1
|
Icard P, Alifano M, Simula L. Citrate oscillations during cell cycle are a targetable vulnerability in cancer cells. Biochim Biophys Acta Rev Cancer 2025; 1880:189313. [PMID: 40216092 DOI: 10.1016/j.bbcan.2025.189313] [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: 09/05/2024] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/20/2025]
Abstract
Cell cycle progression is timely interconnected with oscillations in cellular metabolism. Here, we first describe how these metabolic oscillations allow cycling cells to meet the bioenergetic needs specifically for each phase of the cell cycle. In parallel, we highlight how the cytosolic level of citrate is dynamically regulated during these different phases, being low in G1 phase, increasing in S phase, peaking in G2/M, and decreasing in mitosis. Of note, in cancer cells, a dysregulation of such citrate oscillation can support cell cycle progression by promoting a deregulated Warburg effect (aerobic glycolysis), activating oncogenic signaling pathways (such as PI3K/AKT), and promoting acetyl-CoA production via alternative routes, such as overconsumption of acetate. Then, we review how administration of sodium citrate (at high doses) arrests the cell cycle in G0/G1 or G2/M, inhibits glycolysis and PI3K/AKT, induces apoptosis, and significantly reduces tumor growth in various in vivo models. Last, we reason on the possibility to implement citrate administration to reinforce the effectiveness of cell cycle inhibitors to better cure cancer.
Collapse
Affiliation(s)
- Philippe Icard
- Université de Normandie, UNICAEN, Inserm U1086 Interdisciplinary Research Unit for Cancer Prevention and Treatment, Caen, France; Thoracic Surgery Department, Cochin Hospital, APHP-Centre, Université Paris-Descartes, Paris, France.
| | - Marco Alifano
- Thoracic Surgery Department, Cochin Hospital, APHP-Centre, Université Paris-Descartes, Paris, France; Inserm U1138, Integrative Cancer Immunology, University of Paris, 75006 Paris, France
| | - Luca Simula
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris-Cité, Paris 75014, France
| |
Collapse
|
2
|
Goncu E, Tinartas EP, Gunay B, Ordu T, Turgay Izzetoglu G. Role of Atg3, Atg5 and Atg12 in the crosstalk between apoptosis and autophagy in the posterior silk gland of Bombyx mori. INSECT MOLECULAR BIOLOGY 2025; 34:470-485. [PMID: 39910402 PMCID: PMC12054345 DOI: 10.1111/imb.12985] [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: 09/04/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025]
Abstract
Autophagy is a cellular mechanism that enhances cell survival in response to various stressors, including nutrient deprivation; however, it also plays a pivotal role in the regulation of programmed cell death. This study examined the effects of autophagy-related genes Atg3, Atg5 and Atg12 on apoptosis and autophagy during the degeneration of the posterior silk gland in Bombyx mori, employing RNA interference techniques. Apoptosis-specific markers and autophagic processes were evaluated in both control and treatment groups. The knockdown of all three genes resulted in a significant reduction in autophagy, modifications in the apoptosis process, aberrant expression of p53 and impaired lysosomal function. It was determined that Atg3 is involved in the regulation of intracellular mitochondrial homeostasis. Following the silencing of Atg5, evidence was obtained indicating the gene's role in regulating lysosomal pH. Notably, the loss of Atg3 and Atg5 was associated with an increase in apoptotic markers, whereas the silencing of Atg12 inhibited apoptosis. Elevated levels of the p53 transcription factor following gene silencing suggested a potential interaction between these genes and p53. Our findings further underscore the importance of autophagy-mediated cell death, involving Atg3, Atg5 and Atg12, in the proper progression of degeneration in the posterior silk gland. A comprehensive understanding of the molecular mechanisms that mediate the interaction between apoptosis and autophagy is essential for elucidating their roles in both physiological and pathological contexts.
Collapse
Affiliation(s)
- Ebru Goncu
- Faculty of Science, Department of BiologyEge UniversityBornovaİzmirTürkiye
| | - Esen Poyraz Tinartas
- Faculty of Engineering and Natural Sciences, Department of BiologyManisa Celal Bayar UniversityMuradiyeManisaTürkiye
| | - Busra Gunay
- Faculty of Science, Department of BiologyEge UniversityBornovaİzmirTürkiye
| | - Tugce Ordu
- Faculty of Science, Department of BiologyEge UniversityBornovaİzmirTürkiye
| | | |
Collapse
|
3
|
Plessis-Belair J, Russo T, Riessland M, Sher RB. Nuclear Import Defects Drive Cell Cycle Dysregulation in Neurodegeneration. Aging Cell 2025:e70091. [PMID: 40377023 DOI: 10.1111/acel.70091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/31/2025] [Accepted: 04/14/2025] [Indexed: 05/18/2025] Open
Abstract
Neurodegenerative diseases (NDDs) and other age-related disorders have been classically defined by a set of key pathological hallmarks. Two of these hallmarks, cell cycle dysregulation (CCD) and nucleocytoplasmic transport (NCT) defects, have long been debated as being either causal or consequential in the pathology of accelerated aging. Specifically, aberrant cell cycle activation in post-mitotic neurons has been shown to trigger neuronal cell death pathways and cellular senescence. Additionally, NCT has been observed to be progressively dysregulated during aging and in neurodegeneration, where the increased subcellular redistribution of nuclear proteins, such as TAR DNA-Binding Protein-43 (TDP-43), to the cytoplasm is a primary driver of disease. However, the functional significance of NCT defects as either a causal mechanism or consequence of pathology, and how the redistribution of cell cycle machinery contributes to neurodegeneration, remains unclear. Here, we describe that pharmacological inhibition of importin-β nuclear import is capable of perturbing cell cycle machinery both in mitotic neuronal cell lines and post-mitotic primary neurons in vitro. Our NemfR86S mouse model of motor neuron disease, characterized by nuclear import defects, further recapitulates the hallmarks of CCD we observed in mitotic cell lines and in post-mitotic primary neurons in vitro, and in spinal motor neurons in vivo. The observed CCD is consistent with the transcriptional and phenotypical dysregulation commonly associated with neuronal cell death and senescence-like features in NDDs. Together, this evidence suggests that impairment of nuclear import pathways resulting in CCD may be a common driver of pathology in neurodegeneration.
Collapse
Affiliation(s)
- Jonathan Plessis-Belair
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, New York, USA
| | - Taylor Russo
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, New York, USA
| | - Markus Riessland
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, New York, USA
| | - Roger B Sher
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
4
|
Bi J, Guo Q, Gong Y, Chen X, Wu H, Song L, Xu Y, Ou M, Wang Z, Chen J, Jiang C, Liu A, Li G, Zhang G. Troglitazone Reduction of Intracellular Mycobacterium tuberculosis Survival Via Macrophage Autophagy Through LKB1-AMPKα Signaling. J Infect Dis 2025; 231:e553-e565. [PMID: 39450555 PMCID: PMC11911799 DOI: 10.1093/infdis/jiae523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 10/07/2024] [Accepted: 10/23/2024] [Indexed: 10/26/2024] Open
Abstract
Tuberculosis caused by Mycobacterium tuberculosis (Mtb), results in significant disease and death worldwide. Host-directed therapy, including conventional drugs, is a promising antituberculosis strategy that shows synergistic antibacterial effects when combined with antituberculosis drugs. Here, the mycobactericidal effect of 3 antidiabetic drugs was examined. Of these, only troglitazone (Trog) enhanced the antimycobacterial effect in vitro and in vivo. This was due to Trog-mediated autophagy activation. Moreover, a knock-down experiment revealed that Trog activated autophagy and exhibited antimycobacterial activity through the serine/threonine-protein kinase STK11 (LKB1)-5'-AMP-activated protein kinase (AMPK) signaling pathway. Molecular docking and coimmunoprecipitation experiments demonstrated that Trog promoted LKB1 phosphorylation and activation by targeting STE20-related kinase adapter protein alpha (STRADA). Finally, we found that Trog inhibited the intracellular survival of clinical isoniazid-resistant Mtb, and the combination of Trog and isoniazid showed additive antibacterial effects against Mtb H37Rv. Taken together, antidiabetic Trog may be repurposed as a candidate for host-directed therapy and combined with first-line antituberculosis drugs.
Collapse
Affiliation(s)
- Jing Bi
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Qinglong Guo
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Yaqi Gong
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Xi Chen
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Haojia Wu
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Li Song
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Yating Xu
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Min Ou
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Zhaoqin Wang
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Jiean Chen
- Shenzhen Bay Laboratory, Pingshan Translational Medicine Center, Shenzhen, China
| | - Chenran Jiang
- Shenzhen Bay Laboratory, Pingshan Translational Medicine Center, Shenzhen, China
| | - Aimei Liu
- Department of Tuberculosis, Guangxi Chest Hospital, Liuzhou, China
| | - Guobao Li
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Guoliang Zhang
- National Clinical Research Center for Infectious Diseases, Guangdong Provincial Clinical Research Center for Tuberculosis, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
5
|
Basheer HA, Salman NM, Abdullah RM, Elsalem L, Afarinkia K. Metformin and glioma: Targeting metabolic dysregulation for enhanced therapeutic outcomes. Transl Oncol 2025; 53:102323. [PMID: 39970627 DOI: 10.1016/j.tranon.2025.102323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/09/2024] [Accepted: 02/13/2025] [Indexed: 02/21/2025] Open
Abstract
Glioma, a highly aggressive form of brain cancer, continues to pose significant therapeutic challenges in the field of medicine. Its invasive nature and resistance to traditional treatments make it particularly difficult to combat. This review examines the potential of metformin, a commonly prescribed antidiabetic medication, as a promising new treatment option for glioma. The potential of metformin to target crucial metabolic pathways in cancer cells presents an encouraging approach to improve therapeutic outcomes. The review explores the complexities of metabolic reprogramming in glioma and metformin's role in inhibiting these metabolic pathways. Preclinical studies demonstrate metformin's efficacy in reducing tumor growth and enhancing the sensitivity of glioma cells to chemotherapy and radiotherapy. Furthermore, clinical studies highlight metformin's potential in improving progression-free survival and overall survival rates in glioma patients. The review also addresses the synergistic effects of combining metformin with other therapeutic agents, such as temozolomide and radiotherapy, to overcome drug resistance and improve treatment efficacy. Despite the promising findings, the review acknowledges the need for further clinical trials to establish optimal dosing regimens, understand the molecular mechanisms underlying metformin's antitumor effects, and identify patient populations that would benefit the most from metformin-based therapies. Additionally, the potential side effects and the long-term impact of metformin on Glioma patients require careful evaluation. In conclusion, this review underscores the potential of metformin as a repurposed drug in glioma treatment, emphasizing its multifaceted role in targeting metabolic dysregulation. Metformin holds promise as part of a combination therapy approach to improve the therapeutic landscape of glioma and offers hope for better patient outcomes.
Collapse
Affiliation(s)
- Haneen A Basheer
- Department of Pharmacy, Faculty of Pharmacy, Zarqa University, Zarqa 13110, Jordan.
| | - Nadeem M Salman
- Department of Pharmacy, Faculty of Pharmacy, Zarqa University, Zarqa 13110, Jordan
| | - Rami M Abdullah
- Department of Pharmacy, Faculty of Pharmacy, Zarqa University, Zarqa 13110, Jordan
| | - Lina Elsalem
- Jordan University of Science and Technology, Faculty of Medicine, Department of Pharmacology, Irbid, Jordan
| | - Kamyar Afarinkia
- School of Medicine and Biosciences, University of West London, London W5 5RF, UK
| |
Collapse
|
6
|
Strang J, Astridge DD, Nguyen VT, Reigan P. Small Molecule Modulators of AMP-Activated Protein Kinase (AMPK) Activity and Their Potential in Cancer Therapy. J Med Chem 2025; 68:2238-2254. [PMID: 39879193 PMCID: PMC11831681 DOI: 10.1021/acs.jmedchem.4c02354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/02/2025] [Accepted: 01/17/2025] [Indexed: 01/31/2025]
Abstract
AMP-activated protein kinase (AMPK) is a central mediator of cellular metabolism and is activated in direct response to low ATP levels. Activated AMPK inhibits anabolic pathways and promotes catabolic activities that generate ATP through the phosphorylation of multiple target substrates. AMPK is a therapeutic target for activation in several chronic metabolic diseases, and there is increasing interest in targeting AMPK activity in cancer where it can act as a tumor suppressor or conversely it can support cancer cell survival. Small molecule AMPK activators and inhibitors have demonstrated some success in suppressing cancer growth, survival, and drug resistance in preclinical cancer models. In this perspective, we summarize the role of AMPK in cancer and drug resistance, the influence of the tumor microenvironment on AMPK activity, and AMPK activator and inhibitor development. In addition, we discuss the potential importance of isoform-selective targeting of AMPK and approaches for selective AMPK targeting in cancer.
