphorylation of eIF2α to promote the translation of pro-survival effector genes, such as GCN4 in yeast. Complementing the ISR is the target of rapamycin (TOR) pathway, which regulates eIF4E function. Here, we probe translational control in the absence of eIF4E in Saccharomyces cerevisiae. Intriguingly, we find that loss of eIF4E leads to de-repression of GCN4 translation. In addition, we find that de-repression of GCN4 translation is accompanied by neither eIF2α phosphorylation nor reduction in initiator ternary complex (TC). Our data suggest that when eIF4E levels are depleted, GCN4 translation is de-repressed via a unique mechanism that may involve faster scanning by the small ribosome subunit due to increased local concentration of eIF4A. Overall, our findings suggest that relative levels of eIF4F components are key to ribosome dynamics and may play important roles in translational control of gene expression.
Collapse
Affiliation(s)
- Kyusik Q Kim
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | | | - Victor Lasehinde
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Alison C Greenlaw
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Benjamin H Hudson
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Hani S Zaher
- Department of Biology, Washington University in St. Louis, St. Louis, MO 63130, USA.
| |
Collapse
|
4
|
Chang I, Loo YL, Patel J, Nguyen JT, Kim JK, Krebsbach PH. Targeting of lysosomal-bound protein mEAK-7 for cancer therapy. Front Oncol 2024; 14:1375498. [PMID: 38532930 PMCID: PMC10963491 DOI: 10.3389/fonc.2024.1375498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/29/2024] [Indexed: 03/28/2024] Open
Abstract
mEAK-7 (mammalian EAK-7 or MTOR-associated protein, eak-7 homolog), is an evolutionarily conserved lysosomal membrane protein that is highly expressed in several cancer cells. Multiple recent studies have identified mEAK-7 as a positive activator of mTOR (mammalian/mechanistic target of rapamycin) signaling via an alternative mTOR complex, implying that mEAK-7 plays an important role in the promotion of cancer proliferation and migration. In addition, structural analyses investigating interactions between mEAK-7 and V-ATPase, a protein complex responsible for regulating pH homeostasis in cellular compartments, have suggested that mEAK-7 may contribute to V-ATPase-mediated mTORC1 activation. The C-terminal α-helix of mEAK-7 binds to the D and B subunits of the V-ATPase, creating a pincer-like grip around its B subunit. This binding undergoes partial disruption during ATP hydrolysis, potentially enabling other proteins such as mTOR to bind to the α-helix of mEAK-7. mEAK-7 also promotes chemoresistance and radiation resistance by sustaining DNA damage-mediated mTOR signaling through interactions with DNA-PKcs (DNA-dependent protein kinase catalytic subunit). Taken together, these findings indicate that mEAK-7 may be a promising therapeutic target against tumors. However, the precise molecular mechanisms and signal transduction pathways of mEAK-7 in cancer remain largely unknown, motivating the need for further investigation. Here, we summarize the current known roles of mEAK-7 in normal physiology and cancer development by reviewing the latest studies and discuss potential future developments of mEAK-7 in targeted cancer therapy.
Collapse
Affiliation(s)
- Insoon Chang
- Section of Endodontics, Division of Regenerative and Reconstructive Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yi-Ling Loo
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jay Patel
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Joe Truong Nguyen
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- Oral Immunobiology Unit, National Institutes of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Jin Koo Kim
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Paul H Krebsbach
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
5
|
Arnold BH, Sanislav O, Fisher PR, Annesley SJ. Plate-Based Assays for the Characterization of Mitochondrial and Cellular Phenotypes. Methods Mol Biol 2024; 2746:1-20. [PMID: 38070076 DOI: 10.1007/978-1-0716-3585-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
The mitochondria are essential to eukaryotic life, acting as key drivers of energy generation while also being involved in the regulation of many cellular processes including apoptosis, cell proliferation, calcium homeostasis, and metabolism. Mitochondrial diseases which disrupt these processes lead to a diverse range of pathologies and lack consistency in symptom presentation. In disease, mitochondrial activity and energy homeostasis can be adapted to cellular requirements, and studies using Dictyostelium and human lymphoblastoid cell lines have shown that such changes can be facilitated by the key cellular and energy regulators, TORC1 and AMPK. Fluorescence-based assays are increasingly utilized to measure mitochondrial and cell signalling function in mitochondrial disease research. Here, we describe a streamlined method for the simultaneous measurement of mitochondrial mass, membrane potential, and reactive oxygen species production using MitoTracker Green™ FM, MitoTracker Red™ CMXRos, and DCFH-DA probes. This protocol has been adapted for both Dictyostelium and human lymphoblastoid cell lines. We also describe a method for assessing TORC1 and AMPK activity simultaneously in lymphoblastoid cells. These techniques allow for the characterization of mitochondrial defects in a rapid and easy to implement manner.
Collapse
Affiliation(s)
- Benjamin Henry Arnold
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, VIC, Australia
| | - Oana Sanislav
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, VIC, Australia
| | - Paul Robert Fisher
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, VIC, Australia
| | - Sarah Jane Annesley
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, VIC, Australia.
| |
Collapse
|
6
|
Mane RR, Kale PP. The roles of HDAC with IMPDH and mTOR with JAK as future targets in the treatment of rheumatoid arthritis with combination therapy. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2023; 20:689-706. [PMID: 36409592 DOI: 10.1515/jcim-2022-0114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 07/19/2022] [Indexed: 06/16/2023]
Abstract
Various studies have shown that cytokines are important regulators in rheumatoid arthritis (RA). In synovial inflammation alteration of the enzyme HDAC, IMPDH enzyme, mTOR pathway, and JAK pathway increase cytokine level. These increased cytokine levels are responsible for the inflammation in RA. Inflammation is a physiological and normal reaction of the immune system against dangerous stimuli such as injury and infection. The cytokine-based approach improves the treatment of RA. To reach this goal, various researchers and scientists are working more aggressively by using a combination approach. The present review of combination therapy provides essential evidence about the possible synergistic effect of combinatorial agents. We have focused on the effects of HDAC inhibitor with IMPDH inhibitor and mTOR inhibitor with JAK inhibitor in combination for the treatment of RA. Combining various targeted strategies can be helpful for the treatment of RA.
Collapse
Affiliation(s)
- Reshma Rajendra Mane
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Pravin Popatrao Kale
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| |
Collapse
|
7
|
Soleimani M. Not all kidney cysts are created equal: a distinct renal cystogenic mechanism in tuberous sclerosis complex (TSC). Front Physiol 2023; 14:1289388. [PMID: 38028758 PMCID: PMC10663234 DOI: 10.3389/fphys.2023.1289388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Tuberous Sclerosis Complex (TSC) is an autosomal dominant genetic disease caused by mutations in either TSC1 or TSC2 genes. Approximately, two million individuals suffer from this disorder worldwide. TSC1 and TSC2 code for the proteins harmartin and tuberin, respectively, which form a complex that regulates the mechanistic target of rapamycin complex 1 (mTORC1) and prevents uncontrollable cell growth. In the kidney, TSC presents with the enlargement of benign tumors (angiomyolipomas) and cysts whose presence eventually causes kidney failure. The factors promoting cyst formation and tumor growth in TSC are poorly understood. Recent studies on kidney cysts in various mouse models of TSC, including mice with principal cell- or pericyte-specific inactivation of TSC1 or TSC2, have identified a unique cystogenic mechanism. These studies demonstrate the development of numerous cortical cysts that are predominantly comprised of hyperproliferating A-intercalated (A-IC) cells that express both TSC1 and TSC2. An analogous cellular phenotype in cystic epithelium is observed in both humans with TSC and in TSC2+/- mice, confirming a similar kidney cystogenesis mechanism in TSC. This cellular phenotype profoundly contrasts with kidney cysts found in Autosomal Dominant Polycystic Kidney Disease (ADPKD), which do not show any notable evidence of A-IC cells participating in the cyst lining or expansion. RNA sequencing (RNA-Seq) and confirmatory expression studies demonstrate robust expression of Forkhead Box I1 (FOXI1) transcription factor and its downstream targets, including apical H+-ATPase and cytoplasmic carbonic anhydrase 2 (CAII), in the cyst epithelia of Tsc1 (or Tsc2) knockout (KO) mice, but not in Polycystic Kidney Disease (Pkd1) mutant mice. Deletion of FOXI1, which is vital to H+-ATPase expression and intercalated (IC) cell viability, completely inhibited mTORC1 activation and abrogated the cyst burden in the kidneys of Tsc1 KO mice. These results unequivocally demonstrate the critical role that FOXI1 and A-IC cells, along with H+-ATPase, play in TSC kidney cystogenesis. This review article will discuss the latest research into the causes of kidney cystogenesis in TSC with a focus on possible therapeutic options for this devastating disease.
Collapse
Affiliation(s)
- Manoocher Soleimani
- Department of Medicine, New Mexico Veterans Health Care Center, Albuquerque, NM, United States
- Department of Medicine, University of New Mexico School of Medicine, Albuquerque, NM, United States
| |
Collapse
|
8
|
Zhang X, Luo Z, Li J, Lin Y, Li Y, Li W. Sestrin2 in diabetes and diabetic complications. Front Endocrinol (Lausanne) 2023; 14:1274686. [PMID: 37920252 PMCID: PMC10619741 DOI: 10.3389/fendo.2023.1274686] [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: 08/08/2023] [Accepted: 10/03/2023] [Indexed: 11/04/2023] Open
Abstract
Diabetes is a global health problem which is accompanied with multi-systemic complications. It is of great significance to elucidate the pathogenesis and to identify novel therapies of diabetes and diabetic complications. Sestrin2, a stress-inducible protein, is primarily involved in cellular responses to various stresses. It plays critical roles in regulating a series of cellular events, such as oxidative stress, mitochondrial function and endoplasmic reticulum stress. Researches investigating the correlations between Sestrin2, diabetes and diabetic complications are increasing in recent years. This review incorporates recent findings, demonstrates the diverse functions and regulating mechanisms of Sestrin2, and discusses the potential roles of Sestrin2 in the pathogenesis of diabetes and diabetic complications, hoping to highlight a promising therapeutic direction.
Collapse
Affiliation(s)
- Xiaodan Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zirui Luo
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou, China
| | - Jiahong Li
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou, China
| | - Yaxuan Lin
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou, China
| | - Yu Li
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wangen Li
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
9
|
Moslehian MS, Shabkhizan R, Asadi MR, Bazmani A, Mahdipour M, Haiaty S, Rahbarghazi R, Sakhinia E. Interaction of lncRNAs with mTOR in colorectal cancer: a systematic review. BMC Cancer 2023; 23:512. [PMID: 37280524 DOI: 10.1186/s12885-023-11008-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/25/2023] [Indexed: 06/08/2023] Open
Abstract
Colorectal cancer (CRC) is the third most widespread cancer and the fourth leading lethal disease among different societies. It is thought that CRC accounts for about 10% of all newly diagnosed cancer cases with high-rate mortality. lncRNAs, belonging to non-coding RNAs, are involved in varied cell bioactivities. Emerging data have confirmed a significant alteration in lncRNA transcription under anaplastic conditions. This systematic review aimed to assess the possible influence of abnormal mTOR-associated lncRNAs in the tumorigenesis of colorectal tissue. In this study, the PRISMA guideline was utilized based on the systematic investigation of published articles from seven databases. Of the 200 entries, 24 articles met inclusion criteria and were used for subsequent analyses. Of note, 23 lncRNAs were prioritized in association with the mTOR signaling pathway with up-regulation (79.16%) and down-regulation (20.84%) trends. Based on the obtained data, mTOR can be stimulated or inhibited during CRC by the alteration of several lncRNAs. Determining the dynamic activity of mTOR and relevant signaling pathways via lncRNAs can help us progress novel molecular therapeutics and medications.
Collapse
Affiliation(s)
- Marziyeh Sadat Moslehian
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roya Shabkhizan
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Asadi
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Bazmani
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University Of Mashhad, Mashhad, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, Iran
| | - Sanya Haiaty
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ebrahim Sakhinia
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Tabriz Genetic Analysis Centre (TGAC), Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
10
|
Santibanez JF, Villar VH, Echeverria C. Current and Future Cancer Chemoprevention Strategies. Pharmaceutics 2023; 15:pharmaceutics15051543. [PMID: 37242785 DOI: 10.3390/pharmaceutics15051543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
Cancer is a leading cause of death worldwide, accounting for nearly 10 million deaths in 2020 and ranking as the second-leading cause of death in economically developed countries [...].
