1
|
Yuan Z, He J, Li Z, Fan B, Zhang L, Man X. Targeting autophagy in urological system cancers: From underlying mechanisms to therapeutic implications. Biochim Biophys Acta Rev Cancer 2024; 1879:189196. [PMID: 39426690 DOI: 10.1016/j.bbcan.2024.189196] [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: 07/29/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
The urological system, including kidneys, ureters, bladder, urethra and prostate is known to be vital for blood filtration, waste elimination and electrolyte balance. Notably, urological system cancers represent a significant portion of global cancer diagnoses and mortalities. The current therapeutic strategies for early-stage cancer primarily involve resection surgery, which significantly affects the quality of life of patients, whereas advanced-stage cancer often relies on less effective chemo- or radiotherapy. Recently, accumulating evidence has revealed that autophagy, a crucial process in which excess organelles or inclusions within cells are removed to maintain cell homeostasis, has numerous links to urological system cancers. In this review, we focus on summarizing the underlying two-sided mechanisms of autophagy in urological system cancers. We also review the current clinical drugs targeting autophagy, which demonstrate significant potential in improving treatment outcomes for urological system cancers. In addition, we provide an overview of the research status of novel small molecule compounds targeting autophagy that are in the preclinical stages of investigation. Furthermore, drug combinations based on autophagy modulation strategies in urological system cancers are systematically summarized and discussed. These findings provide comprehensive new insight for the future discovery of more autophagy-related drug candidates.
Collapse
Affiliation(s)
- Ziyue Yuan
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiani He
- Department of Urology, Department of Surgical Oncology and Breast Surgery, Institute of Urology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zhijia Li
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Bo Fan
- Department of Urology, Institute of Precision Drug Innovation and Cancer Center, Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China.
| | - Lan Zhang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China; Department of Urology, Institute of Precision Drug Innovation and Cancer Center, Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China.
| | - Xiaojun Man
- Department of Urology, Department of Surgical Oncology and Breast Surgery, Institute of Urology, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
2
|
Sonis ST, Villa A. A New Hypothesis Describing the Pathogenesis of Oral Mucosal Injury Associated with the Mammalian Target of Rapamycin (mTOR) Inhibitors. Cancers (Basel) 2023; 16:68. [PMID: 38201496 PMCID: PMC10777973 DOI: 10.3390/cancers16010068] [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: 11/27/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
It has been 24 years since rapamycin (sirolimus) was approved to mitigate solid organ transplant rejection and 16 years since mTOR (mammalian/mechanistic target of rapamycin) inhibitors reached patients as a cancer therapy. While the clinical benefits of mTOR inhibitors (mTORi) are robust, so too are their toxicities. Among the most common issues is the development of ulcers of the oral mucosa (mTOR-inhibitor associated stomatitis; mIAS). These lesions are distinct from those of other anti-cancer agents, occur with regularity, and impact patient outcomes. mIAS' pathogenesis has been the subject of speculation, and its similar presentation to recurrent aphthous stomatitis (RAS) has led to the hypothesis that it might serve as a surrogate to better understand RAS. Based on a review of the literature, the current manuscript provides a hypothesis regarding the mechanisms by which mTORis uniquely initiate mucosal injury and an explanation for the observation that steroids (also an immunosuppressive) are effective in its treatment through a non-immunologic mechanism. Unexplained unique features of mIAS are discussed in this review in the context of future investigation.