Collapse
Affiliation(s)
- Juliet
E. Strang
- Department
of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical
Sciences, University of Colorado Anschutz
Medical Campus, 12850 East Montview Boulevard, Aurora, Colorado 80045, United States
| | - Daniel D. Astridge
- Department
of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical
Sciences, University of Colorado Anschutz
Medical Campus, 12850 East Montview Boulevard, Aurora, Colorado 80045, United States
| | - Vu T. Nguyen
- Department
of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical
Sciences, University of Colorado Anschutz
Medical Campus, 12850 East Montview Boulevard, Aurora, Colorado 80045, United States
| | - Philip Reigan
- Department
of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical
Sciences, University of Colorado Anschutz
Medical Campus, 12850 East Montview Boulevard, Aurora, Colorado 80045, United States
| |
Collapse
|
7
|
Arbogast F, Sal-Carro R, Boufenghour W, Frenger Q, Bouis D, Filippi De La Palavesa L, Fauny JD, Griso O, Puccio H, Fima R, Huby T, Gautier EL, Molitor A, Carapito R, Bahram S, Romani N, Clausen BE, Voisin B, Mueller CG, Gros F, Flacher V. Epidermal maintenance of Langerhans cells relies on autophagy-regulated lipid metabolism. J Cell Biol 2025; 224:e202403178. [PMID: 39535446 PMCID: PMC11561468 DOI: 10.1083/jcb.202403178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 09/12/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Macroautophagy (often-named autophagy), a catabolic process involving autophagy-related (Atg) genes, prevents the accumulation of harmful cytoplasmic components and mobilizes energy reserves in long-lived and self-renewing cells. Autophagy deficiency affects antigen presentation in conventional dendritic cells (DCs) without impacting their survival. However, previous studies did not address epidermal Langerhans cells (LCs). Here, we demonstrate that deletion of either Atg5 or Atg7 in LCs leads to their gradual depletion. ATG5-deficient LCs showed metabolic dysregulation and accumulated neutral lipids. Despite increased mitochondrial respiratory capacity, they were unable to process lipids, eventually leading them to ferroptosis. Finally, metabolically impaired LCs upregulated proinflammatory transcripts and showed decreased expression of neuronal interaction receptors. Altogether, autophagy represents a critical regulator of lipid storage and metabolism in LCs, allowing their maintenance in the epidermis.
Collapse
Affiliation(s)
- Florent Arbogast
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Raquel Sal-Carro
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Wacym Boufenghour
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | | | - Delphine Bouis
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Louise Filippi De La Palavesa
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Jean-Daniel Fauny
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Olivier Griso
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U1258/CNRS UMR7104, Illkirch, France
| | - Hélène Puccio
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U1258/CNRS UMR7104, Illkirch, France
| | - Rebecca Fima
- Sorbonne Université, INSERM UMR_S 1166 ICAN, Paris, France
| | - Thierry Huby
- Sorbonne Université, INSERM UMR_S 1166 ICAN, Paris, France
| | | | - Anne Molitor
- Laboratoire d’Immunorhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Université de Strasbourg, Strasbourg, France
- Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France
| | - Raphaël Carapito
- Laboratoire d’Immunorhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Université de Strasbourg, Strasbourg, France
- Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France
- Service d’Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Seiamak Bahram
- Laboratoire d’Immunorhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Université de Strasbourg, Strasbourg, France
- Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France
- Service d’Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Nikolaus Romani
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Björn E. Clausen
- Institute for Molecular Medicine and Paul Klein Center for Immunotherapy (PKZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Benjamin Voisin
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Christopher G. Mueller
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Frédéric Gros
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Vincent Flacher
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| |
Collapse
|
8
|
Plessis-Belair J, Russo T, Riessland M, Sher RB. Nuclear Import Defects Drive Cell Cycle Dysregulation in Neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635269. [PMID: 39975276 PMCID: PMC11838365 DOI: 10.1101/2025.01.28.635269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Neurodegenerative diseases (NDDs) and other age-related disorders have been classically defined by a set of key pathological hallmarks. Two of these hallmarks, cell cycle dysregulation (CCD) and nucleocytoplasmic transport (NCT) defects, have long been debated as being either causal or consequential in the pathology of accelerated aging. Specifically, aberrant cell cycle activation in post-mitotic neurons has been shown to trigger neuronal cell death pathways and cellular senescence. Additionally, NCT has been observed to be progressively dysregulated during aging and in neurodegeneration, where the increased subcellular redistribution of nuclear proteins such as TAR DNA-Binding Protein-43 (TDP43) to the cytoplasm is a primary driver of many NDDs. However, the functional significance of NCT defects as either a primary driver or consequence of pathology, and how the redistribution of cell cycle machinery contributes to neurodegeneration, remains unclear. Here, we describe that pharmacological inhibition of importin-β nuclear import is capable of perturbing cell cycle machinery both in mitotic neuronal cell lines and post-mitotic primary neurons in vitro. Our Nemf R86S mouse model of motor neuron disease, characterized by nuclear import defects, further recapitulates the hallmarks of CCD in mitotic cell lines and in post-mitotic primary neurons in vitro, and in spinal motor neurons in vivo. The observed CCD is consistent with the transcriptional and phenotypical dysregulation observed in neuronal cell death and cellular senescence in NDDs. Together, this evidence suggests that impairment of nuclear import pathways resulting in CCD may be a common driver of pathology in neurodegeneration.
Collapse
Affiliation(s)
- Jonathan Plessis-Belair
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| | - Taylor Russo
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| | - Markus Riessland
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| | - Roger B Sher
- Department of Neurobiology and Behavior; Stony Brook University, Stony Brook, NY 11794, USA
- Center for Nervous System Disorders; Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
9
|
Zaidalkilani AT, Al‐kuraishy HM, Fahad EH, Al‐Gareeb AI, Elewa YHA, Zahran MH, Alexiou A, Papadakis M, AL‐Farga A, Batiha GE. Autophagy modulators in type 2 diabetes: A new perspective. J Diabetes 2024; 16:e70010. [PMID: 39676616 PMCID: PMC11647182 DOI: 10.1111/1753-0407.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 05/31/2024] [Accepted: 08/27/2024] [Indexed: 12/17/2024] Open
Abstract
Type 2 diabetes (T2D) is a chronic metabolic disorder caused by defective insulin signaling, insulin resistance, and impairment of insulin secretion. Autophagy is a conserved lysosomal-dependent catabolic cellular pathway involved in the pathogenesis of T2D and its complications. Basal autophagy regulates pancreatic β-cell function by enhancing insulin release and peripheral insulin sensitivity. Therefore, defective autophagy is associated with impairment of pancreatic β-cell function and the development of insulin rersistance (IR). However, over-activated autophagy increases apoptosis of pancreatic β-cells leading to pancreatic β-cell dysfunction. Hence, autophagy plays a double-edged sword role in T2D. Therefore, the use of autophagy modulators including inhibitors and activators may affect the pathogenesis of T2D. Hence, this review aims to clarify the potential role of autophagy inhibitors and activators in T2D.
Collapse
Affiliation(s)
- Ayah Talal Zaidalkilani
- Department of Nutrition, Faculty of Pharmacy and Medical SciencesUniversity of PetraAmmanJordan
| | - Hayder M. Al‐kuraishy
- Department of Clinical Pharmacology and MedicineCollege of Medicine, Al‐Mustansiriyah UniversityBaghdadIraq
| | - Esraa H. Fahad
- Department of Pharmacology and ToxicologyCollege of Pharmacy, Mustansiriyah UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and MedicineCollege of Medicine, Al‐Mustansiriyah UniversityBaghdadIraq
| | - Yaser Hosny Ali Elewa
- Department of Histology and Cytology, Faculty of Veterinary MedicineZagazig UniversityZagazigEgypt
- Faculty of Veterinary MedicineHokkaido UniversitySapporoJapan
| | | | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh UniversityMohaliPunjabIndia
- Department of Research & DevelopmentFunogenAthensGreece
- Department of Research & DevelopmentAFNP MedWienAustria
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐HerdeckeWuppertalGermany
| | - Ammar AL‐Farga
- Department of BiochemistryCollege of Science University of JeddahJeddahSaudi Arabia
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhur UniversityDamanhurAlBeheiraEgypt
| |
Collapse
|
10
|
Lee J, Geum D, Park DH, Kim JH. Molecular Targeting of Ischemic Stroke: The Promise of Naïve and Engineered Extracellular Vesicles. Pharmaceutics 2024; 16:1492. [PMID: 39771472 PMCID: PMC11678501 DOI: 10.3390/pharmaceutics16121492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 01/04/2025] Open
Abstract
Ischemic stroke (IS) remains a leading cause of mortality and long-term disability worldwide, with limited therapeutic options available. Despite the success of early interventions, such as tissue-type plasminogen activator administration and mechanical thrombectomy, many patients continue to experience persistent neurological deficits. The pathophysiology of IS is multifaceted, encompassing excitotoxicity, oxidative and nitrosative stress, inflammation, and blood-brain barrier disruption, all of which contribute to neural cell death, further complicating the treatment of IS. Recently, extracellular vesicles (EVs) secreted naturally by various cell types have emerged as promising therapeutic agents because of their ability to facilitate selective cell-to-cell communication, neuroprotection, and tissue regeneration. Furthermore, engineered EVs, designed to enhance targeted delivery and therapeutic cargo, hold the potential to improve their therapeutic benefits by mitigating neuronal damage and promoting neurogenesis and angiogenesis. This review summarizes the characteristics of EVs, the molecular mechanisms underlying IS pathophysiology, and the emerging role of EVs in IS treatment at the molecular level. This review also explores the recent advancements in EV engineering, including the incorporation of specific proteins, RNAs, or pharmacological agents into EVs to enhance their therapeutic efficacy.
Collapse
Affiliation(s)
- Jihun Lee
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea;
| | - Dongho Geum
- Department of Medical Science, College of Medicine, Korea University, Seoul 02841, Republic of Korea;
| | - Dong-Hyuk Park
- Department of Neurosurgery, Anam Hospital, College of Medicine, Korea University, Seoul 02841, Republic of Korea;
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea;
| |
Collapse
|
11
|
Wang M, Chen X, Li S, Wang L, Tang H, Pu Y, Zhang D, Fang B, Bai X. A crosstalk between autophagy and apoptosis in intracerebral hemorrhage. Front Cell Neurosci 2024; 18:1445919. [PMID: 39650799 PMCID: PMC11622039 DOI: 10.3389/fncel.2024.1445919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 10/31/2024] [Indexed: 12/11/2024] Open
Abstract
Intracerebral hemorrhage (ICH) is a severe condition that devastatingly harms human health and poses a financial burden on families and society. Bcl-2 Associated X-protein (Bax) and B-cell lymphoma 2 (Bcl-2) are two classic apoptotic markers post-ICH. Beclin 1 offers a competitive architecture with that of Bax, both playing a vital role in autophagy. However, the interaction between Beclin 1 and Bcl-2/Bax has not been conjunctively analyzed. This review aims to examine the crosstalk between autophagy and apoptosis in ICH by focusing on the interaction and balance of Beclin 1, Bax, and Bcl-2. We also explored the therapeutic potential of Western conventional medicine and traditional Chinese medicine (TCM) in ICH via controlling the crosstalk between autophagy and apoptosis.
Collapse
Affiliation(s)
- Moyan Wang
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Xin Chen
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Shuangyang Li
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Lingxue Wang
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Hongmei Tang
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Yuting Pu
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Dechou Zhang
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Bangjiang Fang
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- Department of Emergency, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xue Bai
- Department of Neurology, National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| |
Collapse
|
12
|
Smiles WJ, Ovens AJ, Kemp BE, Galic S, Petersen J, Oakhill JS. New developments in AMPK and mTORC1 cross-talk. Essays Biochem 2024; 68:321-336. [PMID: 38994736 PMCID: PMC12055038 DOI: 10.1042/ebc20240007] [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: 05/09/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024]
Abstract
Metabolic homeostasis and the ability to link energy supply to demand are essential requirements for all living cells to grow and proliferate. Key to metabolic homeostasis in all eukaryotes are AMPK and mTORC1, two kinases that sense nutrient levels and function as counteracting regulators of catabolism (AMPK) and anabolism (mTORC1) to control cell survival, growth and proliferation. Discoveries beginning in the early 2000s revealed that AMPK and mTORC1 communicate, or cross-talk, through direct and indirect phosphorylation events to regulate the activities of each other and their shared protein substrate ULK1, the master initiator of autophagy, thereby allowing cellular metabolism to rapidly adapt to energy and nutritional state. More recent reports describe divergent mechanisms of AMPK/mTORC1 cross-talk and the elaborate means by which AMPK and mTORC1 are activated at the lysosome. Here, we provide a comprehensive overview of current understanding in this exciting area and comment on new evidence showing mTORC1 feedback extends to the level of the AMPK isoform, which is particularly pertinent for some cancers where specific AMPK isoforms are implicated in disease pathogenesis.
Collapse
Affiliation(s)
- William J Smiles
- Metabolic Signalling Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Research Program for Receptor Biochemistry and Tumour Metabolism, Department of Paediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Ashley J Ovens
- Protein Engineering in Immunity and Metabolism, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Bruce E Kemp
- Protein Chemistry and Metabolism, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
- Mary Mackillop Institute for Health Research, Australian Catholic University, Fitzroy, Vic 3065, Vic. Australia
| | - Sandra Galic
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
- Metabolic Physiology, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Janni Petersen
- Flinders Health and Medical Research Institute, Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, SA 5042, Australia
- Nutrition and Metabolism, South Australia Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Jonathan S Oakhill
- Metabolic Signalling Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
13
|
Chu Y, Yuan X, Tao Y, Yang B, Luo J. Autophagy in Muscle Regeneration: Mechanisms, Targets, and Therapeutic Perspective. Int J Mol Sci 2024; 25:11901. [PMID: 39595972 PMCID: PMC11593790 DOI: 10.3390/ijms252211901] [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: 09/30/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Autophagy maintains the stability of eukaryotic cells by degrading unwanted components and recycling nutrients and plays a pivotal role in muscle regeneration by regulating the quiescence, activation, and differentiation of satellite cells. Effective muscle regeneration is vital for maintaining muscle health and homeostasis. However, under certain disease conditions, such as aging, muscle regeneration can fail due to dysfunctional satellite cells. Dysregulated autophagy may limit satellite cell self-renewal, hinder differentiation, and increase susceptibility to apoptosis, thereby impeding muscle regeneration. This review explores the critical role of autophagy in muscle regeneration, emphasizing its interplay with apoptosis and recent advances in autophagy research related to diseases characterized by impaired muscle regeneration. Additionally, we discuss new approaches involving autophagy regulation to promote macrophage polarization, enhancing muscle regeneration. We suggest that utilizing cell therapy and biomaterials to modulate autophagy could be a promising strategy for supporting muscle regeneration. We hope that this review will provide new insights into the treatment of muscle diseases and promote muscle regeneration.