Collapse
Affiliation(s)
- Juan F Santibanez
- Group for Molecular Oncology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Dr. Subotica 4, POB 102, 11129 Belgrade, Serbia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago 8370993, Chile
| | - Victor H Villar
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Cesar Echeverria
- Laboratory of Molecular Biology, Nanomedicine and Genomic, Faculty of Medicine, University of Atacama, Copiapo 1532502, Chile
| |
Collapse
|
11
|
Zhu QY, He ZM, Cao WM, Li B. The role of TSC2 in breast cancer: a literature review. Front Oncol 2023; 13:1188371. [PMID: 37251941 PMCID: PMC10213421 DOI: 10.3389/fonc.2023.1188371] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/03/2023] [Indexed: 05/31/2023] Open
Abstract
TSC2 is a tumor suppressor gene as well as a disease-causing gene for autosomal dominant disorder tuberous sclerosis complex (TSC). Research has found that some tumor tissues have lower TSC2 expression levels than normal tissues. Furthermore, low expression of TSC2 is associated with poor prognosis in breast cancer. TSC2 acts as a convergence point of a complex network of signaling pathways and receives signals from the PI3K, AMPK, MAPK, and WNT pathways. It also regulates cellular metabolism and autophagy through inhibition of a mechanistic target of rapamycin complex, which are processes relevant to the progression, treatment, and prognosis of breast cancer. In-depth study of TSC2 functions provides significant guidance for clinical applications in breast cancer, including improving the treatment efficacy, overcoming drug resistance, and predicting prognosis. In this review, protein structure and biological functions of TSC2 were described and recent advances in TSC2 research in different molecular subtypes of breast cancer were summarized.
Collapse
Affiliation(s)
- Qiao-Yan Zhu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Zhe-Min He
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Wen-Ming Cao
- Department of Breast Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, China
| | - Bei Li
- Department of Geriatric, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
12
|
Dong J, Xu Q, Chen S, Lei H, Wang J, Yan S, Qian C, Wang X. Comparative Proteomic and Phospho-proteomic Analysis of Mouse Placentas Generated via In Vivo and In Vitro Fertilization. Reprod Sci 2023; 30:1143-1156. [PMID: 36280645 DOI: 10.1007/s43032-022-01109-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/13/2022] [Indexed: 10/31/2022]
Abstract
Offspring conceived by assisted reproductive technologies (ART) have increased risk of suffering from gestational complications, and placental dysfunction is related with the adverse outcomes. Studies have revealed that abnormal or adaptive changes can occur in ART placentas, but the potential reasons are not fully understood. Hereby, we tried to use proteomics and phospho-proteomics to find the underlying mechanisms responsible for the changes of ART placentas. Liquid chromatography-tandem mass spectrometry was utilized to perform proteome and phospho-proteome detection on mouse placentas. The differential expressed proteins (DEPs) or phospho-proteins (DEPPs) were analyzed based on subcellular localization, functional classification, and enrichment. Western blot was used to verify the DEPs (Afadin, ZO-1, Ace2, Agt, Slc7a5, and Slc38a10) and measure mTOR signaling activities (mTOR, Rps6, and 4Ebp1). The data showed that 161 DEPs and 304 DEPPs were found in proteome and phospho-proteome, respectively. Multiple biological processes were enriched based on those DEPs and DEPPs, and renin-angiotensin system, cell junction, and PI3K-Akt pathway were investigated. By protein expression identification, two key proteins associated with renin-angiotensin system (Ace2 and Agt) were down-regulated, and the levels of Afadin and ZO-1 (related with cell junction) as well as Slc38a10 were increased in IVF placentas. In addition, mTOR downstream activities were increased as shown by p-Rps6 and p-4Ebp1 in IVF placentas. In conclusion, IVF leads to the changes of cell junction, renin-angiotensin system, amino acid transport, and increased mTOR signaling in mouse placentas, which may be associated with the altered structure and function of IVF placentas.
Collapse
Affiliation(s)
- Jie Dong
- Department of Obstetrics and Gynaecology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Qian Xu
- Department of Obstetrics and Gynaecology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Shuqiang Chen
- Department of Obstetrics and Gynaecology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Hui Lei
- Department of Obstetrics and Gynaecology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Jingjing Wang
- Department of Obstetrics and Gynaecology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Song Yan
- Department of Obstetrics and Gynaecology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Chenxi Qian
- Department of Obstetrics and Gynaecology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Xiaohong Wang
- Department of Obstetrics and Gynaecology, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, Shaanxi Province, China.
| |
Collapse
|
13
|
Huang Y. Targeting glycolysis for cancer therapy using drug delivery systems. J Control Release 2023; 353:650-662. [PMID: 36493949 DOI: 10.1016/j.jconrel.2022.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 12/03/2022] [Indexed: 12/15/2022]
Abstract
There is close crosstalk between cancer metabolism and immunity. Cancer metabolism regulation is a promising therapeutic target for cancer immunotherapy. Warburg effect is characterized by abnormal glucose metabolism that includes common features of increased glucose uptake and lactate production. The aerobic glycolysis can reprogram the cancer cells and promote the formation of a suppressive immune microenvironment. As a case in point, lactate plays an essential role in tumorigenesis, which is the end product of glycolysis as well as serves as a fuel supporting cancer cell survival. Meanwhile, it is also an important immune regulator that drives immunosuppression in tumors. Immunometabolic therapy is to intervene tumor metabolism and regulate the related metabolites that participate in the innate and acquired immunity, thereby reinstalling the immune balance and eliciting anticancer immune responses. In this contribution to the Orations - New Horizons of the Journal of controlled Release I will provide an overview of glucose metabolism in tumors and its effects on drug resistance and tumor metastasis, and present the advance of glycolysis-targeting therapy strategies with drug delivery techniques, as well as discuss the challenges in glycolysis-targeting immunometabolic therapy.
Collapse
Affiliation(s)
- Yongzhuo Huang
- Zhongshan Institute for Drug Discovery, SIMM, CAS, China; Shanghai Institute of Materia Medica Chinese Academy of Science, China.
| |
Collapse
|
14
|
Watany MM, El-Horany HE, Elhosary MM, Elhadidy AA. Clinical application of RUBCN/SESN2 mediated inhibition of autophagy as biomarkers of diabetic kidney disease. Mol Med 2022; 28:147. [PMID: 36476132 PMCID: PMC9730641 DOI: 10.1186/s10020-022-00580-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Deregulated autophagy in diabetes has been a field of many experimental studies recently. Impaired autophagy in diabetic kidneys orchestrates every step of diabetic nephropathy (DN) pathogenesis. This study aimed to evaluate three autophagy regulators; RUBCN, mTOR, and SESN2 as clinically applicable indicators of DN progression and as early predictors of DN. METHODS This retrospective study included 120 participants in 4 groups; G1: diabetic patients without albuminuria, G2: diabetic patients with microalbuminuria, G3: diabetic patients with macroalbuminuria and G4: healthy controls. RUBCN and SESN2 genes expression were tested by RT-qPCR. RUBCN, mTOR, and SESN2 serum proteins were quantitated by ELISA. RESULTS RUBCN mRNA was over-expressed in diabetic patients relative to controls with the highest level found in G3 followed by G2 then G1; (9.04 ± 0.64, 5.18 ± 0.73, 1.94 ± 0.41 respectively. P < 0.001). SESN2 mRNA expression was at its lowest level in G3 followed by G2 then G1 (0.1 ± 0.06, 0.48 ± 0.11, 0.78 ± 0.13 respectively. P < 0.001). Similar parallel reduction in serum SENS2 was observed. Serum RUBCN and mTOR were significantly elevated in diabetic patients compared to controls, with the increase parallel to albuminuria degree. RUBCN expression, serum RUBCN and mTOR strongly correlated with albuminuria (r = 0.912, 0.925 and 0.867 respectively). SESN2 expression and serum level negatively correlated with albuminuria (r = - 0.897 and -0.828 respectively); (All p < 0.001). Regression analysis showed that serum RUBCN, mTOR, RUBCN and SESN2 mRNAs could successfully predict DN. CONCLUSIONS The study proves the overexpression of RUBCN and mTOR in DN and the down-expression of SESN2. The three markers can be clinically used to predict DN and to monitor disease progression.
Collapse
Affiliation(s)
- Mona M. Watany
- grid.412258.80000 0000 9477 7793Clinical Pathology Department, Faculty of Medicine, Tanta University, El Geish Street, Tanta, 31527 El-Gharbia Governorate Egypt
| | - Hemat E. El-Horany
- grid.412258.80000 0000 9477 7793Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, 31527 Egypt ,grid.443320.20000 0004 0608 0056Biochemistry Department, College of Medicine, Ha’il University, Ha’il, 55211 Saudi Arabia
| | - Marwa M. Elhosary
- grid.412258.80000 0000 9477 7793Msc Immunology from Tanta Faculty of Science, Tanta, 31527 Egypt
| | - Ahmed A. Elhadidy
- grid.412258.80000 0000 9477 7793Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta, 31527 Egypt
| |
Collapse
|
15
|
Abou Daher A, Alkhansa S, Azar WS, Rafeh R, Ghadieh HE, Eid AA. Translational Aspects of the Mammalian Target of Rapamycin Complexes in Diabetic Nephropathy. Antioxid Redox Signal 2022; 37:802-819. [PMID: 34544257 DOI: 10.1089/ars.2021.0217] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Despite the many efforts put into understanding diabetic nephropathy (DN), direct treatments for DN have yet to be discovered. Understanding the mechanisms behind DN is an essential step in the development of novel therapeutic regimens. The mammalian target of rapamycin (mTOR) pathway has emerged as an important candidate in the quest for drug discovery because of its role in regulating growth, proliferation, as well as protein and lipid metabolism. Recent Advances: Kidney cells have been found to rely on basal autophagy for survival and for conserving kidney integrity. Recent studies have shown that diabetes induces renal autophagy deregulation, leading to kidney injury. Hyper-activation of the mTOR pathway and oxidative stress have been suggested to play a role in diabetes-induced autophagy imbalance. Critical Issues: A detailed understanding of the role of mTOR signaling in diabetes-associated complications is of major importance in the search for a cure. In this review, we provide evidence that mTOR is heavily implicated in diabetes-induced kidney injury. We suggest possible mechanisms through which mTOR exerts its negative effects by increasing insulin resistance, upregulating oxidative stress, and inhibiting autophagy. Future Directions: Both increased oxidative stress and autophagy deregulation are deeply embedded in DN. However, the mechanisms controlling oxidative stress and autophagy are not well understood. Although Akt/mTOR signaling seems to play an important role in oxidative stress and autophagy, further investigation is required to uncover the details of this signaling pathway. Antioxid. Redox Signal. 37, 802-819.
Collapse
Affiliation(s)
- Alaa Abou Daher
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Sahar Alkhansa
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon.,AUB Diabetes, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - William S Azar
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon.,AUB Diabetes, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon.,Department of Physiology and Biophysics, Georgetown University Medical School, Washington, District of Columbia, USA
| | - Rim Rafeh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon.,AUB Diabetes, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Hilda E Ghadieh
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon.,AUB Diabetes, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon.,AUB Diabetes, Faculty of Medicine and Medical Center, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
16
|
Melnik BC, Schmitz G. Milk Exosomal microRNAs: Postnatal Promoters of β Cell Proliferation but Potential Inducers of β Cell De-Differentiation in Adult Life. Int J Mol Sci 2022; 23:ijms231911503. [PMID: 36232796 PMCID: PMC9569743 DOI: 10.3390/ijms231911503] [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: 08/29/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic β cell expansion and functional maturation during the birth-to-weaning period is driven by epigenetic programs primarily triggered by growth factors, hormones, and nutrients provided by human milk. As shown recently, exosomes derived from various origins interact with β cells. This review elucidates the potential role of milk-derived exosomes (MEX) and their microRNAs (miRs) on pancreatic β cell programming during the postnatal period of lactation as well as during continuous cow milk exposure of adult humans to bovine MEX. Mechanistic evidence suggests that MEX miRs stimulate mTORC1/c-MYC-dependent postnatal β cell proliferation and glycolysis, but attenuate β cell differentiation, mitochondrial function, and insulin synthesis and secretion. MEX miR content is negatively affected by maternal obesity, gestational diabetes, psychological stress, caesarean delivery, and is completely absent in infant formula. Weaning-related disappearance of MEX miRs may be the critical event switching β cells from proliferation to TGF-β/AMPK-mediated cell differentiation, whereas continued exposure of adult humans to bovine MEX miRs via intake of pasteurized cow milk may reverse β cell differentiation, promoting β cell de-differentiation. Whereas MEX miR signaling supports postnatal β cell proliferation (diabetes prevention), persistent bovine MEX exposure after the lactation period may de-differentiate β cells back to the postnatal phenotype (diabetes induction).