Collapse
Affiliation(s)
- Stephen T. Sonis
- Divisions of Oral Medicine and Dentistry, Brigham and Women’s Hospital and the Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02114, USA
- Biomodels, LLC, Waltham, MA 02451, USA
| | - Alessandro Villa
- Oral Medicine, Oral Oncology and Dentistry, Miami Cancer Institute, Miami, FL 33176, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
3
|
Roth L, Dogan S, Tuna BG, Aranyi T, Benitez S, Borrell-Pages M, Bozaykut P, De Meyer GRY, Duca L, Durmus N, Fonseca D, Fraenkel E, Gillery P, Giudici A, Jaisson S, Johansson M, Julve J, Lucas-Herald AK, Martinet W, Maurice P, McDonnell BJ, Ozbek EN, Pucci G, Pugh CJA, Rochfort KD, Roks AJM, Rotllan N, Shadiow J, Sohrabi Y, Spronck B, Szeri F, Terentes-Printzios D, Tunc Aydin E, Tura-Ceide O, Ucar E, Yetik-Anacak G. Pharmacological modulation of vascular ageing: A review from VascAgeNet. Ageing Res Rev 2023; 92:102122. [PMID: 37956927 DOI: 10.1016/j.arr.2023.102122] [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: 07/05/2023] [Revised: 10/27/2023] [Accepted: 11/09/2023] [Indexed: 11/20/2023]
Abstract
Vascular ageing, characterized by structural and functional changes in blood vessels of which arterial stiffness and endothelial dysfunction are key components, is associated with increased risk of cardiovascular and other age-related diseases. As the global population continues to age, understanding the underlying mechanisms and developing effective therapeutic interventions to mitigate vascular ageing becomes crucial for improving cardiovascular health outcomes. Therefore, this review provides an overview of the current knowledge on pharmacological modulation of vascular ageing, highlighting key strategies and promising therapeutic targets. Several molecular pathways have been identified as central players in vascular ageing, including oxidative stress and inflammation, the renin-angiotensin-aldosterone system, cellular senescence, macroautophagy, extracellular matrix remodelling, calcification, and gasotransmitter-related signalling. Pharmacological and dietary interventions targeting these pathways have shown potential in ameliorating age-related vascular changes. Nevertheless, the development and application of drugs targeting vascular ageing is complicated by various inherent challenges and limitations, such as certain preclinical methodological considerations, interactions with exercise training and sex/gender-related differences, which should be taken into account. Overall, pharmacological modulation of endothelial dysfunction and arterial stiffness as hallmarks of vascular ageing, holds great promise for improving cardiovascular health in the ageing population. Nonetheless, further research is needed to fully elucidate the underlying mechanisms and optimize the efficacy and safety of these interventions for clinical translation.
Collapse
Affiliation(s)
- Lynn Roth
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.
| | - Soner Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, Istanbul, Turkiye
| | - Bilge Guvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkiye
| | - Tamas Aranyi
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary; Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Sonia Benitez
- CIBER de Diabetes y enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Cardiovascular Biochemistry, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Maria Borrell-Pages
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
| | - Perinur Bozaykut
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkiye
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France
| | - Nergiz Durmus
- Department of Pharmacology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkiye
| | - Diogo Fonseca
- Laboratory of Pharmacology and Pharmaceutical Care, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Emil Fraenkel
- 1st Department of Internal Medicine, University Hospital, Pavol Jozef Šafárik University of Košice, Košice, Slovakia
| | - Philippe Gillery
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France; Laboratoire de Biochimie-Pharmacologie-Toxicologie, Centre Hospitalier et Universitaire de Reims, Reims, France
| | - Alessandro Giudici
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, the Netherlands
| | - Stéphane Jaisson
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France; Laboratoire de Biochimie-Pharmacologie-Toxicologie, Centre Hospitalier et Universitaire de Reims, Reims, France
| | | | - Josep Julve
- CIBER de Diabetes y enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Endocrinology, Diabetes and Nutrition group, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | | | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Pascal Maurice
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2 "Matrix Aging and Vascular Remodelling", Université de Reims Champagne Ardenne (URCA), Reims, France
| | - Barry J McDonnell
- Centre for Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, UK
| | - Emine Nur Ozbek
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkiye
| | - Giacomo Pucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Christopher J A Pugh
- Centre for Cardiovascular Health and Ageing, Cardiff Metropolitan University, Cardiff, UK
| | - Keith D Rochfort
- School of Nursing, Psychotherapy, and Community Health, Dublin City University, Dublin, Ireland
| | - Anton J M Roks
- Department of Internal Medicine, Division of Vascular Disease and Pharmacology, Erasmus Medical Center, Erasmus University, Rotterdam, the Netherlands
| | - Noemi Rotllan
- CIBER de Diabetes y enfermedades Metabólicas asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Pathophysiology of lipid-related diseases, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - James Shadiow
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Yahya Sohrabi
- Molecular Cardiology, Dept. of Cardiology I - Coronary and Peripheral Vascular Disease, University Hospital Münster, Westfälische Wilhelms-Universität, 48149 Münster, Germany; Department of Medical Genetics, Third Faculty of Medicine, Charles University, 100 00 Prague, Czechia
| | - Bart Spronck
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, the Netherlands; Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Australia
| | - Flora Szeri
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
| | - Dimitrios Terentes-Printzios
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Elif Tunc Aydin
- Department of Cardiology, Hospital of Ataturk Training and Research Hospital, Katip Celebi University, Izmir, Turkiye
| | - Olga Tura-Ceide
- Biomedical Research Institute-IDIBGI, Girona, Spain; Department of Pulmonary Medicine, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS); University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias, Madrid, Spain
| | - Eda Ucar
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkiye
| | - Gunay Yetik-Anacak
- Department of Pharmacology, Faculty of Pharmacy, Ege University, Izmir, Turkiye; Department of Pharmacology, Faculty of Pharmacy, Acıbadem Mehmet Aydinlar University, Istanbul, Turkiye.