Collapse
Affiliation(s)
- Yun Chu
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.C.); (Y.T.); (B.Y.)
| | - Xinrun Yuan
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Yiming Tao
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.C.); (Y.T.); (B.Y.)
| | - Bin Yang
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.C.); (Y.T.); (B.Y.)
| | - Jinlong Luo
- Department of Emergency, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| |
Collapse
|
14
|
Ghareghomi S, Arghavani P, Mahdavi M, Khatibi A, García-Jiménez C, Moosavi-Movahedi AA. Hyperglycemia-driven signaling bridges between diabetes and cancer. Biochem Pharmacol 2024; 229:116450. [PMID: 39059774 DOI: 10.1016/j.bcp.2024.116450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/21/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Growing epidemiological evidence indicates an association between obesity, type 2 diabetes, and certain cancers, suggesting the existence of common underlying mechanisms in these diseases. Frequent hyperglycemias in type 2 diabetes promote pro-inflammatory responses and stimulate intracellular metabolic flux which rewires signaling pathways and influences the onset and advancement of different types of cancers. Here, we review the provocative impact of hyperglycemia on a subset of interconnected signalling pathways that regulate (i) cell growth and survival, (ii) metabolism adjustments, (iii) protein function modulation in response to nutrient availability (iv) and cell fate and proliferation and which are driven respectively by PI3K (Phosphoinositide 3-kinase), AMPK (AMP-activated protein kinase), O-GlcNAc (O-linked N-acetylglucosamine) and Wnt/β-catenin. Specifically, we will elaborate on their involvement in glucose metabolism, inflammation, and cell proliferation, highlighting their interplay in the pathogenesis of diabetes and cancer. Furthermore, the influence of antineoplastic and antidiabetic drugs on the unbridled cellular pathways will be examined. This review aims to inspire the next molecular studies to understand how type 2 diabetes may lead to certain cancers. This will contribute to personalized medicine and direct better prevention strategies.
Collapse
Affiliation(s)
- Somayyeh Ghareghomi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran; Department of Biotechnology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Payam Arghavani
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Majid Mahdavi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Ali Khatibi
- Department of Biotechnology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran.
| | - Custodia García-Jiménez
- Department of Basic Health Sciences, Faculty of Health Sciences, University Rey Juan Carlos. Alcorcón, Madrid, Spain.
| | - Ali A Moosavi-Movahedi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran; UNESCO Chair on Interdisciplinary Research in Diabetes, University of Tehran, Tehran, Iran.
| |
Collapse
|
15
|
Niharika, Garg M. Understanding the autophagic functions in cancer stem cell maintenance and therapy resistance. Expert Rev Mol Med 2024; 26:e23. [PMID: 39375840 PMCID: PMC11488345 DOI: 10.1017/erm.2024.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/25/2023] [Accepted: 06/25/2024] [Indexed: 10/09/2024]
Abstract
Complex tumour ecosystem comprising tumour cells and its associated tumour microenvironment (TME) constantly influence the tumoural behaviour and ultimately impact therapy failure, disease progression, recurrence and poor overall survival of patients. Crosstalk between tumour cells and TME amplifies the complexity by creating metabolic changes such as hypoxic environment and nutrient fluctuations. These changes in TME initiate stem cell-like programmes in cancer cells, contribute to tumoural heterogeneity and increase tumour robustness. Recent studies demonstrate the multifaceted role of autophagy in promoting fibroblast production, stemness, cancer cell survival during longer periods of dormancy, eventual growth of metastatic disease and disease resistance. Recent ongoing studies examine autophagy/mitophagy as a powerful survival strategy in response to environmental stress including nutrient deprivation, hypoxia and environmental stress in TME. It prevents irreversible senescence, promotes dormant stem-like state, induces epithelial-mesenchymal transition and increases migratory and invasive potential of tumour cells. The present review discusses various theories and mechanisms behind the autophagy-dependent induction of cancer stem cell (CSC) phenotype. Given the role of autophagic functions in CSC aggressiveness and therapeutic resistance, various mechanisms and studies based on suppressing cellular plasticity by blocking autophagy as a powerful therapeutic strategy to kill tumour cells are discussed.
Collapse
Affiliation(s)
- Niharika
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India
| | - Minal Garg
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India
| |
Collapse
|
16
|
Ding W, Bao S, Zhao Q, Hao W, Fang K, Xiao Y, Lin X, Zhao Z, Xu X, Cui X, Yang X, Yao L, Jin H, Zhang K, Guo J. Blocking ACSL6 Compromises Autophagy via FLI1-Mediated Downregulation of COLs to Radiosensitize Lung Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403202. [PMID: 39206814 PMCID: PMC11516120 DOI: 10.1002/advs.202403202] [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: 03/26/2024] [Revised: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Lung cancer (LC) is the leading cause of cancer-related mortality worldwide. Radiotherapy is the main component of LC treatment; however, its efficacy is often limited by radioresistance development, resulting in unsatisfactory clinical outcomes. Here, we found that LC radiosensitivity is up-regulated by decreased expression of long-chain acyl-CoA synthase 6 (ACSL6) after irradiation. Deletion of ACSL6 results in significant elevation of Friend leukemia integration 1 transcription factor (FLI1) and a marked decline of collagens (COLs). Blocking of ACSL6 impairs the tumor growth and upregulates FLI1, which reduces the levels of COLs and compromises irradiation-induced autophagy, leading to considerable therapeutic benefits during radiotherapy. Moreover, the direct interaction between ACSL6 and FLI1 and engagement between FLI1 and COLs indicates the involvement of the ACSL6-FLI1-COL axis. Finally, the potently adjusted autophagy flux reduces its otherwise contributive capability in surviving irradiation stress and leads to satisfactory radiosensitization for LC radiotherapy. These results demonstrate that enhanced ACSL6 expression promotes the aggressive performance of irradiated LC through increased FLI1-COL-mediated autophagy flux. Thus, the ACSL6-FLI1-Col-autophagy axis may be targeted to enhance the radiosensitivity of LC and improve the management of LC in radiotherapy.
Collapse
Affiliation(s)
- Wen Ding
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Shijun Bao
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Qingwei Zhao
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Wei Hao
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Kai Fang
- Department of Medicine CollegeJiangnan UniversityWuxiJiangsu214000P. R. China
| | - Yanlan Xiao
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Xiaoting Lin
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Zhemeng Zhao
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Xinyi Xu
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
- College of Basic MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Xinyue Cui
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Xiwen Yang
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Liuhuan Yao
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Hai Jin
- Department of Cardiothoracic SurgeryChanghai HospitalNaval Medical UniversityShanghai200433P. R. China
| | - Kun Zhang
- Department of Laboratory Medicine and Central LaboratorySichuan Academy of Medical SciencesSichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaNo. 32, West Second Section, First Ring RoadChengduSichuan610072P. R. China
| | - Jiaming Guo
- Department of Radiation MedicineCollege of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| |
Collapse
|
17
|
Siegman A, Shaykevich A, Chae D, Silverman I, Goel S, Maitra R. Erlotinib Treatment in Colorectal Cancer Suppresses Autophagy Based on KRAS Mutation. Curr Issues Mol Biol 2024; 46:7530-7547. [PMID: 39057088 PMCID: PMC11276370 DOI: 10.3390/cimb46070447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
The KRAS gene is mutated in approximately 45% of colorectal cancer patients. There are currently very few targeted treatments or therapies equipped to directly inhibit KRAS due to its unusual structural intricacies. Erlotinib, an EGFR inhibitor, has previously been demonstrated to reduce cell viability by inducing autophagy in lung cancer cell lines with varying EGFR mutations. In contrast to lung cancer cells, evidence is provided herein for the first time that erlotinib treatment in colorectal cancer (CRC) cell lines reduces autophagy and still results in decreased cell viability. However, the effects of erlotinib in CRC cell lines containing a wildtype KRAS gene were different than in cells carrying a mutant KRAS gene. We show that there is significantly more downregulation of autophagy in KRAS mutant CRC cells compared to KRAS wildtype cells, both at transcriptional and translational levels, suggesting that the KRAS mutation is advantageous for cancer growth, even in the presence of erlotinib. Cell viability results determined that KRAS wildtype CRC cells had significantly more cell death compared to KRAS mutant cells. Using patient mRNA datasets, we showed that there was a significant correlation between the presence of the KRAS mutation and the expression of autophagy proteins. Additionally, through molecular dynamics simulations, we develop a blueprint for KRAS and autophagy protein interaction and the impact of the KRAS mutation on autophagy protein regulation. Overall, this is the first report of erlotinib treatment in CRC cells that assesses autophagy, and we demonstrate that autophagy activity is downregulated in these cells. This effect is not only greater in cells carrying a KRAS mutation compared to wildtype cells, but the KRAS mutant cells also have increased cell viability compared to wildtype cells. We hypothesize that the difference in cell viability and autophagy expression between KRAS mutant and KRAS wildtype cells after treatment with erlotinib can be of therapeutic value to treat CRC patients carrying KRAS mutations.
Collapse
Affiliation(s)
- Alexander Siegman
- Department of Biology, Yeshiva University, New York, NY 10033, USA (D.C.)
| | - Aaron Shaykevich
- Department of Biology, Yeshiva University, New York, NY 10033, USA (D.C.)
| | - Danbee Chae
- Department of Biology, Yeshiva University, New York, NY 10033, USA (D.C.)
| | - Isaac Silverman
- Department of Biology, Yeshiva University, New York, NY 10033, USA (D.C.)
| | - Sanjay Goel
- Department of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Radhashree Maitra
- Department of Biology, Yeshiva University, New York, NY 10033, USA (D.C.)
| |
Collapse
|
18
|
Madsen JF, Ernst EH, Amoushahi M, Dueholm M, Ernst E, Lykke-Hartmann K. Dorsomorphin inhibits AMPK, upregulates Wnt and Foxo genes and promotes the activation of dormant follicles. Commun Biol 2024; 7:747. [PMID: 38902324 PMCID: PMC11190264 DOI: 10.1038/s42003-024-06418-9] [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: 06/22/2023] [Accepted: 06/06/2024] [Indexed: 06/22/2024] Open
Abstract
AMPK is a well-known energy sensor regulating cellular metabolism. Metabolic disorders such as obesity and diabetes are considered detrimental factors that reduce fecundity. Here, we show that pharmacologically induced in vitro activation (by metformin) or inhibition (by dorsomorphin) of the AMPK pathway inhibits or promotes activation of ovarian primordial follicles in cultured murine ovaries and human ovarian cortical chips. In mice, activation of primordial follicles in dorsomorphin in vitro-treated ovaries reduces AMPK activation and upregulates Wnt and FOXO genes, which, interestingly, is associated with decreased phosphorylation of β-catenin. The dorsomorphin-treated ovaries remain of high quality, with no detectable difference in reactive oxygen species production, apoptosis or mitochondrial cytochrome c oxidase activity, suggesting safe activation. Subsequent maturation of in vitro-treated follicles, using a 3D alginate cell culture system, results in mature metaphase eggs with protruding polar bodies. These findings demonstrate that the AMPK pathway can safely regulate primordial follicles by modulating Wnt and FOXO genes, and reduce β-catenin phosphorylation.
Collapse
Affiliation(s)
- Julie Feld Madsen
- Department of Biomedicine, Aarhus University, DK-8000, Aarhus C, Denmark
| | - Emil Hagen Ernst
- Department of Obstetrics and Gynaecology, Aarhus University Hospital, DK-8000, Aarhus C, Denmark
- Department of Gynaecology and Obstetrics, Gødstrup Hospital, DK-7400, Herning, Denmark
| | | | - Margit Dueholm
- Department of Obstetrics and Gynaecology, Aarhus University Hospital, DK-8000, Aarhus C, Denmark
| | - Erik Ernst
- Department of Biomedicine, Aarhus University, DK-8000, Aarhus C, Denmark
- Fertility Clinic Regional Hospital Horsens, DK-8700, Horsens, Denmark
| | - Karin Lykke-Hartmann
- Department of Biomedicine, Aarhus University, DK-8000, Aarhus C, Denmark.
- Department of Clinical Genetics, Aarhus University Hospital, DK-8200, Aarhus N, Denmark.
| |
Collapse
|
19
|
Razavipour SF, Yoon H, Jang K, Kim M, Nawara HM, Bagheri A, Huang WC, Shin M, Zhao D, Zhou Z, Van Boven D, Briegel K, Morey L, Ince TA, Johnson M, Slingerland JM. C-terminally phosphorylated p27 activates self-renewal driver genes to program cancer stem cell expansion, mammary hyperplasia and cancer. Nat Commun 2024; 15:5152. [PMID: 38886396 PMCID: PMC11183067 DOI: 10.1038/s41467-024-48742-y] [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: 07/14/2023] [Accepted: 05/14/2024] [Indexed: 06/20/2024] Open
Abstract
In many cancers, a stem-like cell subpopulation mediates tumor initiation, dissemination and drug resistance. Here, we report that cancer stem cell (CSC) abundance is transcriptionally regulated by C-terminally phosphorylated p27 (p27pT157pT198). Mechanistically, this arises through p27 co-recruitment with STAT3/CBP to gene regulators of CSC self-renewal including MYC, the Notch ligand JAG1, and ANGPTL4. p27pTpT/STAT3 also recruits a SIN3A/HDAC1 complex to co-repress the Pyk2 inhibitor, PTPN12. Pyk2, in turn, activates STAT3, creating a feed-forward loop increasing stem-like properties in vitro and tumor-initiating stem cells in vivo. The p27-activated gene profile is over-represented in STAT3 activated human breast cancers. Furthermore, mammary transgenic expression of phosphomimetic, cyclin-CDK-binding defective p27 (p27CK-DD) increases mammary duct branching morphogenesis, yielding hyperplasia and microinvasive cancers that can metastasize to liver, further supporting a role for p27pTpT in CSC expansion. Thus, p27pTpT interacts with STAT3, driving transcriptional programs governing stem cell expansion or maintenance in normal and cancer tissues.