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
- Correspondence: ; Tel.: +49-52-4198-8060
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, University of Regensburg, D-93053 Regensburg, Germany
| |
Collapse
|
17
|
Implications of a Neuronal Receptor Family, Metabotropic Glutamate Receptors, in Cancer Development and Progression. Cells 2022; 11:cells11182857. [PMID: 36139432 PMCID: PMC9496915 DOI: 10.3390/cells11182857] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/29/2022] [Accepted: 09/07/2022] [Indexed: 12/03/2022] Open
Abstract
Cancer is the second leading cause of death, and incidences are increasing globally. Simply defined, cancer is the uncontrolled proliferation of a cell, and depending on the tissue of origin, the cancer etiology, biology, progression, prognosis, and treatment will differ. Carcinogenesis and its progression are associated with genetic factors that can either be inherited and/or acquired and are classified as an oncogene or tumor suppressor. Many of these genetic factors converge on common signaling pathway(s), such as the MAPK and PI3K/AKT pathways. In this review, we will focus on the metabotropic glutamate receptor (mGluR) family, an upstream protein that transmits extracellular signals into the cell and has been shown to regulate many aspects of tumor development and progression. We explore the involvement of members of this receptor family in various cancers that include breast cancer, colorectal cancer, glioma, kidney cancer, melanoma, oral cancer, osteosarcoma, pancreatic cancer, prostate cancer, and T-cell cancers. Intriguingly, depending on the member, mGluRs can either be classified as oncogenes or tumor suppressors, although in general most act as an oncogene. The extensive work done to elucidate the role of mGluRs in various cancers suggests that it might be a viable strategy to therapeutically target glutamatergic signaling.
Collapse
|
18
|
Castro‐Guarda M, Arancibia Y, Chipón C, Matamala C, Oyarzo P, Vargas G, Reyes A, Salas M, Morera FJ, Zambrano A. Metabolic changes induced by DNA damage in Ramos cells: exploring the role of mTORC1 complex. FEBS Open Bio 2022; 12:1509-1522. [PMID: 35538662 PMCID: PMC9340868 DOI: 10.1002/2211-5463.13436] [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: 09/16/2021] [Revised: 02/26/2022] [Accepted: 05/09/2022] [Indexed: 11/09/2022] Open
Abstract
DNA damage induces the activation of many different signals associated with repair or cell death, but it is also connected with physiological events, such as adult neurogenesis and B-cell differentiation. DNA damage induces different signaling pathways, some of them linked to important metabolic changes. The mTORC1 pathway has a central role in the regulation of growth processes and cell division in response to environmental changes and also controls protein synthesis, lipid biogenesis, nucleotide synthesis, and expression of glycolytic genes. Here, we report that double-strand breaks induced with etoposide affect the expression of genes encoding different enzymes associated with specific metabolic pathways in Ramos cells. We also analyzed the role of mTOR signaling, demonstrating that double-strand breaks induce downregulation of mTOR signaling. Specific inhibition of mTORC1 using rapamycin also induced changes in the expression of metabolic genes. Finally, we demonstrated that DNA damage and rapamycin can regulate glucose uptake. In summary, our findings show that etoposide and rapamycin affect the expression of metabolic genes as well as apoptotic and proliferation markers in Ramos cells, increasing our understanding of cancer metabolism.
Collapse
Affiliation(s)
- Marcos Castro‐Guarda
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Yennyfer Arancibia
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Carina Chipón
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Christofer Matamala
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Paola Oyarzo
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Gabriela Vargas
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Alejandro Reyes
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
- Universidad Austral de ChileCoyhaiqueChile
| | - Mónica Salas
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Francisco J. Morera
- Facultad de Ciencias VeterinariasInstituto de Farmacología y MorfofisiologíaUniversidad Austral de ChileValdiviaChile
| | - Angara Zambrano
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
- Center for Interdisciplinary Studies on the Nervous System (CISNe)Universidad Austral de ChileValdiviaChile
| |
Collapse
|
19
|
Deshpande R, Lee B, Qiao Y, Grewal SS. TOR signalling is required for host lipid metabolic remodelling and survival following enteric infection in Drosophila. Dis Model Mech 2022; 15:dmm049551. [PMID: 35363274 PMCID: PMC9118046 DOI: 10.1242/dmm.049551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 03/22/2022] [Indexed: 12/29/2022] Open
Abstract
When infected by enteric pathogenic bacteria, animals need to initiate local and whole-body defence strategies. Although most attention has focused on the role of innate immune anti-bacterial responses, less is known about how changes in host metabolism contribute to host defence. Using Drosophila as a model system, we identify induction of intestinal target-of-rapamycin (TOR) kinase signalling as a key adaptive metabolic response to enteric infection. We find that enteric infection induces both local and systemic induction of TOR independently of the Immune deficiency (IMD) innate immune pathway, and we see that TOR functions together with IMD signalling to promote infection survival. These protective effects of TOR signalling are associated with remodelling of host lipid metabolism. Thus, we see that TOR is required to limit excessive infection-mediated wasting of host lipid stores by promoting an increase in the levels of gut- and fat body-expressed lipid synthesis genes. Our data support a model in which induction of TOR represents a host tolerance response to counteract infection-mediated lipid wasting in order to promote survival. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | | | | | - Savraj S. Grewal
- Clark H Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children's Hospital Research Institute and Department of Biochemistry and Molecular Biology Calgary, University of Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
20
|
Kisla MM, Ates-Alagoz Z. Benzimidazoles Against Certain Breast Cancer Drug Targets: A Review. Mini Rev Med Chem 2022; 22:2463-2477. [PMID: 35345997 DOI: 10.2174/1389557522666220328161217] [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: 11/26/2021] [Revised: 01/01/2022] [Accepted: 02/09/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Benzimidazoles are widely used scaffolds against various types of cancer including breast cancer. To this end, anticancer agents must be developed using the knowledge of the specific targets of BC. OBJECTIVE In this study, we aim to review the compounds used against some of the biomolecular targets of breast cancer. To this end, we present information about the various targets, with their latest innovative studies. CONCLUSION Benzimidazole ring is an important building block that can target diverse cancer scenarios since it can structurally mimic biomolecules in the human body. Additionally, many studies imply the involvement of this moiety on a plethora of pathways and enzymes related to BC. Herein, our target-based collection of benzimidazole derivatives strongly suggests the utilization of benzimidazole derivatives against BC.
Collapse
Affiliation(s)
- Mehmet Murat Kisla
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Zeynep Ates-Alagoz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
21
|
Chen N, Zhou YS, Wang LC, Huang JB. Advances in metformin‑based metabolic therapy for non‑small cell lung cancer (Review). Oncol Rep 2022; 47:55. [PMID: 35039878 PMCID: PMC8808708 DOI: 10.3892/or.2022.8266] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022] Open
Abstract
Therapeutic approaches that target the metabolism of tumor cells have been a popular research topic in recent years. Previous studies have demonstrated that glycolysis inhibitors reduce the proliferation of non‑small cell lung cancer (NSCLC) cells by interfering with the aerobic glycolytic pathway. However, the mitochondrial oxidative phosphorylation (OXPHOS) pathway in tumor cells has also been implicated in lung cancer metabolism. Metformin, a known inhibitor of mitochondrial OXPHOS, has been indicated to reduce NSCLC morbidity and mortality in clinical studies. The present article reviewed the therapeutic effects of metformin against NSCLC, both as a single agent and combined with other anticancer treatments, in order to provide a theoretical basis for its clinical use in adjuvant therapy for NSCLC.
Collapse
Affiliation(s)
- Na Chen
- Department of Medical Imaging, Faculty of Medicine, Yangtze University, Yangtze University Research and Experimentation Centre, Jingzhou, Hubei 434000, P.R. China
| | - Yi-Shu Zhou
- Department of Medical Imaging, Faculty of Medicine, Yangtze University, Yangtze University Research and Experimentation Centre, Jingzhou, Hubei 434000, P.R. China
| | - Li-Cui Wang
- Department of Medical Imaging, Faculty of Medicine, Yangtze University, Yangtze University Research and Experimentation Centre, Jingzhou, Hubei 434000, P.R. China
| | - Jin-Bai Huang
- Department of Medical Imaging, Faculty of Medicine, Yangtze University, Yangtze University Research and Experimentation Centre, Jingzhou, Hubei 434000, P.R. China
| |
Collapse
|
22
|
Liu C, Jin Y, Fan Z. The Mechanism of Warburg Effect-Induced Chemoresistance in Cancer. Front Oncol 2021; 11:698023. [PMID: 34540667 PMCID: PMC8446599 DOI: 10.3389/fonc.2021.698023] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/11/2021] [Indexed: 12/26/2022] Open
Abstract
Although chemotherapy can improve the overall survival and prognosis of cancer patients, chemoresistance remains an obstacle due to the diversity, heterogeneity, and adaptability to environmental alters in clinic. To determine more possibilities for cancer therapy, recent studies have begun to explore changes in the metabolism, especially glycolysis. The Warburg effect is a hallmark of cancer that refers to the preference of cancer cells to metabolize glucose anaerobically rather than aerobically, even under normoxia, which contributes to chemoresistance. However, the association between glycolysis and chemoresistance and molecular mechanisms of glycolysis-induced chemoresistance remains unclear. This review describes the mechanism of glycolysis-induced chemoresistance from the aspects of glycolysis process, signaling pathways, tumor microenvironment, and their interactions. The understanding of how glycolysis induces chemoresistance may provide new molecular targets and concepts for cancer therapy.
Collapse
Affiliation(s)
- Chang Liu
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, China
| | - Ying Jin
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, China
| | - Zhimin Fan
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
23
|
Devilliers M, Garrido D, Poidevin M, Rubin T, Le Rouzic A, Montagne J. Differential metabolic sensitivity of insulin-like-response- and TORC1-dependent overgrowth in Drosophila fat cells. Genetics 2021; 217:1-12. [PMID: 33683355 DOI: 10.1093/genetics/iyaa010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022] Open
Abstract
Glycolysis and fatty acid (FA) synthesis directs the production of energy-carrying molecules and building blocks necessary to support cell growth, although the absolute requirement of these metabolic pathways must be deeply investigated. Here, we used Drosophila genetics and focus on the TOR (Target of Rapamycin) signaling network that controls cell growth and homeostasis. In mammals, mTOR (mechanistic-TOR) is present in two distinct complexes, mTORC1 and mTORC2; the former directly responds to amino acids and energy levels, whereas the latter sustains insulin-like-peptide (Ilp) response. The TORC1 and Ilp signaling branches can be independently modulated in most Drosophila tissues. We show that TORC1 and Ilp-dependent overgrowth can operate independently in fat cells and that ubiquitous over-activation of TORC1 or Ilp signaling affects basal metabolism, supporting the use of Drosophila as a powerful model to study the link between growth and metabolism. We show that cell-autonomous restriction of glycolysis or FA synthesis in fat cells retrains overgrowth dependent on Ilp signaling but not TORC1 signaling. Additionally, the mutation of FASN (Fatty acid synthase) results in a drop in TORC1 but not Ilp signaling, whereas, at the cell-autonomous level, this mutation affects none of these signals in fat cells. These findings thus reveal differential metabolic sensitivity of TORC1- and Ilp-dependent growth and suggest that cell-autonomous metabolic defects might elicit local compensatory pathways. Conversely, enzyme knockdown in the whole organism results in animal death. Importantly, our study weakens the use of single inhibitors to fight mTOR-related diseases and strengthens the use of drug combination and selective tissue-targeting.