Collapse
Affiliation(s)
- Seyedeh Fatemeh Razavipour
- Cancer Host Interactions Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC, USA
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
| | - Hyunho Yoon
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon-si, South Korea
| | - Kibeom Jang
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
| | - Minsoon Kim
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
| | - Hend M Nawara
- Cancer Host Interactions Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC, USA
| | - Amir Bagheri
- Cancer Host Interactions Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC, USA
| | - Wei-Chi Huang
- Cancer Host Interactions Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC, USA
| | - Miyoung Shin
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Dekuang Zhao
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
| | - Zhiqun Zhou
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
| | - Derek Van Boven
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Karoline Briegel
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, Fl, USA
| | - Lluis Morey
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA
- John P. Hussman Institute for Human Genomics, Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Tan A Ince
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Michael Johnson
- Cancer Host Interactions Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC, USA
| | - Joyce M Slingerland
- Cancer Host Interactions Program, Lombardi Comprehensive Cancer Center, Department of Oncology, Georgetown University, Washington DC, USA.
- Braman Family Breast Cancer Institute, Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Fl, USA.
| |
Collapse
|
20
|
Huang L, Guo H. Acetylation modification in the regulation of macroautophagy. ADVANCED BIOTECHNOLOGY 2024; 2:19. [PMID: 39883319 PMCID: PMC11740868 DOI: 10.1007/s44307-024-00027-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 01/31/2025]
Abstract
Macroautophagy, commonly referred to as autophagy, is an evolutionarily conserved cellular process that plays a crucial role in maintaining cellular homeostasis. It orchestrates the delivery of dysfunctional or surplus cellular materials to the vacuole or lysosome for degradation and recycling, particularly during adverse conditions. Over the past few decades, research has unveiled intricate regulatory mechanisms governing autophagy through various post-translational modifications (PTMs). Among these PTMs, acetylation modification has emerged as a focal point in yeast and animal studies. It plays a pivotal role in autophagy by directly targeting core components within the central machinery of autophagy, including autophagy initiation, nucleation, phagophore expansion, and autophagosome maturation. Additionally, acetylation modulates autophagy at the transcriptional level by modifying histones and transcription factors. Despite its well-established significance in yeast and mammals, the role of acetylation in plant autophagy remains largely unexplored, and the precise regulatory mechanisms remain enigmatic. In this comprehensive review, we summarize the current understanding of the function and underlying mechanisms of acetylation in regulating autophagy across yeast, mammals, and plants. We particularly highlight recent advances in deciphering the impact of acetylation on plant autophagy. These insights not only provide valuable guidance but also inspire further scientific inquiries into the intricate role of acetylation in plant autophagy.
Collapse
Affiliation(s)
- Li Huang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China
| | - Hongwei Guo
- Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Institute of Plant and Food Science, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
21
|
Trelford CB, Shepherd TG. LKB1 biology: assessing the therapeutic relevancy of LKB1 inhibitors. Cell Commun Signal 2024; 22:310. [PMID: 38844908 PMCID: PMC11155146 DOI: 10.1186/s12964-024-01689-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024] Open
Abstract
Liver Kinase B1 (LKB1), encoded by Serine-Threonine Kinase 11 (STK11), is a master kinase that regulates cell migration, polarity, proliferation, and metabolism through downstream adenosine monophosphate-activated protein kinase (AMPK) and AMPK-related kinase signalling. Since genetic screens identified STK11 mutations in Peutz-Jeghers Syndrome, STK11 mutants have been implicated in tumourigenesis labelling it as a tumour suppressor. In support of this, several compounds reduce tumour burden through upregulating LKB1 signalling, and LKB1-AMPK agonists are cytotoxic to tumour cells. However, in certain contexts, its role in cancer is paradoxical as LKB1 promotes tumour cell survival by mediating resistance against metabolic and oxidative stressors. LKB1 deficiency has also enhanced the selectivity and cytotoxicity of several cancer therapies. Taken together, there is a need to develop LKB1-specific pharmacological compounds, but prior to developing LKB1 inhibitors, further work is needed to understand LKB1 activity and regulation. However, investigating LKB1 activity is strenuous as cell/tissue type, mutations to the LKB1 signalling pathway, STE-20-related kinase adaptor protein (STRAD) binding, Mouse protein 25-STRAD binding, splicing variants, nucleocytoplasmic shuttling, post-translational modifications, and kinase conformation impact the functional status of LKB1. For these reasons, guidelines to standardize experimental strategies to study LKB1 activity, associate proteins, spliced isoforms, post-translational modifications, and regulation are of upmost importance to the development of LKB1-specific therapies. Therefore, to assess the therapeutic relevancy of LKB1 inhibitors, this review summarizes the importance of LKB1 in cell physiology, highlights contributors to LKB1 activation, and outlines the benefits and risks associated with targeting LKB1.
Collapse
Affiliation(s)
- Charles B Trelford
- The Mary &, John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, 790 Commissioners Road East, Room A4‑921, London, ON, N6A 4L6, Canada.
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.
| | - Trevor G Shepherd
- The Mary &, John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, 790 Commissioners Road East, Room A4‑921, London, ON, N6A 4L6, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| |
Collapse
|
22
|
Yang C, Rubin L, Yu X, Lazarovici P, Zheng W. Preclinical evidence using synthetic compounds and natural products indicates that AMPK represents a potential pharmacological target for the therapy of pulmonary diseases. Med Res Rev 2024; 44:1326-1369. [PMID: 38229486 DOI: 10.1002/med.22014] [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: 06/05/2023] [Revised: 12/07/2023] [Accepted: 12/30/2023] [Indexed: 01/18/2024]
Abstract
Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) is a highly conserved eukaryotic enzyme discovered as a key regulator of cellular energy homeostasis, with anti-inflammation, antioxidative stress, anticancer, and antifibrosis beneficial effects. AMPK is dysregulated in human pulmonary diseases such as acute lung injury, nonsmall cell lung cancer, pulmonary fibrosis, chronic obstructive pulmonary disease, and asthma. This review provides an overview of the beneficial role of natural, synthetic, and Chinese traditional medicines AMPK modulators in pulmonary diseases, and highlights the role of the AMPK signaling pathway in the lung, emphasizing the importance of finding lead compounds and drugs that can target and modulate AMPK to treat the lung diseases.
Collapse
Affiliation(s)
- Chao Yang
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Limor Rubin
- Allergy and Clinical Immunology Unit, Department of Medicine, Jerusalem, Israel
| | - Xiyong Yu
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wenhua Zheng
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
23
|
Galal MA, Al-Rimawi M, Hajeer A, Dahman H, Alouch S, Aljada A. Metformin: A Dual-Role Player in Cancer Treatment and Prevention. Int J Mol Sci 2024; 25:4083. [PMID: 38612893 PMCID: PMC11012626 DOI: 10.3390/ijms25074083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
Cancer continues to pose a significant global health challenge, as evidenced by the increasing incidence rates and high mortality rates, despite the advancements made in chemotherapy. The emergence of chemoresistance further complicates the effectiveness of treatment. However, there is growing interest in the potential of metformin, a commonly prescribed drug for type 2 diabetes mellitus (T2DM), as an adjuvant chemotherapy agent in cancer treatment. Although the precise mechanism of action of metformin in cancer therapy is not fully understood, it has been found to have pleiotropic effects, including the modulation of metabolic pathways, reduction in inflammation, and the regulation of cellular proliferation. This comprehensive review examines the anticancer properties of metformin, drawing insights from various studies conducted in vitro and in vivo, as well as from clinical trials and observational research. This review discusses the mechanisms of action involving both insulin-dependent and independent pathways, shedding light on the potential of metformin as a therapeutic agent for different types of cancer. Despite promising findings, there are challenges that need to be addressed, such as conflicting outcomes in clinical trials, considerations regarding dosing, and the development of resistance. These challenges highlight the importance of further research to fully harness the therapeutic potential of metformin in cancer treatment. The aims of this review are to provide a contemporary understanding of the role of metformin in cancer therapy and identify areas for future exploration in the pursuit of effective anticancer strategies.
Collapse
Affiliation(s)
- Mariam Ahmed Galal
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1QU, UK
| | - Mohammed Al-Rimawi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | | | - Huda Dahman
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | - Samhar Alouch
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| | - Ahmad Aljada
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia; (M.A.G.); (M.A.-R.); (H.D.); (S.A.)
| |
Collapse
|
24
|
Bildik G, Gray JP, Mao W, Yang H, Ozyurt R, Orellana VR, De Wever O, Carey MS, Bast RC, Lu Z. DIRAS3 induces autophagy and enhances sensitivity to anti-autophagic therapy in KRAS-driven pancreatic and ovarian carcinomas. Autophagy 2024; 20:675-691. [PMID: 38169324 PMCID: PMC10936598 DOI: 10.1080/15548627.2023.2299516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 12/06/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) and low-grade ovarian cancer (LGSOC) are characterized by the prevalence of KRAS oncogene mutations. DIRAS3 is the first endogenous non-RAS protein that heterodimerizes with RAS, disrupts RAS clustering, blocks RAS signaling, and inhibits cancer cell growth. Here, we found that DIRAS3-mediated KRAS inhibition induces ROS-mediated apoptosis in PDAC and LGSOC cells with KRAS mutations, but not in cells with wild-type KRAS, by downregulating NFE2L2/Nrf2 transcription, reducing antioxidants, and inducing oxidative stress. DIRAS3 also induces cytoprotective macroautophagy/autophagy that may protect mutant KRAS cancer cells from oxidative stress, by inhibiting mutant KRAS, activating the STK11/LKB1-PRKAA/AMPK pathway, increasing lysosomal CDKN1B/p27 localization, and inducing autophagic gene expression. Treatment with chloroquine or the novel dimeric chloroquine analog DC661 significantly enhances DIRAS3-mediated inhibition of mutant KRAS tumor cell growth in vitro and in vivo. Taken together, our study demonstrates that DIRAS3 plays a critical role in regulating mutant KRAS-driven oncogenesis in PDAC and LGSOC.Abbreviations: AFR: autophagic flux reporter; ATG: autophagy related; CQ: chloroquine; DCFDA: 2'-7'-dichlorodihydrofluorescein diacetate; DIRAS3: DIRAS family GTPase 3; DOX: doxycycline; KRAS: KRAS proto-oncogene, LGSOC: low-grade serous ovarian cancer; MiT/TFE: microphthalmia family of transcription factors; NAC: N-acetylcysteine; PDAC: pancreatic ductal adenocarcinoma; ROS: reactive oxygen species; TFEB: transcription factor EB.
Collapse
Affiliation(s)
- Gamze Bildik
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joshua P. Gray
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Weiqun Mao
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hailing Yang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rumeysa Ozyurt
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vivian R. Orellana
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Olivier De Wever
- Laboratory of Experimental Cancer Research, Cancer Research Institute Ghent, Belgium; Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Mark S. Carey
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Robert C. Bast
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhen Lu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
25
|
Bosso M, Haddad D, Al Madhoun A, Al-Mulla F. Targeting the Metabolic Paradigms in Cancer and Diabetes. Biomedicines 2024; 12:211. [PMID: 38255314 PMCID: PMC10813379 DOI: 10.3390/biomedicines12010211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Dysregulated metabolic dynamics are evident in both cancer and diabetes, with metabolic alterations representing a facet of the myriad changes observed in these conditions. This review delves into the commonalities in metabolism between cancer and type 2 diabetes (T2D), focusing specifically on the contrasting roles of oxidative phosphorylation (OXPHOS) and glycolysis as primary energy-generating pathways within cells. Building on earlier research, we explore how a shift towards one pathway over the other serves as a foundational aspect in the development of cancer and T2D. Unlike previous reviews, we posit that this shift may occur in seemingly opposing yet complementary directions, akin to the Yin and Yang concept. These metabolic fluctuations reveal an intricate network of underlying defective signaling pathways, orchestrating the pathogenesis and progression of each disease. The Warburg phenomenon, characterized by the prevalence of aerobic glycolysis over minimal to no OXPHOS, emerges as the predominant metabolic phenotype in cancer. Conversely, in T2D, the prevailing metabolic paradigm has traditionally been perceived in terms of discrete irregularities rather than an OXPHOS-to-glycolysis shift. Throughout T2D pathogenesis, OXPHOS remains consistently heightened due to chronic hyperglycemia or hyperinsulinemia. In advanced insulin resistance and T2D, the metabolic landscape becomes more complex, featuring differential tissue-specific alterations that affect OXPHOS. Recent findings suggest that addressing the metabolic imbalance in both cancer and diabetes could offer an effective treatment strategy. Numerous pharmaceutical and nutritional modalities exhibiting therapeutic effects in both conditions ultimately modulate the OXPHOS-glycolysis axis. Noteworthy nutritional adjuncts, such as alpha-lipoic acid, flavonoids, and glutamine, demonstrate the ability to reprogram metabolism, exerting anti-tumor and anti-diabetic effects. Similarly, pharmacological agents like metformin exhibit therapeutic efficacy in both T2D and cancer. This review discusses the molecular mechanisms underlying these metabolic shifts and explores promising therapeutic strategies aimed at reversing the metabolic imbalance in both disease scenarios.
Collapse
Affiliation(s)
- Mira Bosso
- Department of Pathology, Faculty of Medicine, Health Science Center, Kuwait University, Safat 13110, Kuwait
| | - Dania Haddad
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
| | - Ashraf Al Madhoun
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
- Department of Animal and Imaging Core Facilities, Dasman Diabetes Institute, Dasman 15462, Kuwait
| | - Fahd Al-Mulla
- Department of Pathology, Faculty of Medicine, Health Science Center, Kuwait University, Safat 13110, Kuwait
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman 15462, Kuwait; (D.H.); (A.A.M.)
| |
Collapse
|
26
|
Marzoog BA. Autophagy as an Anti-senescent in Aging Neurocytes. Curr Mol Med 2024; 24:182-190. [PMID: 36683318 DOI: 10.2174/1566524023666230120102718] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 01/24/2023]
Abstract
Neuron homeostasis is crucial for the organism, and its maintenance is multifactorial, including autophagy. The turnover of aberrant intracellular components is a fundamental pathogenetic mechanism for cell aging. Autophagy is involved in the acceleration of the neurocyte aging process and the modification of cell longevity. Neurocyte aging is a process of loss of cell identity through cellular and subcellular changes that include molecular loss of epigenetics, transcriptomic, proteomic, and autophagy dysfunction. Autophagy dysfunction is the hallmark of neurocyte aging. Cell aging is the credential feature of neurodegenerative diseases. Pathophysiologically, aged neurocytes are characterized by dysregulated autophagy and subsequently neurocyte metabolic stress, resulting in accelerated neurocyte aging. In particular, chaperone- mediated autophagy perturbation results in upregulated expression of aging and apoptosis genes. Aged neurocytes are also characterized by the down-regulation of autophagy-related genes, such as ATG5-ATG12, LC3-II / LC3-I ratio, Beclin-1, and p62. Slowing aging through autophagy targeting is sufficient to improve prognosis in neurodegenerative diseases. Three primary anti-senescent molecules are involved in the aging process: mTOR, AMPK, and Sirtuins. Autophagy therapeutic effects can be applied to reverse and slow aging. This article discusses current advances in the role of autophagy in neurocyte homeostasis, aging, and potential therapeutic strategies to reduce aging and increase cell longevity.
Collapse
Affiliation(s)
- Basheer Abdullah Marzoog
- National Research Mordovia State University, Bolshevitskaya Street, 68, Saransk, 430005, Rep. Mordovia, Russia
| |
Collapse
|
27
|
Isop LM, Neculau AE, Necula RD, Kakucs C, Moga MA, Dima L. Metformin: The Winding Path from Understanding Its Molecular Mechanisms to Proving Therapeutic Benefits in Neurodegenerative Disorders. Pharmaceuticals (Basel) 2023; 16:1714. [PMID: 38139841 PMCID: PMC10748332 DOI: 10.3390/ph16121714] [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: 09/12/2023] [Revised: 11/25/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Metformin, a widely prescribed medication for type 2 diabetes, has garnered increasing attention for its potential neuroprotective properties due to the growing demand for treatments for Alzheimer's, Parkinson's, and motor neuron diseases. This review synthesizes experimental and clinical studies on metformin's mechanisms of action and potential therapeutic benefits for neurodegenerative disorders. A comprehensive search of electronic databases, including PubMed, MEDLINE, Embase, and Cochrane library, focused on key phrases such as "metformin", "neuroprotection", and "neurodegenerative diseases", with data up to September 2023. Recent research on metformin's glucoregulatory mechanisms reveals new molecular targets, including the activation of the LKB1-AMPK signaling pathway, which is crucial for chronic administration of metformin. The pleiotropic impact may involve other stress kinases that are acutely activated. The precise role of respiratory chain complexes (I and IV), of the mitochondrial targets, or of the lysosomes in metformin effects remains to be established by further research. Research on extrahepatic targets like the gut and microbiota, as well as its antioxidant and immunomodulatory properties, is crucial for understanding neurodegenerative disorders. Experimental data on animal models shows promising results, but clinical studies are inconclusive. Understanding the molecular targets and mechanisms of its effects could help design clinical trials to explore and, hopefully, prove its therapeutic effects in neurodegenerative conditions.
Collapse
Affiliation(s)
- Laura Mihaela Isop
- Department of Fundamental, Prophylactic and Clinical Sciences, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania; (L.M.I.)
| | - Andrea Elena Neculau
- Department of Fundamental, Prophylactic and Clinical Sciences, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania; (L.M.I.)
| | - Radu Dan Necula
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania
| | - Cristian Kakucs
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania
| | - Marius Alexandru Moga
- Department of Medical and Surgical Specialties, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania
| | - Lorena Dima
- Department of Fundamental, Prophylactic and Clinical Sciences, Faculty of Medicine, Transilvania University of Brasov, 500036 Brașov, Romania; (L.M.I.)
| |
Collapse
|
28
|
Latt KZ, Yoshida T, Shrivastav S, Abedini A, Reece JM, Sun Z, Lee H, Okamoto K, Dagur P, Heymann J, Zhao Y, Chung JY, Hewitt S, Jose PA, Lee K, He JC, Winkler CA, Knepper MA, Kino T, Rosenberg AZ, Susztak K, Kopp JB. HIV viral protein R induces loss of DCT1-type renal tubules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.02.526686. [PMID: 36945458 PMCID: PMC10028744 DOI: 10.1101/2023.02.02.526686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hyponatremia and salt wasting is a common occurance in patients with HIV/AIDS, however, the understanding of its contributing factors is limited. HIV viral protein R (Vpr) contributes to HIV-associated nephropathy. To investigate the effects of Vpr on the expression level of the Slc12a3 gene, encoding the Na-Cl cotransporter, which is responsible for sodium reabsorption in distal nephron segments, we performed single-nucleus RNA sequencing of kidney cortices from three wild-type (WT) and three Vpr-transgenic (Vpr Tg) mice. The results showed that the percentage of distal convoluted tubule (DCT) cells was significantly lower in Vpr Tg mice compared with WT mice (P < 0.05), and that in Vpr Tg mice, Slc12a3 expression was not different in DCT cell cluster. The Pvalb+ DCT1 subcluster had fewer cells in Vpr Tg mice compared with WT (P < 0.01). Immunohistochemistry demonstrated fewer Slc12a3+ Pvalb+ DCT1 segments in Vpr Tg mice. Differential gene expression analysis comparing Vpr Tg and WT in the DCT cluster showed Ier3, an inhibitor of apoptosis, to be the most downregulated gene. These observations demonstrate that the salt-wasting effect of Vpr in Vpr Tg mice is mediated by loss of Slc12a3+ Pvalb+ DCT1 segments via apoptosis dysregulation.
Collapse
Affiliation(s)
- Khun Zaw Latt
- Kidney Disease Section, Kidney Diseases Branch, NIDDK, NIH, Bethesda MD
| | - Teruhiko Yoshida
- Kidney Disease Section, Kidney Diseases Branch, NIDDK, NIH, Bethesda MD
| | - Shashi Shrivastav
- Kidney Disease Section, Kidney Diseases Branch, NIDDK, NIH, Bethesda MD
| | - Amin Abedini
- Department of Medicine, Renal Electrolyte and Hypertension Division; Institute for Diabetes, Obesity, and Metabolism; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jeff M. Reece
- Advanced Light Microscopy & Image Analysis Core (ALMIAC), NIDDK, NIH, Bethesda, MD
| | - Zeguo Sun
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Hewang Lee
- Departments of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC
| | - Koji Okamoto
- Kidney Disease Section, Kidney Diseases Branch, NIDDK, NIH, Bethesda MD
- Division of Nephrology, Endocrinology and Vascular Medicine, Department of Medicine, Tohoku University Hospital, Aoba-ku, Sendai, Miyagi, Japan
| | - Pradeep Dagur
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Jurgen Heymann
- Kidney Disease Section, Kidney Diseases Branch, NIDDK, NIH, Bethesda MD
| | - Yongmei Zhao
- Advanced Biomedical and Computational Sciences, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., NCI, Frederick, MD
| | - Joon-Yong Chung
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, NCI, NIH, Bethesda, MD
| | - Stephen Hewitt
- Experimental Pathology Laboratory, Laboratory of Pathology, Center for Cancer Research, NCI, NIH, Bethesda, MD
| | - Pedro A. Jose
- Departments of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC
- Departments of Physiology and Pharmacology, The George Washington University School of Medicine & Health Sciences, Washington, DC
| | - Kyung Lee
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Cheryl A. Winkler
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute and Basic Research Program, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Mark A. Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, Division of Intramural Research, NHLBI, NIH, Bethesda, MD
| | - Tomoshige Kino
- Laboratory for Molecular and Genomic Endocrinology, Division of Translational Medicine, Sidra Medicine, Doha, Qatar
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD
| | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division; Institute for Diabetes, Obesity, and Metabolism; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jeffrey B. Kopp
- Kidney Disease Section, Kidney Diseases Branch, NIDDK, NIH, Bethesda MD
| |
Collapse
|
29
|
Wu Q, Meng W, Zhu B, Chen X, Fu J, Zhao C, Liu G, Luo X, Lv Y, Zhao W, Wang F, Hu S, Zhang S. VEGFC ameliorates salt-sensitive hypertension and hypertensive nephropathy by inhibiting NLRP3 inflammasome via activating VEGFR3-AMPK dependent autophagy pathway. Cell Mol Life Sci 2023; 80:327. [PMID: 37837447 PMCID: PMC11072217 DOI: 10.1007/s00018-023-04978-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/29/2023] [Accepted: 09/23/2023] [Indexed: 10/16/2023]
Abstract
Salt-sensitivity hypertension (SSHTN) is an independent predictor for cardiovascular mortality. VEGFC has been reported to be a protective role in SSHTN and hypertensive kidney injury. However, the underlying mechanisms remain largely unclear. The current study aimed to explore the protective effects and mechanisms of VEGFC against SSHTN and hypertensive nephropathy. Here, we reported that VEGFC attenuated high blood pressure as well as protected against renal inflammation and fibrosis in SSHTN mice. Moreover, VEGFC suppressed the activation of renal NLRP3 inflammasome in SSHTN mice. In vitro, we found VEGFC inhibited NLRP3 inflammasome activation, meanwhile, upregulated autophagy in high-salt-induced macrophages, while these effects were reversed by an autophagy inhibitor 3MA. Furthermore, in vivo, 3MA pretreatment weakened the protective effects of VEGFC on SSHTN and hypertensive nephropathy. Mechanistically, VEGF receptor 3 (VEGFR3) kinase domain activated AMPK by promoting the phosphorylation at Thr183 via binding to AMPK, thus enhancing autophagy activity in the context of high-salt-induced macrophages. These findings indicated that VEGFC inhibited NLRP3 inflammasome activation by promoting VEGFR3-AMPK-dependent autophagy pathway in high-salt-induced macrophages, which provided a mechanistic basis for the therapeutic target in SSHTN and hypertensive kidney injury.
Collapse
Affiliation(s)
- Qiuwen Wu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China
| | - Wei Meng
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China
| | - Bin Zhu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China
| | - Xi Chen
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Jiaxin Fu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Chunyu Zhao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Gang Liu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xing Luo
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Ying Lv
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Wenqi Zhao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Fan Wang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Sining Hu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China.
| | - Shuo Zhang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
30
|
Han S, Choi H, Park H, Kim JJ, Lee EJ, Ham YR, Na KR, Lee KW, Chang YK, Choi DE. Omega-3 Fatty Acids Attenuate Renal Fibrosis via AMPK-Mediated Autophagy Flux Activation. Biomedicines 2023; 11:2553. [PMID: 37760994 PMCID: PMC10525956 DOI: 10.3390/biomedicines11092553] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
The unilateral ureteral obstruction (UUO) injury model is well-known to mimic human chronic kidney disease, promoting the rapid onset and development of kidney injury. ω3-poly unsaturated fatty acids (PUFAs) have been observed to protect against tissue injury in many disease models. In this study, we assessed the efficacy of ω3-PUFAs in attenuating UUO injury and investigated their mechanism of action. The immortalized human proximal tubular cells human kidney-2 (HK2) were incubated for 72 h with docosahexaenoic acid (DHA) or eicosapentaenoic acid (EPA) in various concentrations, in the presence or absence of transforming growth factor (TGF)-β. DHA/EPA reduced the epithelial-mesenchymal transition in the TGF-β-treated HK2 cells by enhancing autophagy flux and adenosine monophosphate-activated protein kinase (AMPK) phosphorylation. C57BL/6 mice were divided into four groups and treated as follows: sham (no treatment, n = 5), sham + ω3-PUFAs (n = 5), UUO (n = 10), and UUO + ω3-PUFAs (n = 10). Their kidneys and blood were harvested on the seventh day following UUO injury. The kidneys of the ω3-PUFAs-treated UUO mice showed less oxidative stress, inflammation, and fibrosis compared to those of the untreated UUO mice. Greater autophagic flux, higher amounts of microtubule-associated protein 1A/1B-light chain 3 (LC3)-II, Beclin-1, and Atg7, lower amounts of p62, and higher levels of cathepsin D and ATP6E were observed in the kidneys of the omega-3-treated UUO mice compared to those of the control UUO mice. In conclusion, ω3-PUFAs enhanced autophagic activation, leading to a renoprotective response against chronic kidney injury.