Collapse
Affiliation(s)
- Maelle Devilliers
- Institute for Integrative Biology of the Cell (I2BC), CNRS, Université Paris-Saclay, CEA, F-91190 Gif-sur-Yvette, France
| | - Damien Garrido
- Institute for Integrative Biology of the Cell (I2BC), CNRS, Université Paris-Saclay, CEA, F-91190 Gif-sur-Yvette, France
| | - Mickael Poidevin
- Institute for Integrative Biology of the Cell (I2BC), CNRS, Université Paris-Saclay, CEA, F-91190 Gif-sur-Yvette, France
| | - Thomas Rubin
- Institute for Integrative Biology of the Cell (I2BC), CNRS, Université Paris-Saclay, CEA, F-91190 Gif-sur-Yvette, France
| | - Arnaud Le Rouzic
- Laboratoire Evolution, Génomes, Comportement et Ecologie, CNRS, Université Paris-Saclay, UMR 9191, F-91190 Gif-sur-Yvette, France
| | - Jacques Montagne
- Institute for Integrative Biology of the Cell (I2BC), CNRS, Université Paris-Saclay, CEA, F-91190 Gif-sur-Yvette, France
| |
Collapse
|
24
|
Bodineau C, Tomé M, Courtois S, Costa ASH, Sciacovelli M, Rousseau B, Richard E, Vacher P, Parejo-Pérez C, Bessede E, Varon C, Soubeyran P, Frezza C, Murdoch PDS, Villar VH, Durán RV. Two parallel pathways connect glutamine metabolism and mTORC1 activity to regulate glutamoptosis. Nat Commun 2021; 12:4814. [PMID: 34376668 PMCID: PMC8355106 DOI: 10.1038/s41467-021-25079-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 07/16/2021] [Indexed: 11/08/2022] Open
Abstract
Glutamoptosis is the induction of apoptotic cell death as a consequence of the aberrant activation of glutaminolysis and mTORC1 signaling during nutritional imbalance in proliferating cells. The role of the bioenergetic sensor AMPK during glutamoptosis is not defined yet. Here, we show that AMPK reactivation blocks both the glutamine-dependent activation of mTORC1 and glutamoptosis in vitro and in vivo. We also show that glutamine is used for asparagine synthesis and the GABA shunt to produce ATP and to inhibit AMPK, independently of glutaminolysis. Overall, our results indicate that glutamine metabolism is connected with mTORC1 activation through two parallel pathways: an acute alpha-ketoglutarate-dependent pathway; and a secondary ATP/AMPK-dependent pathway. This dual metabolic connection between glutamine and mTORC1 must be considered for the future design of therapeutic strategies to prevent cell growth in diseases such as cancer.
Collapse
Affiliation(s)
- Clément Bodineau
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Seville, Spain
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France
| | - Mercedes Tomé
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Seville, Spain
| | - Sarah Courtois
- Bordeaux Research in Translational Oncology, INSERM U1053, Université de Bordeaux, Bordeaux cedex, France
| | - Ana S H Costa
- Medical Research Council Cancer Unit, Hutchison/MRC Research Centre, Box 197, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Marco Sciacovelli
- Medical Research Council Cancer Unit, Hutchison/MRC Research Centre, Box 197, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Benoit Rousseau
- Service Commun des Animaleries, Animalerie A2, University of Bordeaux, Bordeaux, France
| | | | | | - Carlos Parejo-Pérez
- Instituto de Bioquímica Vegetal y Fotosíntesis, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Seville, Spain
| | - Emilie Bessede
- Bordeaux Research in Translational Oncology, INSERM U1053, Université de Bordeaux, Bordeaux cedex, France
| | - Christine Varon
- Bordeaux Research in Translational Oncology, INSERM U1053, Université de Bordeaux, Bordeaux cedex, France
| | | | - Christian Frezza
- Medical Research Council Cancer Unit, Hutchison/MRC Research Centre, Box 197, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Piedad Del Socorro Murdoch
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Seville, Spain
- Departamento de Bioquímica Vegetal y Biología Molecular, Universidad de Sevilla, Seville, Spain
| | | | - Raúl V Durán
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Universidad Pablo de Olavide, Seville, Spain.
- Institut Européen de Chimie et Biologie, INSERM U1218, Université de Bordeaux, Pessac, France.
- INSERM U1218, Institut Bergonié, Bordeaux, France.
| |
Collapse
|
25
|
Pan M, Yin Y, Wang X, Wang Q, Zhang L, Hu H, Wang C. Mice deficient in UXT exhibit retinitis pigmentosa-like features via aberrant autophagy activation. Autophagy 2021; 17:1873-1888. [PMID: 32744119 PMCID: PMC8386600 DOI: 10.1080/15548627.2020.1796015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/29/2020] [Accepted: 07/09/2020] [Indexed: 11/22/2022] Open
Abstract
UXT (ubiquitously expressed prefoldin like chaperone), a small chaperone-like protein, is widely expressed in diverse human and mouse tissues and is more abundant in retina and kidney. However, the functional characterization of UXT at tissue level was largely unknown. Here, we reported that mice deficient in UXT exhibited salient features of retinal degenerative disease, similar to retinitis pigmentosa. Conditional knockout (CKO) of Uxt led to retinal degeneration and pigmentation in mice retina along with significant alterations of retinitis pigmentosa-related genes, which indicated UXT might be associated with retinal degenerative disease sharing key features to retinitis pigmentosa. Consistently, the electroretinogram (ERG) responses were dramatically impaired in uxt CKO retinas. Strong degenerative features were observed in uxt CKO retinas, including specific and progressive reduction of photoreceptor cells and increased numbers of apoptotic cells. Intriguingly, macroautophagic/autophagic flux was enhanced in uxt CKO retina. Mechanistically, we found UXT was indispensable to suppress photoreceptor apoptotic cell death by inhibiting autophagy through regulating the activity of MTOR (mechanistic target of rapamycin kinase), a key negative regulator of autophagy. Conversely, knockdown of UXT induced the robust expression of the canonical autophagy-related genes and boosted autophagic flux and apoptosis, finally resulting in severe retina degeneration in uxt CKO mice. Taken together, our study reveals a vital role of UXT in preventing retina from degeneration. The loss of UXT results in a hyper-autophagic state leading to massive retinal degeneration. Therefore, UXT may be a crucial target for retinal degenerative disease.Abbreviations: 3-ma: 3-methyladenine; casp3: caspase 3; cko: conditional knockout; erg: electroretinogram; gapdh: glyceraldehyde-3-phosphate dehydrogenase; map1lc3b/lc3b: microtubule-associated protein 1 light chain 3; mtor: mechanistic target of rapamycin kinase; parp: poly (adp-ribose) polymerase family; rna-seq: rna sequencing; rp: retinitis pigmentosa; rps6kb1/s6k: ribosomal protein s6 kinase b1; sqstm1: sequestosome 1; tunel: terminal deoxynucleotidyl transferase mediated dutp nick-end labeling; uxt: ubiquitously expressed prefoldin like chaperone.
Collapse
Affiliation(s)
- Mingyu Pan
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yue Yin
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xinxia Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Quanyi Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Lele Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Haiyang Hu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
26
|
Cayo A, Segovia R, Venturini W, Moore-Carrasco R, Valenzuela C, Brown N. mTOR Activity and Autophagy in Senescent Cells, a Complex Partnership. Int J Mol Sci 2021; 22:ijms22158149. [PMID: 34360912 PMCID: PMC8347619 DOI: 10.3390/ijms22158149] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022] Open
Abstract
Cellular senescence is a form of proliferative arrest triggered in response to a wide variety of stimuli and characterized by unique changes in cell morphology and function. Although unable to divide, senescent cells remain metabolically active and acquire the ability to produce and secrete bioactive molecules, some of which have recognized pro-inflammatory and/or pro-tumorigenic actions. As expected, this “senescence-associated secretory phenotype (SASP)” accounts for most of the non-cell-autonomous effects of senescent cells, which can be beneficial or detrimental for tissue homeostasis, depending on the context. It is now evident that many features linked to cellular senescence, including the SASP, reflect complex changes in the activities of mTOR and other metabolic pathways. Indeed, the available evidence indicates that mTOR-dependent signaling is required for the maintenance or implementation of different aspects of cellular senescence. Thus, depending on the cell type and biological context, inhibiting mTOR in cells undergoing senescence can reverse senescence, induce quiescence or cell death, or exacerbate some features of senescent cells while inhibiting others. Interestingly, autophagy—a highly regulated catabolic process—is also commonly upregulated in senescent cells. As mTOR activation leads to repression of autophagy in non-senescent cells (mTOR as an upstream regulator of autophagy), the upregulation of autophagy observed in senescent cells must take place in an mTOR-independent manner. Notably, there is evidence that autophagy provides free amino acids that feed the mTOR complex 1 (mTORC1), which in turn is required to initiate the synthesis of SASP components. Therefore, mTOR activation can follow the induction of autophagy in senescent cells (mTOR as a downstream effector of autophagy). These functional connections suggest the existence of autophagy regulatory pathways in senescent cells that differ from those activated in non-senescence contexts. We envision that untangling these functional connections will be key for the generation of combinatorial anti-cancer therapies involving pro-senescence drugs, mTOR inhibitors, and/or autophagy inhibitors.
Collapse
Affiliation(s)
- Angel Cayo
- Center for Medical Research, University of Talca School of Medicine, Talca 346000, Chile; (A.C.); (R.S.); (W.V.); (C.V.)
| | - Raúl Segovia
- Center for Medical Research, University of Talca School of Medicine, Talca 346000, Chile; (A.C.); (R.S.); (W.V.); (C.V.)
| | - Whitney Venturini
- Center for Medical Research, University of Talca School of Medicine, Talca 346000, Chile; (A.C.); (R.S.); (W.V.); (C.V.)
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, University of Talca, Talca 346000, Chile;
| | - Rodrigo Moore-Carrasco
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, University of Talca, Talca 346000, Chile;
| | - Claudio Valenzuela
- Center for Medical Research, University of Talca School of Medicine, Talca 346000, Chile; (A.C.); (R.S.); (W.V.); (C.V.)
| | - Nelson Brown
- Center for Medical Research, University of Talca School of Medicine, Talca 346000, Chile; (A.C.); (R.S.); (W.V.); (C.V.)
- Correspondence:
| |
Collapse
|
27
|
Hu C, Cao Y, Li P, Tang X, Yang M, Gu S, Xiong K, Li T, Xiao T. Oleanolic Acid Induces Autophagy and Apoptosis via the AMPK-mTOR Signaling Pathway in Colon Cancer. JOURNAL OF ONCOLOGY 2021; 2021:8281718. [PMID: 34326874 PMCID: PMC8310446 DOI: 10.1155/2021/8281718] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/23/2021] [Accepted: 07/06/2021] [Indexed: 12/31/2022]
Abstract
AIMS The purpose of this study was to explore the biological functions of the mTOR and AMPK signaling pathways in colon cancer (CC). The potential molecular mechanisms by which oleanolic acid (OA) induces autophagy and apoptosis were also investigated. METHODS The biological functions of mTOR were analyzed by GeneCards, the Search Tool for the Retrieval of Interacting Genes (STRING), and the Database for Annotation, Visualization and Integrated Discovery (DAVID). Least absolute shrinkage and selection operator (LASSO) regression analysis was used to obtain prognostic and survival data of CC patients from the Gene Expression Omnibus (GEO) database. The effects of OA on the CC cell lines HCT-116 and SW-480 were analyzed by CCK-8, colony formation assay, and high-content system (HCS) array scan. The apoptosis rate of SW-480 and HCT-116 cells was detected by flow cytometry. The mRNA and protein expression levels in HCT-116 and SW-480 cells and NCM-460 normal colonic epithelial cells were detected by RT-PCR and Western blotting. RESULTS mTOR was highly expressed in CC patients and acted as an oncogene. The AMPK signaling pathway mediated by mTOR predicted the poor prognosis of CC patients. OA effectively inhibited the proliferation and viability of CC cells. Furthermore, the apoptosis rate of CC cells was clearly increased following OA administration. Regarding the molecular mechanism of OA, the results indicated that mTOR and the antiapoptosis gene Bcl-2 were downregulated by OA. In addition, regulator genes of autophagy and apoptosis, including BAX, caspase-9, caspase-8, and caspase-3, were significantly upregulated by OA. Moreover, OA upregulated AMPK and its downstream proteins, including TSC2, BAX, Beclin 1, LC3B-II, and ULK1, to induce autophagy and apoptosis in CC cells. CONCLUSION The findings from this study demonstrate that OA could effectively inhibit the proliferation and viability of CC cells. The anti-CC activity of OA is closely related to the activation of the AMPK-mTOR signaling pathway. Activation of AMPK and inhibition of mTOR are involved in the induction of autophagy and apoptosis by OA. OA induced autophagy and apoptosis mainly in an AMPK activation-dependent manner in CC cells.