Collapse
Affiliation(s)
- Suyeon Han
- Department of Nephrology, Chungnam National University Hospital, Daejeon 35015, Republic of Korea; (S.H.); (E.-J.L.); (Y.-R.H.); (K.-R.N.); (K.-W.L.)
| | - Hyunsu Choi
- Clinical Research Institute, Daejeon Saint Mary’s Hospital, Daejeon 34943, Republic of Korea;
| | - Hyerim Park
- Department of Medical Science, Medical School, Chungnam National University, Daejeon 35015, Republic of Korea; (H.P.); (J.-J.K.)
| | - Jwa-Jin Kim
- Department of Medical Science, Medical School, Chungnam National University, Daejeon 35015, Republic of Korea; (H.P.); (J.-J.K.)
| | - Eu-Jin Lee
- Department of Nephrology, Chungnam National University Hospital, Daejeon 35015, Republic of Korea; (S.H.); (E.-J.L.); (Y.-R.H.); (K.-R.N.); (K.-W.L.)
| | - Young-Rok Ham
- Department of Nephrology, Chungnam National University Hospital, Daejeon 35015, Republic of Korea; (S.H.); (E.-J.L.); (Y.-R.H.); (K.-R.N.); (K.-W.L.)
| | - Ki-Rayng Na
- Department of Nephrology, Chungnam National University Hospital, Daejeon 35015, Republic of Korea; (S.H.); (E.-J.L.); (Y.-R.H.); (K.-R.N.); (K.-W.L.)
| | - Kang-Wook Lee
- Department of Nephrology, Chungnam National University Hospital, Daejeon 35015, Republic of Korea; (S.H.); (E.-J.L.); (Y.-R.H.); (K.-R.N.); (K.-W.L.)
| | - Yoon-Kyung Chang
- Department of Nephrology, Daejeon Saint Mary’s Hospital, Catholic University of Korea, Daejeon 34943, Republic of Korea
| | - Dae-Eun Choi
- Department of Nephrology, Chungnam National University Hospital, Daejeon 35015, Republic of Korea; (S.H.); (E.-J.L.); (Y.-R.H.); (K.-R.N.); (K.-W.L.)
- Department of Medical Science, Medical School, Chungnam National University, Daejeon 35015, Republic of Korea; (H.P.); (J.-J.K.)
| |
Collapse
|
31
|
Salete-Granado D, Carbonell C, Puertas-Miranda D, Vega-Rodríguez VJ, García-Macia M, Herrero AB, Marcos M. Autophagy, Oxidative Stress, and Alcoholic Liver Disease: A Systematic Review and Potential Clinical Applications. Antioxidants (Basel) 2023; 12:1425. [PMID: 37507963 PMCID: PMC10376811 DOI: 10.3390/antiox12071425] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Ethanol consumption triggers oxidative stress by generating reactive oxygen species (ROS) through its metabolites. This process leads to steatosis and liver inflammation, which are critical for the development of alcoholic liver disease (ALD). Autophagy is a regulated dynamic process that sequesters damaged and excess cytoplasmic organelles for lysosomal degradation and may counteract the harmful effects of ROS-induced oxidative stress. These effects include hepatotoxicity, mitochondrial damage, steatosis, endoplasmic reticulum stress, inflammation, and iron overload. In liver diseases, particularly ALD, macroautophagy has been implicated as a protective mechanism in hepatocytes, although it does not appear to play the same role in stellate cells. Beyond the liver, autophagy may also mitigate the harmful effects of alcohol on other organs, thereby providing an additional layer of protection against ALD. This protective potential is further supported by studies showing that drugs that interact with autophagy, such as rapamycin, can prevent ALD development in animal models. This systematic review presents a comprehensive analysis of the literature, focusing on the role of autophagy in oxidative stress regulation, its involvement in organ-organ crosstalk relevant to ALD, and the potential of autophagy-targeting therapeutic strategies.
Collapse
Affiliation(s)
- Daniel Salete-Granado
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
| | - Cristina Carbonell
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - David Puertas-Miranda
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Víctor-José Vega-Rodríguez
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Marina García-Macia
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Instituto de Biología Funcional y Genómica (IBFG), Universidad de Salamanca, 37007 Salamanca, Spain
| | - Ana Belén Herrero
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Miguel Marcos
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
32
|
de la Fuente AG, Pelucchi S, Mertens J, Di Luca M, Mauceri D, Marcello E. Novel therapeutic approaches to target neurodegeneration. Br J Pharmacol 2023; 180:1651-1673. [PMID: 36965025 PMCID: PMC10952850 DOI: 10.1111/bph.16078] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/26/2023] [Accepted: 03/17/2023] [Indexed: 03/27/2023] Open
Abstract
Ageing is the main risk factor common to most primary neurodegenerative disorders. Indeed, age-related brain alterations have been long considered to predispose to neurodegeneration. Although protein misfolding and the accumulation of toxic protein aggregates have been considered as causative events in neurodegeneration, several other biological pathways affected by brain ageing also contribute to pathogenesis. Here, we discuss the evidence showing the involvement of the mechanisms controlling neuronal structure, gene expression, autophagy, cell metabolism and neuroinflammation in the onset and progression of neurodegenerative disorders. Furthermore, we review the therapeutic strategies currently under development or as future approaches designed to normalize these pathways, which may then increase brain resilience to cope with toxic protein species. In addition to therapies targeting the insoluble protein aggregates specifically associated with each neurodegenerative disorder, these novel pharmacological approaches may be part of combined therapies designed to rescue brain function.
Collapse
Affiliation(s)
- Alerie G. de la Fuente
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL)AlicanteSpain
- Instituto de Neurociencias CSIC‐UMHAlicanteSpain
- Wellcome‐Wolfson Institute for Experimental MedicineQueen's University BelfastBelfastUK
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
- Institute of Molecular BiologyLeopold‐Franzens‐Universität InnsbruckInnsbruckAustria
| | - Jerome Mertens
- Institute of Molecular BiologyLeopold‐Franzens‐Universität InnsbruckInnsbruckAustria
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| | - Daniela Mauceri
- Institute of Anatomy and Cell BiologyDepartment of Molecular and Cellular Neuroscience, University of MarburgMarburgGermany
- Department of NeurobiologyInterdisciplinary Centre for Neurosciences (IZN), Heidelberg UniversityHeidelbergGermany
| | - Elena Marcello
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| |
Collapse
|
33
|
Yu B, Zhou Y, He J. TRIM13 inhibits cell proliferation and induces autophagy in lung adenocarcinoma by regulating KEAP1/NRF2 pathway. Cell Cycle 2023; 22:1496-1513. [PMID: 37245083 PMCID: PMC10281484 DOI: 10.1080/15384101.2023.2216504] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 05/29/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is the most common type of lung cancer. Tripartite motif 13 (TRIM13) is a member of TRIM protein family and is downregulated in multiple cancers, especially non-small cell lung cancers (NSCLC). In this study, we investigated anti-tumor mechanism of TRIM13 in non-small cell lung cancer tissues and cell lines. First, the mRNA and protein levels of TRIM13 in LUAD tissue and cells were measured. TRIM13 was overexpressed on LUAD cells to investigate the effects on cell proliferation, apoptosis, oxidative stress, p62 ubiquitination, and autophagy activation. Finally, mechanistic role of TRIM13 in regulating the Keap1/Nrf2 pathway was investigated. Results indicated that low level of TRIM13 mRNA and protein expression was found in LUAD tissue and cells. Overexpression of TRIM13 in LUAD cancer cells suppressed their proliferation, increased apoptosis, and oxidative stress, ubiquitinated p62, and activated autophagy via the RING finger domain of TRIM13. Furthermore, TRIM13 showed interaction with p62 and mediated its ubiquitination and degradation in LUAD cells. Mechanistically, TRIM13 exerted the tumor suppressor functions in LUAD cells by negatively regulating Nrf2 signaling and downstream antioxidants, which was further confirmed by in vivo data from xenografts. In conclusion, TRIM13 behaves like a tumor suppressor and triggers autophagy in LUAD cells by mediating p62 ubiquitination via KEAP1/Nrf2 pathway. Our findings provide a novel insight into targeted therapy plans for LUAD.
Collapse
Affiliation(s)
- Bo Yu
- Department of thoracic surgery, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yu Zhou
- Department of Scientific Research, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jinxi He
- Department of thoracic surgery, The General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
34
|
Rho SB, Byun HJ, Kim BR, Lee CH. Liver Kinase B1 Mediates Its Anti-Tumor Function by Binding to the N-Terminus of Malic Enzyme 3. Biomol Ther (Seoul) 2023; 31:330-339. [PMID: 37095735 PMCID: PMC10129855 DOI: 10.4062/biomolther.2023.041] [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: 03/01/2023] [Revised: 03/18/2023] [Accepted: 03/24/2023] [Indexed: 04/26/2023] Open
Abstract
Liver kinase B1 (LKB1) is a crucial tumor suppressor involved in various cellular processes, including embryonic development, tumor initiation and progression, cell adhesion, apoptosis, and metabolism. However, the precise mechanisms underlying its functions remain elusive. In this study, we demonstrate that LKB1 interacts directly with malic enzyme 3 (ME3) through the N-terminus of the enzyme and identified the binding regions necessary for this interaction. The binding activity was confirmed to promote the expression of ME3 in an LKB1-dependent manner and was also shown to induce apoptosis activity. Furthermore, LKB1 and ME3 overexpression upregulated the expression of tumour suppressor proteins (p53 and p21) and downregulated the expression of antiapoptotic proteins (nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and B-cell lymphoma 2 (Bcl-2)). Additionally, LKB1 and ME3 enhanced the transcription of p21 and p53 and inhibited the transcription of NF-κB. Moreover, LKB1 and ME3 suppressed the phosphorylation of various components of the phosphatidylinositol-4,5-bisphosphate 3-kinase/protein kinase B signaling pathway. Overall, these results suggest that LKB1 promotes pro-apoptotic activities by inducing ME3 expression.
Collapse
Affiliation(s)
- Seung Bae Rho
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408
| | - Hyun Jung Byun
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea
| | - Boh-Ram Kim
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea
| | - Chang Hoon Lee
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea
| |
Collapse
|
35
|
Lin M, Yu H, Xie Q, Xu Z, Shang P. Role of microglia autophagy and mitophagy in age-related neurodegenerative diseases. Front Aging Neurosci 2023; 14:1100133. [PMID: 37180741 PMCID: PMC10169626 DOI: 10.3389/fnagi.2022.1100133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 12/28/2022] [Indexed: 05/16/2023] Open
Abstract
Microglia, characterized by responding to damage, regulating the secretion of soluble inflammatory mediators, and engulfing specific segments in the central nervous system (CNS), function as key immune cells in the CNS. Emerging evidence suggests that microglia coordinate the inflammatory responses in CNS system and play a pivotal role in the pathogenesis of age-related neurodegenerative diseases (NDDs). Remarkably, microglia autophagy participates in the regulation of subcellular substances, which includes the degradation of misfolded proteins and other harmful constituents produced by neurons. Therefore, microglia autophagy regulates neuronal homeostasis maintenance and process of neuroinflammation. In this review, we aimed at highlighting the pivotal role of microglia autophagy in the pathogenesis of age-related NDDs. Besides the mechanistic process and the co-interaction between microglia autophagy and different kinds of NDDs, we also emphasized potential therapeutic agents and approaches that could be utilized at the onset and progression of these diseases through modulating microglia autophagy, including promising nanomedicines. Our review provides a valuable reference for subsequent studies focusing on treatments of neurodegenerative disorders. The exploration of microglia autophagy and the development of nanomedicines greatly enhances current understanding of NDDs.
Collapse
Affiliation(s)
- Mingkai Lin
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongwen Yu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiuyan Xie
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiyun Xu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pei Shang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
36
|
Chen L, Zhang W, Chen D, Yang Q, Sun S, Dai Z, Li Z, Liang X, Chen C, Jiao Y, Zhi L, Zhao L, Zhang J, Liu X, Zhao J, Li M, Wang Y, Qi Y. RBM4 dictates ESCC cell fate switch from cellular senescence to glutamine-addiction survival through inhibiting LKB1-AMPK-axis. Signal Transduct Target Ther 2023; 8:159. [PMID: 37080995 PMCID: PMC10119322 DOI: 10.1038/s41392-023-01367-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 01/09/2023] [Accepted: 02/14/2023] [Indexed: 04/22/2023] Open
Abstract
Cellular senescence provides a protective barrier against tumorigenesis in precancerous or normal tissues upon distinct stressors. However, the detailed mechanisms by which tumor cells evade premature senescence to malignant progression remain largely elusive. Here we reported that RBM4 adversely impacted cellular senescence to favor glutamine-dependent survival of esophageal squamous cell carcinoma (ESCC) cells by dictating the activity of LKB1, a critical governor of cancer metabolism. The level of RBM4 was specifically elevated in ESCC compared to normal tissues, and RBM4 overexpression promoted the malignant phenotype. RBM4 contributed to overcome H-RAS- or doxorubicin-induced senescence, while its depletion caused P27-dependent senescence and proliferation arrest by activating LKB1-AMPK-mTOR cascade. Mechanistically, RBM4 competitively bound LKB1 to disrupt the LKB1/STRAD/MO25 heterotrimeric complex, subsequently recruiting the E3 ligase TRIM26 to LKB1, promoting LKB1 ubiquitination and degradation in nucleus. Therefore, such molecular process leads to bypassing senescence and sustaining cell proliferation through the activation of glutamine metabolism. Clinically, the ESCC patients with high RBM4 and low LKB1 have significantly worse overall survival than those with low RBM4 and high LKB1. The RBM4 high/LKB1 low expression confers increased sensitivity of ESCC cells to glutaminase inhibitor CB-839, providing a novel insight into mechanisms underlying the glutamine-dependency to improve the efficacy of glutamine inhibitors in ESCC therapeutics.
Collapse
Affiliation(s)
- Lei Chen
- Institute of Cancer Stem Cells and the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116044, China
| | - Wenjing Zhang
- Institute of Cancer Stem Cells and the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116044, China
| | - Dan Chen
- Department of Pathology, the First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Quan Yang
- Institute of Cancer Stem Cells and the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116044, China
| | - Siwen Sun
- Department of Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Zhenwei Dai
- Institute of Cancer Stem Cells and the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116044, China
| | - Zhengzheng Li
- Institute of Cancer Stem Cells and the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116044, China
| | - Xuemei Liang
- Department of Thoracic Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Chaoqun Chen
- Institute of Cancer Stem Cells and the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116044, China
| | - Yuexia Jiao
- Institute of Cancer Stem Cells and the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116044, China
| | - Lili Zhi
- Institute of Cancer Stem Cells and the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116044, China
| | - Lianmei Zhao
- Research Center, the Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Jinrui Zhang
- Institute of Cancer Stem Cells and the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116044, China
| | - Xuefeng Liu
- Institute of Cancer Stem Cells and the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116044, China
| | - Jinyao Zhao
- Institute of Cancer Stem Cells and the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116044, China
| | - Man Li
- Department of Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China.
| | - Yang Wang
- Institute of Cancer Stem Cells and the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116044, China.
| | - Yangfan Qi
- Institute of Cancer Stem Cells and the Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
37
|
Kouroumalis E, Tsomidis I, Voumvouraki A. Pathogenesis of Hepatocellular Carcinoma: The Interplay of Apoptosis and Autophagy. Biomedicines 2023; 11:1166. [PMID: 37189787 PMCID: PMC10135776 DOI: 10.3390/biomedicines11041166] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/09/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
The pathogenesis of hepatocellular carcinoma (HCC) is a multifactorial process that has not yet been fully investigated. Autophagy and apoptosis are two important cellular pathways that are critical for cell survival or death. The balance between apoptosis and autophagy regulates liver cell turnover and maintains intracellular homeostasis. However, the balance is often dysregulated in many cancers, including HCC. Autophagy and apoptosis pathways may be either independent or parallel or one may influence the other. Autophagy may either inhibit or promote apoptosis, thus regulating the fate of the liver cancer cells. In this review, a concise overview of the pathogenesis of HCC is presented, with emphasis on new developments, including the role of endoplasmic reticulum stress, the implication of microRNAs and the role of gut microbiota. The characteristics of HCC associated with a specific liver disease are also described and a brief description of autophagy and apoptosis is provided. The role of autophagy and apoptosis in the initiation, progress and metastatic potential is reviewed and the experimental evidence indicating an interplay between the two is extensively analyzed. The role of ferroptosis, a recently described specific pathway of regulated cell death, is presented. Finally, the potential therapeutic implications of autophagy and apoptosis in drug resistance are examined.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Department of Gastroenterology, PAGNI University Hospital, University of Crete School of Medicine, 71500 Heraklion, Crete, Greece
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Crete, Greece
| | - Ioannis Tsomidis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Crete, Greece
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Central Macedonia, Greece
| | - Argyro Voumvouraki
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Central Macedonia, Greece
| |
Collapse
|
38
|
Steinberg GR, Hardie DG. New insights into activation and function of the AMPK. Nat Rev Mol Cell Biol 2023; 24:255-272. [PMID: 36316383 DOI: 10.1038/s41580-022-00547-x] [Citation(s) in RCA: 396] [Impact Index Per Article: 198.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2022] [Indexed: 11/06/2022]
Abstract
The classical role of AMP-activated protein kinase (AMPK) is as a cellular energy sensor activated by falling energy status, signalled by increases in AMP to ATP and ADP to ATP ratios. Once activated, AMPK acts to restore energy homeostasis by promoting ATP-producing catabolic pathways while inhibiting energy-consuming processes. In this Review, we provide an update on this canonical (AMP/ADP-dependent) activation mechanism, but focus mainly on recently described non-canonical pathways, including those by which AMPK senses the availability of glucose, glycogen or fatty acids and by which it senses damage to lysosomes and nuclear DNA. We also discuss new findings on the regulation of carbohydrate and lipid metabolism, mitochondrial and lysosomal homeostasis, and DNA repair. Finally, we discuss the role of AMPK in cancer, obesity, diabetes, nonalcoholic steatohepatitis (NASH) and other disorders where therapeutic targeting may exert beneficial effects.
Collapse
Affiliation(s)
- Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - D Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, UK.
| |
Collapse
|
39
|
AMPK Phosphorylation Impacts Apoptosis in Differentiating Myoblasts Isolated from Atrophied Rat Soleus Muscle. Cells 2023; 12:cells12060920. [PMID: 36980261 PMCID: PMC10047078 DOI: 10.3390/cells12060920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/25/2023] [Accepted: 03/10/2023] [Indexed: 03/19/2023] Open
Abstract
Regrowth of atrophied myofibers depends on muscle satellite cells (SCs) that exist outside the plasma membrane. Muscle atrophy appears to result in reduced number of SCs due to apoptosis. Given reduced AMP-activated protein kinase (AMPK) activity during differentiation of primary myoblasts derived from atrophic muscle, we hypothesized that there may be a potential link between AMPK and susceptibility of differentiating myoblasts to apoptosis. The aim of this study was to estimate the effect of AMPK activation (via AICAR treatment) on apoptosis in differentiating myoblasts derived from atrophied rat soleus muscle. Thirty rats were randomly assigned to the following two groups: control (C, n = 10) and 7-day hindlimb suspension (HS, n = 20). Myoblasts derived from the soleus muscles of HS rats were divided into two parts: AICAR-treated cells and non-treated cells. Apoptotic processes were evaluated by using TUNEL assay, RT-PCR and WB. In differentiating myoblasts derived from the atrophied soleus, there was a significant decrease (p < 0.05) in AMPK and ACC phosphorylation in parallel with increased number of apoptotic nuclei and a significant upregulation of pro-apoptotic markers (caspase-3, -9, BAX, p53) compared to the cells derived from control muscles. AICAR treatment of atrophic muscle-derived myoblasts during differentiation prevented reductions in AMPK and ACC phosphorylation as well as maintained the number of apoptotic nuclei and the expression of pro-apoptotic markers at the control levels. Thus, the maintenance of AMPK activity can suppress enhanced apoptosis in differentiating myoblasts derived from atrophied rat soleus muscle.
Collapse
|
40
|
Huang Y, Zhou Q, Li W, Chen Y. The expression of p27 in the adult vestibular sensory organs and its possible roles. Neurosci Lett 2023; 800:137128. [PMID: 36792024 DOI: 10.1016/j.neulet.2023.137128] [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: 11/27/2022] [Revised: 01/26/2023] [Accepted: 02/11/2023] [Indexed: 02/15/2023]
Abstract
Vestibular hair cells (HCs) located in the inner ear are the receptors of vestibular sensory, which facilitates the human sense of balance. The detailed differentiation pattern and maturation process of the vestibular HCs are unclear now. p27, a cyclin/CDK inhibitor, plays a critical role in regulating the exit of cell cycle. We found that p27 was continuously expressed in the terminally differentiated and mature vestibular HCs using p27-P2A-iCreER/+; Rosa26-LSL-tdTomato/+ mice, suggesting p27 might have novel roles independent of its CDK inhibitory action. p27 is also reported to be associated with cell differentiation, cell migration and cell survival. We further explored the difference of p27 expression between two subtypes of vestibular HCs, and found that the proportion of p27-tdTomato positive type I vestibular HCs increased gradually along the subtype determination and maturation of vestibular HCs, suggesting that p27 might play a role in the HC subtype differentiation, maturation and function acquirement.
Collapse
Affiliation(s)
- Yikang Huang
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| | - Qin Zhou
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China
| | - Wenyan Li
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China; Institutesof Biomedical Sciences, Fudan University, Shanghai 200032, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China; The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China.
| | - Yan Chen
- ENT Institute and Otorhinolaryngology Department of Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Hearing Medicine (Fudan University), Shanghai 200031, China.
| |
Collapse
|
41
|
Chakraborthy M, Rao A. A Feedback Loop between TGF-β1 and ATG5 Mediated by miR-122-5p Regulates Fibrosis and EMT in Human Trabecular Meshwork Cells. Curr Issues Mol Biol 2023; 45:2381-2392. [PMID: 36975524 PMCID: PMC10047315 DOI: 10.3390/cimb45030154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/11/2023] [Accepted: 02/20/2023] [Indexed: 03/16/2023] Open
Abstract
Autophagy is a cell’s evolutionary conserved process for degrading and recycling cellular proteins and removing damaged organelles. There has been an increasing interest in identifying the basic cellular mechanism of autophagy and its implications in health and illness during the last decade. Many proteinopathies such as Alzheimer’s and Huntington’s disease are reported to be associated with impaired autophagy. The functional significance of autophagy in exfoliation syndrome/exfoliation glaucoma (XFS/XFG), remains unknown though it is presumed to be impaired autophagy to be responsible for the aggregopathy characteristic of this disease. In the current study we have shown that autophagy or ATG5 is enhanced in response to TGF-β1 in human trabecular meshwork (HTM) cells and TGF-β1 induced autophagy is necessary for increased expression of profibrotic proteins and epithelial to mesenchymal (EMT) through Smad3 that lead to aggregopathy. Inhibition of ATG5 by siRNA mediated knockdown reduced profibrotic and EMT markers and increased protein aggregates in the presence of TGF-β1 stimulation. The miR-122-5p, which was increased upon TGF exposure, was also reduced upon ATG5 inhibition. We thus conclude that TGF-β1 induces autophagy in primary HTM cells and a positive feedback loop exists between TGF-β1 and ATG5 that regulated TGF downstream effects mainly mediated by Smad3 signaling with miR-122-5p also playing a role.
Collapse
Affiliation(s)
- Munmun Chakraborthy
- Hyderabad Eye Research Foundation (HERF), L.V. Prasad Eye Institute, Bhubaneswar 751024, Odisha, India
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Aparna Rao
- Hyderabad Eye Research Foundation (HERF), L.V. Prasad Eye Institute, Bhubaneswar 751024, Odisha, India
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
- Correspondence:
| |
Collapse
|
42
|
Silvis MR, Silva D, Rohweder R, Schuman S, Gudipaty S, Truong A, Yap J, Affolter K, McMahon M, Kinsey C. MYC-mediated resistance to trametinib and HCQ in PDAC is overcome by CDK4/6 and lysosomal inhibition. J Exp Med 2023; 220:e20221524. [PMID: 36719686 PMCID: PMC9930170 DOI: 10.1084/jem.20221524] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 12/02/2022] [Accepted: 12/20/2022] [Indexed: 02/01/2023] Open
Abstract
Pharmacological inhibition of KRAS>RAF>MEK1/2>ERK1/2 signaling has provided no clinical benefit to patients with pancreatic ductal adenocarcinoma (PDAC). Interestingly, combined inhibition of MEK1/2 (with trametinib [T]) plus autophagy (with chloroquine [CQ] or hydroxychloroquine [HCQ]) demonstrated striking anti-tumor effects in preclinical models and in a patient (Patient 1). However, not all patients respond to the T/HCQ regimen, and Patient 1 eventually developed resistant disease. Here we report that primary or acquired resistance is associated with focal DNA copy number gains encompassing c-MYC. Furthermore, ectopic expression of c-MYC in PDAC cell lines rendered them T/HCQ resistant. Interestingly, a CDK4/6 inhibitor, palbociclib (P), also induced autophagy and overrode c-MYC-mediated T/HCQ resistance, such that P/HCQ promoted regression of T/HCQ-resistant PDAC tumors with elevated c-MYC expression. Finally, P/HCQ treatment of Patient 1 resulted in a biochemical disease response. These data suggest that elevated c-MYC expression is both a marker and a mediator of T/HCQ resistance, which may be overcome by the use of P/HCQ.
Collapse
Affiliation(s)
- Mark R. Silvis
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Dilru Silva
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Riley Rohweder
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Sophia Schuman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | | | | | - Jeffrey Yap
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Radiology, University of Utah, Salt Lake City, UT, USA
| | - Kajsa Affolter
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Martin McMahon
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
- Department of Dermatology, University of Utah, Salt Lake City, UT, USA
| | - Conan Kinsey
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Oncology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
43
|
Wang L, Han H, Feng Y, Ma J, Han Z, Li R, Zhu W, Li S, Tian J, Zhang L. Capilliposide B inhibits the migration of prostate cancer by inducing autophagy through the ROS/AMPK/mTOR pathway. Phytother Res 2023. [PMID: 36867511 DOI: 10.1002/ptr.7785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Capilliposide B (CPS-B), a novel oleanane triterpenoid saponin derived from Lysimachia capillipes Hemsl, is a potent anticancer agent. However, its anticancer mechanism remains elusive. In the present study, we demonstrated the potent anti-tumor activity and molecular mechanism of CPS-B both in vitro and in vivo. Proteomic analysis using isobaric tags for relative and absolute quantitation techniques suggested that CPS-B modulated autophagy in prostate cancer (PC). Moreover, Western blotting showed that both autophagy and epithelial-mesenchymal transition occurred place after CPS-B treatment in vivo, which was also proven in PC-3 cancer cells. We deduced that CPS-B inhibited migration by inducing autophagy. We examined the accumulation of reactive oxygen species (ROS) in cells, and in downstream pathways, LKB1 and AMPK were activated while mTOR was inhibited. Transwell experiment results showed that CPS-B inhibited the metastasis of PC-3 cells and that this effect was significantly attenuated after pretreatment with chloroquine, indicating that CPS-B inhibited metastasis via autophagy induction. Altogether, these data suggest that CPS-B is a potential therapeutic agent for cancer treatment that acts by inhibiting migration through the ROS/AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Luping Wang
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, People's Republic of China.,Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Haote Han
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Yue Feng
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Jiahui Ma
- Marine Science College, Zhejiang Ocean University, Zhoushan, Zhejiang, People's Republic of China
| | - Zhuo Han
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Ruyi Li
- College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, People's Republic of China
| | - Wei Zhu
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Shouxin Li
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Jingkui Tian
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, People's Republic of China
| | - Lin Zhang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, People's Republic of China
| |
Collapse
|
44
|
Hu L, Liu M, Tang B, Li Q, Pan BS, Xu C, Lin HK. Posttranslational regulation of liver kinase B1 (LKB1) in human cancer. J Biol Chem 2023; 299:104570. [PMID: 36870679 PMCID: PMC10068580 DOI: 10.1016/j.jbc.2023.104570] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/06/2023] Open
Abstract
Liver kinase B1 (LKB1) is a serine-threonine kinase that participates in multiple cellular and biological processes, including energy metabolism, cell polarity, cell proliferation, cell migration, and many others. LKB1 is initially identified as a germline-mutated causative gene in Peutz-Jeghers syndrome (PJS) and is commonly regarded as a tumor suppressor due to frequent inactivation in a variety of cancers. LKB1 directly binds and activates its downstream kinases including the AMP-activated protein kinase (AMPK) and AMPK-related kinases by phosphorylation, which has been intensively investigated for the past decades. An increasing number of studies has uncovered the posttranslational modifications (PTMs) of LKB1 and consequent changes in its localization, activity, and interaction with substrates. The alteration in LKB1 function as a consequence of genetic mutations and aberrant upstream signaling regulation leads to tumor development and progression. Here, we review current knowledge about the mechanism of LKB1 in cancer and the contributions of PTMs, such as phosphorylation, ubiquitination, SUMOylation, acetylation, prenylation, and others, to the regulation of LKB1 function, offering new insights into the therapeutic strategies in cancer.