Collapse
Affiliation(s)
- Changxiao Hu
- College of Clinical Medicine, Guizhou University of Traditional Chinese Medicine, No. 71 Baoshan North Road, Guiyang 550001, China
| | - Yibo Cao
- Colorectal and Anal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Baoshan North Road, Guiyang 550001, China
| | - Ping Li
- Colorectal and Anal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Baoshan North Road, Guiyang 550001, China
| | - Xiaorong Tang
- Colorectal and Anal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Baoshan North Road, Guiyang 550001, China
| | - Minhui Yang
- College of Clinical Medicine, Guizhou University of Traditional Chinese Medicine, No. 71 Baoshan North Road, Guiyang 550001, China
| | - Shengliang Gu
- College of Clinical Medicine, Guizhou University of Traditional Chinese Medicine, No. 71 Baoshan North Road, Guiyang 550001, China
| | - Kai Xiong
- College of Clinical Medicine, Guizhou University of Traditional Chinese Medicine, No. 71 Baoshan North Road, Guiyang 550001, China
| | - Tian Li
- Fourth Military Medical University, No. 169 Changle West Rd, Xi'an 710032, China
| | - Tianbao Xiao
- Colorectal and Anal Surgery, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, No. 71 Baoshan North Road, Guiyang 550001, China
| |
Collapse
|
28
|
Zhao CY, Hua CH, Li CH, Zheng RZ, Li XY. High PYGL Expression Predicts Poor Prognosis in Human Gliomas. Front Neurol 2021; 12:652931. [PMID: 34177761 PMCID: PMC8225935 DOI: 10.3389/fneur.2021.652931] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Background: PYGL has been reported as a glycogen degradation-related gene, which is up-regulated in many tumors. This study was designed to investigate the predictive value of high PYGL expression in patients with gliomas through bioinformatics analysis of the gene transcriptome and the single-cell sequencing data. Methods: The gene transcriptome data of 595 glioma patients from the TCGA database and the single-cell RNA sequencing data of 7,930 GBM cells from the GEO database were included in the study. Differential analysis was used to find the distribution of expression of PYGL in different groups of glioma patients. OS analysis was used to assess the influence of the high expression of PYGL on the prognosis of patients. The reliability of its prediction was evaluated by the AUC of ROC and the C-index. The GSEA be used to reveal potential mechanisms. The single-cell analysis was used to observe the high expression of PYGL in different cell groups to further analyze the mechanism of its prediction. Results: Differential analysis identified the expression level of PYGL is positively associated with glioma malignancy. OS analysis and Cox regression analyses showed high expression of PYGL was an independent factor for poor prognosis of gliomas (p < 0.05). The AUC values were 0.838 (1-year ROC), 0.864 (3-year ROC) and 0.833 (5-year ROC). The C index was 0.81. The GSEA showed that gene sets related to MTORC1 signaling, glycolysis, hypoxia, PI3K/AKT/mTOR signaling, KRAS signaling up and angiogenesis were differentially enriched in the high PYGL expression phenotype. The single-cell sequencing data analysis showed TAMs and malignant cells in GBM tissues expressed a high level of PYGL. Conclusion: The high expression of PYGL is an independent predictor of poor prognosis in patients with glioma.
Collapse
Affiliation(s)
- Chang-Yi Zhao
- Department of Neurosurgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chun-Hui Hua
- Department of Neurosurgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chang-Hua Li
- Department of Neurosurgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui-Zhe Zheng
- Department of Neurosurgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin-Yuan Li
- Department of Neurosurgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Melnik BC. Lifetime Impact of Cow's Milk on Overactivation of mTORC1: From Fetal to Childhood Overgrowth, Acne, Diabetes, Cancers, and Neurodegeneration. Biomolecules 2021; 11:404. [PMID: 33803410 PMCID: PMC8000710 DOI: 10.3390/biom11030404] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 02/07/2023] Open
Abstract
The consumption of cow's milk is a part of the basic nutritional habits of Western industrialized countries. Recent epidemiological studies associate the intake of cow's milk with an increased risk of diseases, which are associated with overactivated mechanistic target of rapamycin complex 1 (mTORC1) signaling. This review presents current epidemiological and translational evidence linking milk consumption to the regulation of mTORC1, the master-switch for eukaryotic cell growth. Epidemiological studies confirm a correlation between cow's milk consumption and birthweight, body mass index, onset of menarche, linear growth during childhood, acne vulgaris, type 2 diabetes mellitus, prostate cancer, breast cancer, hepatocellular carcinoma, diffuse large B-cell lymphoma, neurodegenerative diseases, and all-cause mortality. Thus, long-term persistent consumption of cow's milk increases the risk of mTORC1-driven diseases of civilization. Milk is a highly conserved, lactation genome-controlled signaling system that functions as a maternal-neonatal relay for optimized species-specific activation of mTORC1, the nexus for regulation of eukaryotic cell growth, and control of autophagy. A deeper understanding of milk´s impact on mTORC1 signaling is of critical importance for the prevention of common diseases of civilization.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Am Finkenhügel 7a, D-49076 Osnabrück, Germany
| |
Collapse
|
30
|
Hepatic DNAJB9 Drives Anabolic Biasing to Reduce Steatosis and Obesity. Cell Rep 2021; 30:1835-1847.e9. [PMID: 32049014 DOI: 10.1016/j.celrep.2020.01.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/23/2019] [Accepted: 01/14/2020] [Indexed: 12/12/2022] Open
Abstract
Nutrients stimulate the anabolic synthesis of proteins and lipids, but selective insulin resistance in obesity biases the anabolic program toward lipogenesis. Here, we report the identification of a DNAJB9-driven program that favors protein synthesis and energy production over lipid accumulation. We show there are two pools of DNAJB9 cochaperone. DNAJB9 in the ER lumen promotes the degradation of the lipogenic transcription factor SREBP1c through ERAD, whereas its counterpart on the ER membrane promotes the assembly of mTORC2 in the cytosol and stimulates the synthesis of proteins and ATP. The expression of Dnajb9 is induced by nutrients and downregulated in the obese mouse liver. Restoration of hepatic DNAJB9 expression effectively improves insulin sensitivity, restores protein synthesis, and suppresses food intake, accompanied by reduced hepatic steatosis and adiposity in multiple mouse models of obesity. Therefore, targeting the anabolic balance may provide a unique opportunity to tackle obesity and diabetes.
Collapse
|
31
|
Sun X, Zhou ZR, Fang Y, Ding S, Lu S, Wang Z, Wang H, Chen X, Shen K. A novel metabolic gene signature-based nomogram to predict overall survival in breast cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:367. [PMID: 33842588 PMCID: PMC8033348 DOI: 10.21037/atm-20-4813] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background Breast cancer risk prediction is often based on clinicopathological characteristics despite the high heterogeneity derived from gene expression. Metabolic alteration is a hallmark of cancer, and thus, the integration of a metabolic signature with clinical parameters is necessary to predict disease outcomes in breast cancers. Methods Metabolic genes were downloaded from the Gene Set Enrichment Analysis (GSEA) dataset. Genes with statistical significance in the univariate analysis were applied in the least absolute shrinkage and selection operator (LASSO) analysis to build a gene signature in the GSE20685 dataset. Clinicopathological characteristics and risk scores with prognostic significance were incorporated into the nomogram to predict the overall survival (OS) of patients. The Cancer Genome Atlas (TCGA) and GSE866166 datasets were used as the validation datasets. Time-dependent receiver operating characteristic (tROC) curves and calibration plots were used to assess the accuracy and discrimination of the model. Results A 55-gene metabolic gene signature (MGS) was constructed, and was significantly related to OS both in the discovery (P<0.001) and validation (P<0.001) datasets. The MGS was an independent prognostic factor and could divide patients into high- and low-risk groups regardless of their different prediction analysis of microarray 50 (PAM50) subtypes. Time-dependent ROC curves indicated that the risk scores based on the MGS [area under the ROC curve (AUC): 0.931] were superior to the those based on the American Joint Committee on Cancer (AJCC) stage (AUC: 0.781) and PAM50 (AUC: 0.675). A nomogram based on the AJCC stage and risk score could predict OS, and the calibration curves showed good agreement to the actual outcome, indicating that the nomogram may have practical utility. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analysis indicated that this MGS was primarily enriched in amino acid pathways. Conclusions Our results demonstrated that the MGS was superior to existing risk predictors such as PAM50 and AJCC stage. By combining clinical factors (AJCC stage) and the MGS, a nomogram was constructed and showed good predictive ability for OS in breast cancer.
Collapse
Affiliation(s)
- Xi Sun
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Rui Zhou
- Radiation Oncology Center, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Fang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuning Ding
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuangshuang Lu
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaosong Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kunwei Shen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
32
|
Kishimoto Y, Kondo K, Momiyama Y. The Protective Role of Sestrin2 in Atherosclerotic and Cardiac Diseases. Int J Mol Sci 2021; 22:ijms22031200. [PMID: 33530433 PMCID: PMC7865804 DOI: 10.3390/ijms22031200] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/21/2021] [Accepted: 01/23/2021] [Indexed: 01/22/2023] Open
Abstract
Atherosclerotic disease, such as coronary artery disease (CAD), is known to be a chronic inflammatory disease, as well as an age-related disease. Excessive oxidative stress produced by reactive oxygen species (ROS) contributes to the pathogenesis of atherosclerosis. Sestrin2 is an anti-oxidant protein that is induced by various stresses such as hypoxia, DNA damage, and oxidative stress. Sestrin2 is also suggested to be associated with aging. Sestrin2 is expressed and secreted mainly by macrophages, endothelial cells, and cardiomyocytes. Sestrin2 plays an important role in suppressing the production and accumulation of ROS, thus protecting cells from oxidative damage. Since sestrin2 is reported to have anti-oxidant and anti-inflammatory properties, it may play a protective role against the progression of atherosclerosis and may be a potential therapeutic target for the amelioration of atherosclerosis. Regarding the association between blood sestrin2 levels and atherosclerotic disease, the blood sestrin2 levels in patients with CAD or carotid atherosclerosis were reported to be high. High blood sestrin2 levels in patients with such atherosclerotic disease may reflect a compensatory response to increased oxidative stress and may help protect against the progression of atherosclerosis. This review describes the protective role of sestrin2 against the progression of atherosclerotic and cardiac diseases.