Collapse
Affiliation(s)
- Lanlin Hu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Mingxin Liu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Bo Tang
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiang Li
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Bo-Syong Pan
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Chuan Xu
- Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Department of Laboratory Medicine and Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.
| |
Collapse
|
45
|
Njeka Wojnarova L, Kutinova Canova N, Arora M, Farghali H. Differentiated modulation of signaling molecules AMPK and SIRT1 in experimentally drug-induced hepatocyte injury. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2023; 167:50-60. [PMID: 35416184 DOI: 10.5507/bp.2022.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/04/2022] [Indexed: 11/23/2022] Open
Abstract
AIM Currently available medicines have little to offer in terms of supporting the regeneration of injured hepatic cells. Previous experimental studies have shown that resveratrol and metformin, less specific activators of AMP-activated protein kinase (AMPK) and sirtuin 1 (SIRT1), can effectively attenuate acute liver injury. The aim of this experimental study was to elucidate whether modulation of AMPK and SIRT1 activity can modify drug/paracetamol (APAP)-induced hepatocyte damage in vitro. METHODS Primary rat hepatocytes were pretreated with mutual combinations of specific synthetic activators and inhibitors of SIRT1 and AMPK and followed by a toxic dose of APAP. At the end of cultivation, medium samples were collected for biochemical analysis of alanine-aminotransferase and nitrite levels. Hepatocyte viability, thiobarbituric reactive substances, SIRT1 and AMPK activity and protein expression were also assessed. RESULTS The harmful effect of APAP was associated with decreased AMPK and SIRT1 activity and protein expression alongside enhanced oxidative stress in hepatocytes. The addition of AMPK activator (AICAR) or SIRT1 activator (CAY10591) significantly attenuated the deleterious effects of AMPK inhibitor (Compound C) on the hepatotoxicity of APAP. Furthermore, CAY10591 but not AICAR markedly decreased the deleterious effect of APAP in combination with SIRT1 inhibitor (EX-527). CONCLUSION Our findings demonstrate that decreased AMPK activity is associated with the hepatotoxic effect of APAP which can be significantly attenuated by the administration of a SIRT1 activator. These findings suggest that differentiated modulation of AMPK and SIRT1 activity could therefore provide an interesting and novel therapeutic opportunity in the future to combat hepatocyte injury.
Collapse
Affiliation(s)
- Lea Njeka Wojnarova
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Nikolina Kutinova Canova
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Mahak Arora
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Hassan Farghali
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| |
Collapse
|
46
|
Kasprzak A. Autophagy and the Insulin-like Growth Factor (IGF) System in Colonic Cells: Implications for Colorectal Neoplasia. Int J Mol Sci 2023; 24:ijms24043665. [PMID: 36835075 PMCID: PMC9959216 DOI: 10.3390/ijms24043665] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/02/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common human malignancies worldwide. Along with apoptosis and inflammation, autophagy is one of three important mechanisms in CRC. The presence of autophagy/mitophagy in most normal mature intestinal epithelial cells has been confirmed, where it has mainly protective functions against reactive oxygen species (ROS)-induced DNA and protein damage. Autophagy regulates cell proliferation, metabolism, differentiation, secretion of mucins and/or anti-microbial peptides. Abnormal autophagy in intestinal epithelial cells leads to dysbiosis, a decline in local immunity and a decrease in cell secretory function. The insulin-like growth factor (IGF) signaling pathway plays an important role in colorectal carcinogenesis. This is evidenced by the biological activities of IGFs (IGF-1 and IGF-2), IGF-1 receptor type 1 (IGF-1R) and IGF-binding proteins (IGF BPs), which have been reported to regulate cell survival, proliferation, differentiation and apoptosis. Defects in autophagy are found in patients with metabolic syndrome (MetS), inflammatory bowel diseases (IBD) and CRC. In neoplastic cells, the IGF system modulates the autophagy process bidirectionally. In the current era of improving CRC therapies, it seems important to investigate the exact mechanisms not only of apoptosis, but also of autophagy in different populations of tumor microenvironment (TME) cells. The role of the IGF system in autophagy in normal as well as transformed colorectal cells still seems poorly understood. Hence, the aim of the review was to summarize the latest knowledge on the role of the IGF system in the molecular mechanisms of autophagy in the normal colon mucosa and in CRC, taking into account the cellular heterogeneity of the colonic and rectal epithelium.
Collapse
Affiliation(s)
- Aldona Kasprzak
- Department of Histology and Embryology, University of Medical Sciences, Swiecicki Street 6, 60-781 Poznan, Poland
| |
Collapse
|
47
|
Son S, Im JH, Ko J, Han K. SNF1-related protein kinase 1 represses Arabidopsis growth through post-translational modification of E2Fa in response to energy stress. THE NEW PHYTOLOGIST 2023; 237:823-839. [PMID: 36478538 PMCID: PMC10107498 DOI: 10.1111/nph.18597] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 10/08/2022] [Indexed: 06/01/2023]
Abstract
Cellular sugar starvation and/or energy deprivation serves as an important signaling cue for the live cells to trigger the necessary stress adaptation response. When exposed to cellular energy stress (ES) conditions, the plants reconfigure metabolic pathways and rebalance energy status while restricting vegetative organ growth. Despite the vital importance of this ES-induced growth restriction, the regulatory mechanism underlying the response remains largely elusive in plants. Using plant cell- and whole plant-based functional analyses coupled with extended genetic validation, we show that cellular ES-activated SNF1-related protein kinase 1 (SnRK1.1) directly interacts with and phosphorylates E2Fa transcription factor, a critical cell cycle regulator. Phosphorylation of E2Fa by SnRK1.1 leads to its proteasome-mediated protein degradation, resulting in S-phase repression and organ growth restriction. Our findings show that ES-dependently activated SnRK1.1 adjusts cell proliferation and vegetative growth for plants to cope with constantly fluctuating environments.
Collapse
Affiliation(s)
- Seungmin Son
- Department of Life SciencesKorea University145 Anamro, Sungbuk‐guSeoul02841Korea
- National Institute of Agricultural Sciences, Rural Development AdministrationJeonju54874Korea
| | - Jong Hee Im
- Department of Life SciencesKorea University145 Anamro, Sungbuk‐guSeoul02841Korea
- Department of HorticultureMichigan State UniversityEast LansingMI48824USA
| | - Jae‐Heung Ko
- Department of Plant & Environmental New Resources, College of Life Science and Graduate School of BiotechnologyKyung Hee UniversityYongin‐siGyeonggi‐do17104Korea
| | - Kyung‐Hwan Han
- Department of HorticultureMichigan State UniversityEast LansingMI48824USA
- Department of ForestryMichigan State UniversityEast LansingMI48824USA
| |
Collapse
|
48
|
Al-Huseini I, Sirasanagandla SR, Babu KS, Sofin RGS, Das S. Kinase Inhibitors Involved in the Regulation of Autophagy: Molecular Concepts and Clinical Implications. Curr Med Chem 2023; 30:1502-1528. [PMID: 35078392 DOI: 10.2174/0929867329666220117114306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/08/2021] [Accepted: 11/22/2021] [Indexed: 11/22/2022]
Abstract
All cells and intracellular components are remodeled and recycled in order to replace the old and damaged cells. Autophagy is a process by which damaged, and unwanted cells are degraded in the lysosomes. There are three different types of autophagy: macroautophagy, microautophagy, and chaperone-mediated autophagy. Autophagy has an effect on adaptive and innate immunity, suppression of any tumour, and the elimination of various microbial pathogens. The process of autophagy has both positive and negative effects, and this pertains to any specific disease or its stage of progression. Autophagy involves various processes which are controlled by various signaling pathways, such as Jun N-terminal kinase, GSK3, ERK1, Leucine-rich repeat kinase 2, and PTEN-induced putative kinase 1 and parkin RBR E3. Protein kinases are also important for the regulation of autophagy as they regulate the process of autophagy either by activation or inhibition. The present review discusses the kinase catalyzed phosphorylated reactions, the kinase inhibitors, types of protein kinase inhibitors and their binding properties to protein kinase domains, the structures of active and inactive kinases, and the hydrophobic spine structures in active and inactive protein kinase domains. The intervention of autophagy by targeting specific kinases may form the mainstay of treatment of many diseases and lead the road to future drug discovery.
Collapse
Affiliation(s)
- Isehaq Al-Huseini
- Department of Physiology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Al-Khodh 123, Oman
| | - Srinivasa Rao Sirasanagandla
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Al-Khodh 123, Oman
| | - Kondaveeti Suresh Babu
- Department of Biochemistry, Symbiosis Medical College for Women, Symbiosis International (Deemed) University, Pune, Maharashtra, India
| | | | - Srijit Das
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Al-Khodh 123, Oman
| |
Collapse
|
49
|
Qiu Y, Ke S, Chen J, Qin Z, Zhang W, Yuan Y, Meng D, Zhao G, Wu K, Li B, Li D. FOXP3+ regulatory T cells and the immune escape in solid tumours. Front Immunol 2022; 13:982986. [PMID: 36569832 PMCID: PMC9774953 DOI: 10.3389/fimmu.2022.982986] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/01/2022] [Indexed: 01/15/2023] Open
Abstract
FOXP3+ regulatory T (Treg) cells play critical roles in establishing the immunosuppressive tumour microenvironment, which is achieved and dynamically maintained with the contribution of various stromal and immune cell subsets. However, the dynamics of non-lymphoid FOXP3+ Treg cells and the mutual regulation of Treg cells and other cell types in solid tumour microenvironment remains largely unclear. In this review, we summarize the latest findings on the dynamic connections and reciprocal regulations of non-lymphoid Treg cell subsets in accordance with well-established and new emerging hallmarks of cancer, especially on the immune escape of tumour cells in solid tumours. Our comprehension of the interplay between FOXP3+ Treg cells and key hallmarks of cancer may provide new insights into the development of next-generation engineered T cell-based immune treatments for solid tumours.
Collapse
Affiliation(s)
- Yiran Qiu
- Department of Breast Surgery, Obstetrics and Gynecology Hospital, Fudan University School of Medicine, Shanghai, China
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shouyu Ke
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieqiong Chen
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhizhen Qin
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenle Zhang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaqin Yuan
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dehua Meng
- Department of Orthopedics, Zhongshan Hospital, Fudan University School of Medicine, Shanghai, China
| | - Gang Zhao
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kejin Wu
- Department of Breast Surgery, Obstetrics and Gynecology Hospital, Fudan University School of Medicine, Shanghai, China
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Arthritis Research, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Integrated TCM & Western Medicine at Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dan Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
50
|
Hsu CC, Peng D, Cai Z, Lin HK. AMPK signaling and its targeting in cancer progression and treatment. Semin Cancer Biol 2022; 85:52-68. [PMID: 33862221 PMCID: PMC9768867 DOI: 10.1016/j.semcancer.2021.04.006] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/24/2022]
Abstract
The intrinsic mechanisms sensing the imbalance of energy in cells are pivotal for cell survival under various environmental insults. AMP-activated protein kinase (AMPK) serves as a central guardian maintaining energy homeostasis by orchestrating diverse cellular processes, such as lipogenesis, glycolysis, TCA cycle, cell cycle progression and mitochondrial dynamics. Given that AMPK plays an essential role in the maintenance of energy balance and metabolism, managing AMPK activation is considered as a promising strategy for the treatment of metabolic disorders such as type 2 diabetes and obesity. Since AMPK has been attributed to aberrant activation of metabolic pathways, mitochondrial dynamics and functions, and epigenetic regulation, which are hallmarks of cancer, targeting AMPK may open up a new avenue for cancer therapies. Although AMPK is previously thought to be involved in tumor suppression, several recent studies have unraveled its tumor promoting activity. The double-edged sword characteristics for AMPK as a tumor suppressor or an oncogene are determined by distinct cellular contexts. In this review, we will summarize recent progress in dissecting the upstream regulators and downstream effectors for AMPK, discuss the distinct roles of AMPK in cancer regulation and finally offer potential strategies with AMPK targeting in cancer therapy.
Collapse
Affiliation(s)
- Che-Chia Hsu
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC, 27101, USA
| | - Danni Peng
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC, 27101, USA
| | - Zhen Cai
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC, 27101, USA.
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston-Salem, NC, 27101, USA.
| |
Collapse
|