Collapse
Affiliation(s)
- Yoshimi Kishimoto
- Department of Food Science and Human Nutrition, Faculty of Agriculture, Setsunan University, 45-1 Nagaotouge-cho, Hirakata, Osaka 573-0101, Japan
- Correspondence: ; Tel.: +81-72-896-6352
| | - Kazuo Kondo
- Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo 112-8610, Japan;
| | - Yukihiko Momiyama
- Department of Cardiology, National Hospital Organization Tokyo Medical Center, 2-5-1 Higashigaoka, Meguro-ku, Tokyo 152-8902, Japan;
| |
Collapse
|
33
|
Melnik BC. Synergistic Effects of Milk-Derived Exosomes and Galactose on α-Synuclein Pathology in Parkinson's Disease and Type 2 Diabetes Mellitus. Int J Mol Sci 2021; 22:1059. [PMID: 33494388 PMCID: PMC7865729 DOI: 10.3390/ijms22031059] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 12/11/2022] Open
Abstract
Epidemiological studies associate milk consumption with an increased risk of Parkinson's disease (PD) and type 2 diabetes mellitus (T2D). PD is an α-synucleinopathy associated with mitochondrial dysfunction, oxidative stress, deficient lysosomal clearance of α-synuclein (α-syn) and aggregation of misfolded α-syn. In T2D, α-syn promotes co-aggregation with islet amyloid polypeptide in pancreatic β-cells. Prion-like vagal nerve-mediated propagation of exosomal α-syn from the gut to the brain and pancreatic islets apparently link both pathologies. Exosomes are critical transmitters of α-syn from cell to cell especially under conditions of compromised autophagy. This review provides translational evidence that milk exosomes (MEX) disturb α-syn homeostasis. MEX are taken up by intestinal epithelial cells and accumulate in the brain after oral administration to mice. The potential uptake of MEX miRNA-148a and miRNA-21 by enteroendocrine cells in the gut, dopaminergic neurons in substantia nigra and pancreatic β-cells may enhance miRNA-148a/DNMT1-dependent overexpression of α-syn and impair miRNA-148a/PPARGC1A- and miRNA-21/LAMP2A-dependent autophagy driving both diseases. MiRNA-148a- and galactose-induced mitochondrial oxidative stress activate c-Abl-mediated aggregation of α-syn which is exported by exosome release. Via the vagal nerve and/or systemic exosomes, toxic α-syn may spread to dopaminergic neurons and pancreatic β-cells linking the pathogenesis of PD and T2D.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
| |
Collapse
|
34
|
Smith EM, Benbahouche N, Morris K, Wilczynska A, Gillen S, Schmidt T, Meijer H, Jukes-Jones R, Cain K, Jones C, Stoneley M, Waldron J, Bell C, Fonseca B, Blagden S, Willis A, Bushell M. The mTOR regulated RNA-binding protein LARP1 requires PABPC1 for guided mRNA interaction. Nucleic Acids Res 2021; 49:458-478. [PMID: 33332560 PMCID: PMC7797073 DOI: 10.1093/nar/gkaa1189] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 11/16/2020] [Accepted: 12/11/2020] [Indexed: 12/16/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) is a critical regulator of cell growth, integrating multiple signalling cues and pathways. Key among the downstream activities of mTOR is the control of the protein synthesis machinery. This is achieved, in part, via the co-ordinated regulation of mRNAs that contain a terminal oligopyrimidine tract (TOP) at their 5'ends, although the mechanisms by which this occurs downstream of mTOR signalling are still unclear. We used RNA-binding protein (RBP) capture to identify changes in the protein-RNA interaction landscape following mTOR inhibition. Upon mTOR inhibition, the binding of LARP1 to a number of mRNAs, including TOP-containing mRNAs, increased. Importantly, non-TOP-containing mRNAs bound by LARP1 are in a translationally-repressed state, even under control conditions. The mRNA interactome of the LARP1-associated protein PABPC1 was found to have a high degree of overlap with that of LARP1 and our data show that PABPC1 is required for the association of LARP1 with its specific mRNA targets. Finally, we demonstrate that mRNAs, including those encoding proteins critical for cell growth and survival, are translationally repressed when bound by both LARP1 and PABPC1.
Collapse
Affiliation(s)
- Ewan M Smith
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Nour El Houda Benbahouche
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Katherine Morris
- MRC Toxicology Unit, University of Cambridge, Leicester LE1 9HN, UK
| | - Ania Wilczynska
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1QH, UK
| | - Sarah Gillen
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Tobias Schmidt
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Hedda A Meijer
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | | | - Kelvin Cain
- MRC Toxicology Unit, University of Cambridge, Leicester LE1 9HN, UK
| | - Carolyn Jones
- MRC Toxicology Unit, University of Cambridge, Leicester LE1 9HN, UK
| | - Mark Stoneley
- MRC Toxicology Unit, University of Cambridge, Leicester LE1 9HN, UK
| | - Joseph A Waldron
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Cameron Bell
- Cancer Research UK Therapeutic Discovery Laboratories, London Bioscience Innovation Centre, 2 Royal College Street, London NW1 0NH, UK
| | | | - Sarah Blagden
- Department of Oncology, University of Oxford, Oxford, OX3 7LE, UK
| | - Anne E Willis
- MRC Toxicology Unit, University of Cambridge, Leicester LE1 9HN, UK
| | - Martin Bushell
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1QH, UK
| |
Collapse
|
35
|
O'Leary EM, Tian Y, Nigdelioglu R, Witt LJ, Cetin-Atalay R, Meliton AY, Woods PS, Kimmig LM, Sun KA, Gökalp GA, Mutlu GM, Hamanaka RB. TGF-β Promotes Metabolic Reprogramming in Lung Fibroblasts via mTORC1-dependent ATF4 Activation. Am J Respir Cell Mol Biol 2020; 63:601-612. [PMID: 32668192 DOI: 10.1165/rcmb.2020-0143oc] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a fatal interstitial lung disease characterized by the TGF-β (transforming growth factor-β)-dependent differentiation of lung fibroblasts into myofibroblasts, which leads to excessive deposition of collagen proteins and progressive scarring. We have previously shown that synthesis of collagen by myofibroblasts requires de novo synthesis of glycine, the most abundant amino acid found in collagen protein. TGF-β upregulates the expression of the enzymes of the de novo serine-glycine synthesis pathway in lung fibroblasts; however, the transcriptional and signaling regulators of this pathway remain incompletely understood. Here, we demonstrate that TGF-β promotes accumulation of ATF4 (activating transcription factor 4), which is required for increased expression of the serine-glycine synthesis pathway enzymes in response to TGF-β. We found that induction of the integrated stress response (ISR) contributes to TGF-β-induced ATF4 activity; however, the primary driver of ATF4 downstream of TGF-β is activation of mTORC1 (mTOR Complex 1). TGF-β activates the PI3K-Akt-mTOR pathway, and inhibition of PI3K prevents activation of downstream signaling and induction of ATF4. Using a panel of mTOR inhibitors, we found that ATF4 activation is dependent on mTORC1, independent of mTORC2. Rapamycin, which incompletely and allosterically inhibits mTORC1, had no effect on TGF-β-mediated induction of ATF4; however, Rapalink-1, which specifically targets the kinase domain of mTORC1, completely inhibited ATF4 induction and metabolic reprogramming downstream of TGF-β. Our results provide insight into the mechanisms of metabolic reprogramming in myofibroblasts and clarify contradictory published findings on the role of mTOR inhibition in myofibroblast differentiation.
Collapse
Affiliation(s)
- Erin M O'Leary
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Yufeng Tian
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Recep Nigdelioglu
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois; and
| | - Leah J Witt
- Division of Geriatrics and Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, The University of California San Francisco, San Francisco, California
| | - Rengul Cetin-Atalay
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Angelo Y Meliton
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Parker S Woods
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Lucas M Kimmig
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Kaitlyn A Sun
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Gizem A Gökalp
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Gökhan M Mutlu
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Robert B Hamanaka
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, The University of Chicago, Chicago, Illinois
| |
Collapse
|
36
|
Larsen LJ, Møller LB. Crosstalk of Hedgehog and mTORC1 Pathways. Cells 2020; 9:cells9102316. [PMID: 33081032 PMCID: PMC7603200 DOI: 10.3390/cells9102316] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/30/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
Hedgehog (Hh) signaling and mTOR signaling, essential for embryonic development and cellular metabolism, are both coordinated by the primary cilium. Observations from cancer cells strongly indicate crosstalk between Hh and mTOR signaling. This hypothesis is supported by several studies: Evidence points to a TGFβ-mediated crosstalk; Increased PI3K/AKT/mTOR activity leads to increased Hh signaling through regulation of the GLI transcription factors; increased Hh signaling regulates mTORC1 activity positively by upregulating NKX2.2, leading to downregulation of negative mTOR regulators; GSK3 and AMPK are, as members of both signaling pathways, potentially important links between Hh and mTORC1 signaling; The kinase DYRK2 regulates Hh positively and mTORC1 signaling negatively. In contrast, both positive and negative regulation of Hh has been observed for DYRK1A and DYRK1B, which both regulate mTORC1 signaling positively. Based on crosstalk observed between cilia, Hh, and mTORC1, we suggest that the interaction between Hh and mTORC1 is more widespread than it appears from our current knowledge. Although many studies focusing on crosstalk have been carried out, contradictory observations appear and the interplay involving multiple partners is far from solved.
Collapse
|
37
|
Soliman GA, Schooling CM. Causal association between mTOR-dependent EIF-4E and EIF-4A circulating protein levels and type 2 diabetes: a Mendelian randomization study. Sci Rep 2020; 10:15737. [PMID: 32978410 PMCID: PMC7519073 DOI: 10.1038/s41598-020-71987-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 08/19/2020] [Indexed: 12/22/2022] Open
Abstract
The mammalian Target of Rapamycin complex 1 (mTORC1) nutrient-sensing pathway is a central regulator of cell growth and metabolism and is dysregulated in diabetes. The eukaryotic translation initiation factor 4E (EIF-4E) protein, a key regulator of gene translation and protein function, is controlled by mTORC1 and EIF-4E Binding Proteins (EIF4EBPs). Both EIF4EBPs and ribosomal protein S6K kinase (RP-S6K) are downstream effectors regulated by mTORC1 but converge to regulate two independent pathways. We investigated whether the risk of type 2 diabetes varied with genetically predicted EIF-4E, EIF-4A, EIF-4G, EIF4EBP, and RP-S6K circulating levels using Mendelian Randomization. We estimated the causal role of EIF-4F complex, EIF4EBP, and S6K in the circulation on type 2 diabetes, based on independent single nucleotide polymorphisms strongly associated (p = 5 × 10–6) with EIF-4E (16 SNPs), EIF-4A (11 SNPs), EIF-4G (6 SNPs), EIF4EBP2 (12 SNPs), and RP-S6K (16 SNPs). The exposure data were obtained from the INTERVAL study. We applied these SNPs for each exposure to publically available genetic associations with diabetes from the DIAbetes Genetics Replication And Meta-analysis (DIAGRAM) case (n = 26,676) and control (n = 132,532) study (mean age 57.4 years). We meta-analyzed SNP-specific Wald-estimates using inverse variance weighting with multiplicative random effects and conducted sensitivity analysis. Mendelian Randomization (MR-Base) R package was used in the analysis. The PhenoScanner curated database was used to identify disease associations with SNP gene variants. EIF-4E is associated with a lowered risk of type 2 diabetes with an odds ratio (OR) 0.94, 95% confidence interval (0.88, 0.99, p = 0.03) with similar estimates from the weighted median and MR-Egger. Similarly, EIF-4A was associated with lower risk of type 2 diabetes with odds ratio (OR) 0.90, 95% confidence interval (0.85, 0.97, p = 0.0003). Sensitivity analysis using MR-Egger and weighed median analysis does not indicate that there is a pleiotropic effect. This unbiased Mendelian Randomization estimate is consistent with a protective causal association of EIF-4E and EIF-4A on type 2 diabetes. EIF-4E and EIF-4A may be targeted for intervention by repurposing existing therapeutics to reduce the risk of type 2 diabetes.
Collapse
Affiliation(s)
- Ghada A Soliman
- Department of Environmental, Occupational and Geospatial Health Sciences, The City University of New York, Graduate School of Public Health and Health Policy, 55 West 125th St, New York, NY, 10027, USA.
| | - C Mary Schooling
- Department of Environmental, Occupational and Geospatial Health Sciences, The City University of New York, Graduate School of Public Health and Health Policy, 55 West 125th St, New York, NY, 10027, USA.,School of Public Health, Li Ka Shing, Faculty of Medicine, The University of Hong Kong, 7 Sassoon Road, Hong Kong, China
| |
Collapse
|
38
|
Eldad S, Hertz R, Vainer G, Saada A, Bar-Tana J. Treatment of ErbB2 breast cancer by mitochondrial targeting. Cancer Metab 2020; 8:17. [PMID: 32695336 PMCID: PMC7362624 DOI: 10.1186/s40170-020-00223-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/01/2020] [Indexed: 12/14/2022] Open
Abstract
Background ErbB2 breast cancer still remains an unmet need due to primary and/or acquired resistance to current treatment strategies. MEDICA compounds consist of synthetic long-chain α,ω-dicarboxylic acids previously reported to suppress breast cancer in PyMT transgenic mice. Methods MEDICA efficacy and mode of action in the ErbB2 context was studied in ErbB2 transgenic mice and human breast cancer cells. Results MEDICA treatment is shown here to suppress ErbB2 breast tumors and lung metastasis in ErbB2/neu MMTV transgenic mice, to suppress ErbB2/neu xenografts in nod/scid mice, and to suppress survival of AU565 and BT474 human ErbB2 breast cancer cells. Suppression of ErbB2 breast tumors by MEDICA is due to lipid raft disruption with loss of ErbB family members, including EGFR, ErbB2, and ErbB3. In addition, MEDICA inhibits mTORC1 activity, independently of abrogating the ErbB receptors and their signaling cascades. The double hit of MEDICA in abrogating ErbB and mTORC1 is partly accounted for by targeting mitochondria complex I. Conclusions Mitochondrial targeting by MEDICA suppresses ErbB2 breast tumors and metastasis due to lipid raft disruption and inhibition of mTORC1 activity. Inhibition of mTORC1 activity by MEDICA avoids the resistance acquired by canonical mTORC1 inhibitors like rapalogs or mTOR kinase inhibitors.
Collapse
Affiliation(s)
- Sophia Eldad
- Dept of Human Nutrition and Metabolism, Hebrew University Medical School, 91120 Jerusalem, Israel
| | - Rachel Hertz
- Dept of Human Nutrition and Metabolism, Hebrew University Medical School, 91120 Jerusalem, Israel
| | - Gilad Vainer
- Dept of Pathology, Hadassah-Hebrew University Medical Center, 91120 Jerusalem, Israel
| | - Ann Saada
- Department of Genetics and Metabolic Diseases, Hadassah-Hebrew University Medical Center, 91120 Jerusalem, Israel
| | - Jacob Bar-Tana
- Dept of Human Nutrition and Metabolism, Hebrew University Medical School, 91120 Jerusalem, Israel
| |
Collapse
|
39
|
Abstract
Celiac disease (CD) is an enteropathy triggered by the ingestion of gluten proteins in genetically predisposed individuals and characterized by excessive activation of effector immune cells and enhanced production of inflammatory cytokines. However, factors/mechanisms that amplify the ongoing mucosal inflammation in CD are not fully understood. In this study, we assessed whether mammalian target of Rapamycin (mTOR), a pathway that combines intra- and extra-cellular signals and acts as a central regulator for the metabolism, growth, and function of immune and non-immune cells, sustains CD-associated immune response. Our findings indicate that expression of phosphorylated (p)/active form of mTOR is increased in protein lysates of duodenal biopsy samples taken from patients with active CD (ACD) as compared to normal controls. In ACD, activation of mTOR occurs mainly in the epithelial compartment and associates with enhanced expression of p-4EBP, a downstream target of mTOR complex (mTORC)1, while expression of p-Rictor, a component of mTORC2, is not increased. Stimulation of mucosal explants of inactive CD patients with pepsin-trypsin-digested (PT)-gliadin or IFN-γ/IL-21, two cytokines produced in CD by gluten-specific T cells, increases p-4EBP expression. Consistently, blockade of such cytokines in cultures of ACD mucosal explants reduces p-4EBP. Finally, we show that inhibition of mTORC1 with rapamycin in ACD mucosal explants reduces p-4EBP and production of IL-15, a master cytokine produced by epithelial cells in this disorder. Our data suggest that ACD inflammation is marked by activation of mTORC1 in the epithelial compartment.
Collapse
|
40
|
Michalopoulou E, Auciello FR, Bulusu V, Strachan D, Campbell AD, Tait-Mulder J, Karim SA, Morton JP, Sansom OJ, Kamphorst JJ. Macropinocytosis Renders a Subset of Pancreatic Tumor Cells Resistant to mTOR Inhibition. Cell Rep 2020; 30:2729-2742.e4. [PMID: 32101748 PMCID: PMC7043007 DOI: 10.1016/j.celrep.2020.01.080] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 10/14/2019] [Accepted: 01/21/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) features a near-universal mutation in KRAS. Additionally, the tumor suppressor PTEN is lost in ∼10% of patients, and in mouse models, this dramatically accelerates tumor progression. While oncogenic KRAS and phosphatidylinositol 3-kinase (PI3K) cause divergent metabolic phenotypes individually, how they synergize to promote tumor metabolic alterations and dependencies remains unknown. We show that in KRAS-driven murine PDAC cells, loss of Pten strongly enhances both mTOR signaling and macropinocytosis. Protein scavenging alleviates sensitivity to mTOR inhibition by rescuing AKT phosphorylation at serine 473 and consequently cell proliferation. Combined inhibition of mTOR and lysosomal processing of internalized protein eliminates the macropinocytosis-mediated resistance. Our results indicate that mTORC2, rather than mTORC1, is an important regulator of protein scavenging and that protein-mediated resistance could explain the lack of effectiveness of mTOR inhibitors in certain genetic backgrounds. Concurrent inhibition of mTOR and protein scavenging might be a valuable therapeutic approach.
Collapse
Affiliation(s)
- Evdokia Michalopoulou
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Francesca R Auciello
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Vinay Bulusu
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - David Strachan
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Andrew D Campbell
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Jacqueline Tait-Mulder
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Saadia A Karim
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Jennifer P Morton
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Jurre J Kamphorst
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.
| |
Collapse
|
41
|
mTOR Regulation of Metabolism in Hematologic Malignancies. Cells 2020; 9:cells9020404. [PMID: 32053876 PMCID: PMC7072383 DOI: 10.3390/cells9020404] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/02/2020] [Accepted: 02/07/2020] [Indexed: 02/06/2023] Open
Abstract
Neoplastic cells rewire their metabolism, acquiring a selective advantage over normal cells and a protection from therapeutic agents. The mammalian Target of Rapamycin (mTOR) is a serine/threonine kinase involved in a variety of cellular activities, including the control of metabolic processes. mTOR is hyperactivated in a large number of tumor types, and among them, in many hematologic malignancies. In this article, we summarized the evidence from the literature that describes a central role for mTOR in the acquisition of new metabolic phenotypes for different hematologic malignancies, in concert with other metabolic modulators (AMPK, HIF1α) and microenvironmental stimuli, and shows how these features can be targeted for therapeutic purposes.
Collapse
|
42
|
Association of serum sestrin 2 and betatrophin with serum neutrophil gelatinase associated lipocalin levels in type 2 diabetic patients with diabetic nephropathy. J Diabetes Metab Disord 2020; 19:249-256. [PMID: 32548072 PMCID: PMC7270235 DOI: 10.1007/s40200-020-00498-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/28/2020] [Indexed: 01/21/2023]
Abstract
Purpose Understanding the pathogenesis and the molecular mechanisms of diabetic nephropathy (DN) helps its timely detection and prevention. The current work aims tomeasure serum sestrin 2 and betatrophin levels in healthy and type diabetic (T2DM)subjects with/or without diabetic nephropathy (DN) and also to test their correlation with serum neutrophil gelatinase associated lipocalin (sNGAL); indicator of DN. Methods This study included 96 subjects; 20 healthy (G1) and 76 T2DM [22 normoalbuminuric (G2), 35 microalbuminuric (G3) and 19 macroalbuminuric (G4)]. Serum sestrin 2, betatrophin and NGAL were measured by their corresponding kits. Results Significant low levels of serum sestrin 2 andhigh levels of serum betatrophin were found in T2DM group when compared to G1 (p = 0.002,p > 0.001, respectively) and this difference is manifested in G4 followed, in order, by G3, G2 then G1 (p= > 0.001 for both). Also, serum sestrin2 levels showed significant negative correlations with sNGAL in G1 (r = -0.497, p = 0.026), G2 (r = -0.784, p > 0.001), G3 (r = -0.894, p > 0.001) and G4 (r = -0.896, pp. > 0.001) while serum betatrophin levels showed significant positive correlations with sNGAL in G2 (r = 0.681, p > 0.001), G3 (r = 0.518, p > 0.001) and G4 (r = 0.727, p > 0.001). Conclusion Serum sestrin 2 levels decrease significantly while betatrophin levels increase significantly in T2DM patients with DN especially those with macroalbuminuria. These levels have significant effect strengths on the indicator of diabetic nephropathy; sNGAL which might indicate theirvaluablerole in the timely detection and prevention of the development of DN.
Collapse
|
43
|
Ma L, Zong X. Metabolic Symbiosis in Chemoresistance: Refocusing the Role of Aerobic Glycolysis. Front Oncol 2020; 10:5. [PMID: 32038983 PMCID: PMC6992567 DOI: 10.3389/fonc.2020.00005] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 01/06/2020] [Indexed: 12/12/2022] Open
Abstract
Cellular metabolic reprogramming is now recognized as a hallmark of tumors. Altered tumor metabolism determines the malignant biological behaviors and phenotypes of cancer. More recently, studies have begun to reveal that cancer cells generally exhibit increased glycolysis or oxidative phosphorylation (OXPHOS) for Adenosine Triphosphate(ATP)generation, which is frequently associated with drug resistance. The metabolism of drug-resistant cells is regulated by the PI3K/AKT/mTOR pathway which ultimately confer cancer cells drug resistance phenotype. The key enzymes involved in glycolysis and the key molecules in relevant pathways have been used as targets to reverse drug resistance. In this review, we highlight our current understanding of the role of metabolic symbiosis in therapeutic resistance and discuss the ongoing effort to develop metabolic inhibitors as anti-cancer drugs to overcome drug resistance to classical chemotherapy.
Collapse
Affiliation(s)
- Lisi Ma
- Department of Breast Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiangyun Zong
- Department of Breast Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
44
|
He X, Yang Y, Yao MW, Ren TT, Guo W, Li L, Xu X. Full title: High glucose protects mesenchymal stem cells from metformin-induced apoptosis through the AMPK-mediated mTOR pathway. Sci Rep 2019; 9:17764. [PMID: 31780804 PMCID: PMC6882892 DOI: 10.1038/s41598-019-54291-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 09/02/2019] [Indexed: 12/14/2022] Open
Abstract
Micro- and macro-vascular events are directly associated with hyperglycemia in patients with type 2 diabetes mellitus (T2DM), but whether intensive glucose control decreases the risk of diabetic cardiovascular complications remains uncertain. Many studies have confirmed that impaired quality and quantity of mesenchymal stem cells (MSCs) plays a pathogenic role in diabetes. Our previous study found that the abundance of circulating MSCs was significantly decreased in patients with T2DM, which was correlated with the progression of diabetic complications. In addition, metformin-induced MSC apoptosis is one of the reasons for the decreased quantity of endogenous or exogenous MSCs during intensive glucose control. However, the role of glucose in metformin-induced MSC apoptosis during intensive glucose control in T2DM remains unknown. In this study, we found that metformin induces MSC apoptosis during intensive glucose control, while high glucose (standard glucose control) could significantly reverse its adverse effect in an AMPK-mTOR pathway dependent manner. Thus, our results indicate that the poorer clinical benefit of the intensive glucose control strategy may be related to an adverse effect due to metformin-induced MSC apoptosis during intensive glucose control therapy in patients with T2DM.
Collapse
Affiliation(s)
- Xiao He
- Department of Stem Cell and Regenerative Medicine, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
- Central Laboratory, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
- PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Yi Yang
- Central Laboratory, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
- Department of Rheumatology and Clinical Immunology, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
| | - Meng-Wei Yao
- Department of Stem Cell and Regenerative Medicine, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
- Department Biochemistry and Molecular Biology, College of Basic Medical Sciences, Army Medical University, Chongqing, P.R. China
| | - Ting-Ting Ren
- Department of Stem Cell and Regenerative Medicine, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
- Central Laboratory, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
- Department of Histology and Embryology, Qingdao University Medical College, Qingdao, Shandong, P.R. China
| | - Wei Guo
- Department of Stem Cell and Regenerative Medicine, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
- Central Laboratory, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
| | - Ling Li
- Department of Histology and Embryology, Qingdao University Medical College, Qingdao, Shandong, P.R. China
| | - Xiang Xu
- Department of Stem Cell and Regenerative Medicine, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China.
- Central Laboratory, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China.
- Department Biochemistry and Molecular Biology, College of Basic Medical Sciences, Army Medical University, Chongqing, P.R. China.
| |
Collapse
|
45
|
Brito AS, Soto Diaz S, Van Vooren P, Godard P, Marini AM, Boeckstaens M. Pib2-Dependent Feedback Control of the TORC1 Signaling Network by the Npr1 Kinase. iScience 2019; 20:415-433. [PMID: 31622882 PMCID: PMC6817644 DOI: 10.1016/j.isci.2019.09.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 05/10/2019] [Accepted: 09/13/2019] [Indexed: 01/21/2023] Open
Abstract
To adjust cell growth and metabolism according to environmental conditions, the conserved TORC1 signaling network controls autophagy, protein synthesis, and turnover. Here, we dissected the signals controlling phosphorylation and activity of the TORC1-effector kinase Npr1, involved in tuning the plasma membrane permeability to nitrogen sources. By evaluating a role of pH as a signal, we show that, although a transient cytosolic acidification accompanies nitrogen source entry and is correlated to a rapid TORC1-dependent phosphorylation of Npr1, a pH drop is not a prerequisite for TORC1 activation. We show that the Gtr1/Gtr2 and Pib2 regulators of TORC1 both independently and differently contribute to regulate Npr1 phosphorylation and activity. Finally, our data reveal that Npr1 mediates nitrogen-dependent phosphorylation of Pib2, as well as a Pib2-dependent inhibition of TORC1. This work highlights a feedback control loop likely enabling efficient downregulation and faster re-activation of TORC1 in response to a novel stimulating signal.
Collapse
Affiliation(s)
- Ana Sofia Brito
- Laboratory of Biology of Membrane Transport, IBMM, Université Libre de Bruxelles, rue des Professeurs Jeener et Brachet 12, 6041 Gosselies, Belgium
| | - Silvia Soto Diaz
- Laboratory of Biology of Membrane Transport, IBMM, Université Libre de Bruxelles, rue des Professeurs Jeener et Brachet 12, 6041 Gosselies, Belgium
| | - Pascale Van Vooren
- Laboratory of Biology of Membrane Transport, IBMM, Université Libre de Bruxelles, rue des Professeurs Jeener et Brachet 12, 6041 Gosselies, Belgium
| | - Patrice Godard
- UCB Pharma, Chemin du Foriest, 1420 Braine-l'Alleud, Belgium
| | - Anna Maria Marini
- Laboratory of Biology of Membrane Transport, IBMM, Université Libre de Bruxelles, rue des Professeurs Jeener et Brachet 12, 6041 Gosselies, Belgium
| | - Mélanie Boeckstaens
- Laboratory of Biology of Membrane Transport, IBMM, Université Libre de Bruxelles, rue des Professeurs Jeener et Brachet 12, 6041 Gosselies, Belgium.
| |
Collapse
|
46
|
de Oliveira Boldrini V, Dos Santos Farias A, Degasperi GR. Deciphering targets of Th17 cells fate: From metabolism to nuclear receptors. Scand J Immunol 2019; 90:e12793. [PMID: 31141182 DOI: 10.1111/sji.12793] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/19/2019] [Accepted: 05/24/2019] [Indexed: 12/17/2022]
Abstract
Evidence indicates that reprogramming of metabolism is critically important for the differentiation of CD4 + T lymphocytes, and the manipulation of metabolic pathways in these cells may shape their fate and function. Distinct subgroups from T lymphocytes, such as Th17, adopt specific metabolic programmes to support their needs. Some important metabolic reactions, such as glycolysis, oxidative phosphorylation, are considered important for the differentiation of these lymphocytes. Since their discovery nearly a decade ago, Th17 lymphocytes have received significant attention because of their role in the pathology of several immune-mediated inflammatory diseases such as multiple sclerosis. In this review, it will be discussed as the involvement of T cell metabolism and as metabolic reprogramming in activated T cells dictates fate decisions to Th17. The involvement of nuclear receptors such as RORyt e PPARs in the induction of Th17 cells was also discussed. Understanding the metabolic pathways involved in the differentiation of the distinct subgroups of T lymphocytes helps in the design of promising therapeutic proposals.
Collapse
Affiliation(s)
- Vinícius de Oliveira Boldrini
- Autoimmune Research Laboratory, Department of Genetics, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil.,Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Brazil
| | - Alessandro Dos Santos Farias
- Autoimmune Research Laboratory, Department of Genetics, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil.,Neuroimmunology Unit, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Brazil
| | | |
Collapse
|
47
|
Torp MK, Yang K, Ranheim T, Husø Lauritzen K, Alfsnes K, Vinge LE, Aukrust P, Stensløkken KO, Yndestad A, Sandanger Ø. Mammalian Target of Rapamycin (mTOR) and the Proteasome Attenuates IL-1β Expression in Primary Mouse Cardiac Fibroblasts. Front Immunol 2019; 10:1285. [PMID: 31244838 PMCID: PMC6563870 DOI: 10.3389/fimmu.2019.01285] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 05/20/2019] [Indexed: 12/21/2022] Open
Abstract
Background: IL-1β is a highly potent pro-inflammatory cytokine and its secretion is tightly regulated. Inactive pro-IL-1β is transcribed in response to innate immune receptors activating NFκB. If tissue damage occurs, danger signals released from necrotic cells, such as ATP, can activate NLRP3-inflammasomes (multiprotein complexes consisting of NLRP3, ASC, and active caspase-1) which cleaves and activates pro-IL-1β. NLRP3 activation also depends on NEK7 and mitochondrial ROS-production. Thus, IL-1β secretion may be regulated at the level of each involved component. We have previously shown that NLRP3-dependent IL-1β release can be induced in cardiac fibroblasts by pro-inflammatory stimuli. However, anti-inflammatory mechanisms targeting IL-1β release in cardiac cells have not been investigated. mTOR is a key regulator of protein metabolism, including autophagy and proteasome activity. In this study we explored whether autophagy or proteasomal degradation are regulators of NLRP3 inflammasome activation and IL-1β release from cardiac fibroblasts. Methods and Results: Serum starvation selectively reduced LPS/ATP-induced IL-1β secretion from cardiac fibroblasts. However, no other inflammasome components, nor mitochondrial mass, were affected. The mTOR inhibitor rapamycin restored pro-IL-1β protein levels as well as LPS/ATP-induced IL-1β release from serum starved cells. However, neither serum starvation nor rapamycin induced autophagy in cardiac fibroblasts. Conversely, chloroquine and bafilomycin A (inhibitors of autophagy) and betulinic acid (a proteasome activator) effectively reduced LPS-induced pro-IL-1β protein levels. Key findings were reinvestigated in human monocyte-derived macrophages. Conclusion: In cardiac fibroblasts, mTOR inhibition selectively favors pro-IL-1β synthesis while proteasomal degradation and not autophagy is the major catabolic anti-inflammatory mechanism for degradation of this cytokine.
Collapse
Affiliation(s)
- May-Kristin Torp
- Division of Physiology, Department of Molecular Medicine, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Kuan Yang
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Trine Ranheim
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Knut Husø Lauritzen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Katrine Alfsnes
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Leif E. Vinge
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Internal Medicine, Diakonhjemmet Hospital, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kåre-Olav Stensløkken
- Division of Physiology, Department of Molecular Medicine, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Arne Yndestad
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Øystein Sandanger
- Centre for Heart Failure Research, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section of Dermatology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
48
|
A Drosophila genetic screen for suppressors of S6kinase-dependent growth identifies the F-box subunit Archipelago/FBXW7. Mol Genet Genomics 2019; 294:573-582. [PMID: 30656413 DOI: 10.1007/s00438-018-01529-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 12/26/2018] [Indexed: 12/12/2022]
Abstract
This study was designed to identify novel negative regulators of the Drosophila S6kinase (dS6K). S6K is a downstream effector of the growth-regulatory complex mTORC1 (mechanistic-Target-of-Rapamycin complex 1). Nutrients activate mTORC1, which in turn induces the phosphorylation of S6K to promote cell growth, whereas fasting represses mTORC1 activity. Here, we screened 11,000 RNA-interfering (RNAi) lines and retained those that enhanced a dS6K-dependent growth phenotype. Since RNAi induces gene knockdown, enhanced tissue growth supports the idea that the targeted gene acts as a growth suppressor. To validate the resulting candidate genes, we monitored dS6K phosphorylation and protein levels in double-stranded RNAi-treated S2 cells. We identified novel dS6K negative regulators, including gene products implicated in basal cellular functions, suggesting that feedback inputs modulate mTORC1/dS6K signaling. We also identified Archipelago (Ago), the Drosophila homologue of FBXW7, which is an E3-ubiquitin-ligase subunit that loads ubiquitin units onto target substrates for proteasome-mediated degradation. Despite a previous report showing an interaction between Ago/FBXW7 and dS6K in a yeast two-hybrid assay and the presence of an Ago/FBXW7-consensus motif in the dS6K polypeptide, we could not see a direct interaction in immunoprecipitation assay. Nevertheless, we observed that loss-of-ago/fbxw7 in larvae resulted in an increase in dS6K protein levels, but no change in the levels of phosphorylated dS6K or dS6K transcripts, suggesting that Ago/FBXW7 indirectly controls dS6K translation or stability. Through the identification of novel negative regulators of the downstream target, dS6K, our study may help deciphering the underlying mechanisms driving deregulations of mTORC1, which underlies several human diseases.
Collapse
|
49
|
Clatici VG, Voicu C, Voaides C, Roseanu A, Icriverzi M, Jurcoane S. Diseases of Civilization - Cancer, Diabetes, Obesity and Acne - the Implication of Milk, IGF-1 and mTORC1. MAEDICA 2018; 13:273-281. [PMID: 30774725 PMCID: PMC6362881 DOI: 10.26574/maedica.2018.13.4.273] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Nutrition and food are one of the most complex aspects of human lives, being influenced by biochemical, psychological, social and cultural factors. The Western diet is the prototype of modern dietary pattern and is mainly characterized by the intake of large amounts of red meat, dairy products, refined grains and sugar. Large amounts of scientific evidence positively correlate Western diet to acne, obesity, diabetes, heart disease and cancer, the so-called "diseases of civilization". The pathophysiological common ground of all these pathologies is the IGF-1 and mTORC pathways, which will be disscussed further in this paper.
Collapse
Affiliation(s)
| | | | | | - Anca Roseanu
- Department of Ligand-Receptor Interaction, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Madalina Icriverzi
- Department of Ligand-Receptor Interaction, Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | | |
Collapse
|
50
|
Santoleri D, Titchenell PM. Resolving the Paradox of Hepatic Insulin Resistance. Cell Mol Gastroenterol Hepatol 2018; 7:447-456. [PMID: 30739869 PMCID: PMC6369222 DOI: 10.1016/j.jcmgh.2018.10.016] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/30/2018] [Accepted: 10/30/2018] [Indexed: 12/12/2022]
Abstract
Insulin resistance is associated with numerous metabolic disorders, such as obesity and type II diabetes, that currently plague our society. Although insulin normally promotes anabolic metabolism in the liver by increasing glucose consumption and lipid synthesis, insulin-resistant individuals fail to inhibit hepatic glucose production and paradoxically have increased liver lipid synthesis, leading to hyperglycemia and hypertriglyceridemia. Here, we detail the intrahepatic and extrahepatic pathways mediating insulin's control of glucose and lipid metabolism. We propose that the interplay between both of these pathways controls insulin signaling and that mis-regulation between the 2 results in the paradoxic effects seen in the insulin-resistant liver instead of the commonly proposed deficiencies in particular branches of only the direct hepatic pathway.
Collapse
Affiliation(s)
- Dominic Santoleri
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Paul M. Titchenell
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania,Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania,Correspondence Address correspondence to: Paul M. Titchenell, PhD, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania 19104. fax: (215) 898-5408.
| |
Collapse
|