| |
Collapse
|
4
|
Optimization of the Solvent and In Vivo Administration Route of Auranofin in a Syngeneic Non-Small Cell Lung Cancer and Glioblastoma Mouse Model. Pharmaceutics 2022; 14:pharmaceutics14122761. [PMID: 36559255 PMCID: PMC9783082 DOI: 10.3390/pharmaceutics14122761] [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: 10/24/2022] [Revised: 11/29/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
The antineoplastic activity of the thioredoxin reductase 1 (TrxR) inhibitor, auranofin (AF), has already been investigated in various cancer mouse models as a single drug, or in combination with other molecules. However, there are inconsistencies in the literature on the solvent, dose and administration route of AF treatment in vivo. Therefore, we investigated the solvent and administration route of AF in a syngeneic SB28 glioblastoma (GBM) C57BL/6J and a 344SQ non-small cell lung cancer 129S2/SvPasCrl (129) mouse model. Compared to daily intraperitoneal injections and subcutaneous delivery of AF via osmotic minipumps, oral gavage for 14 days was the most suitable administration route for high doses of AF (10-15 mg/kg) in both mouse models, showing no measurable weight loss or signs of toxicity. A solvent comprising 50% DMSO, 40% PEG300 and 10% ethanol improved the solubility of AF for oral administration in mice. In addition, we confirmed that AF was a potent TrxR inhibitor in SB28 GBM tumors at high doses. Taken together, our results and results in the literature indicate the therapeutic value of AF in several in vivo cancer models, and provide relevant information about AF's optimal administration route and solvent in two syngeneic cancer mouse models.
Collapse
|
5
|
Gui Z, Suo C, Tao J, Wang Z, Zheng M, Fei S, Chen H, Sun L, Han Z, Ju X, Zhang H, Gu M, Tan R. Everolimus Alleviates Renal Allograft Interstitial Fibrosis by Inhibiting Epithelial-to-Mesenchymal Transition Not Only via Inducing Autophagy but Also via Stabilizing IκB-α. Front Immunol 2022; 12:753412. [PMID: 35140705 PMCID: PMC8818677 DOI: 10.3389/fimmu.2021.753412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 12/14/2021] [Indexed: 11/25/2022] Open
Abstract
Chronic allograft dysfunction (CAD) is the major cause of late graft loss in long-term renal transplantation. In our previous study, we found that epithelial–mesenchymal transition (EMT) is a significant event in the progression of renal allograft tubulointerstitial fibrosis, and impaired autophagic flux plays a critical role in renal allograft fibrosis. Everolimus (EVR) has been reported to be widely used to prevent the progression of organ fibrosis and graft rejection. However, the pharmacological mechanism of EVR in kidney transplantation remains to be determined. We used CAD rat model and the human kidney 2 (HK2) cell line treated with tumor necrosis factor-α (TNF-α) and EVR to examine the role of EVR on TNF-α-induced EMT and transplanted renal interstitial fibrosis. Here, we found that EVR could attenuate the progression of EMT and renal allograft interstitial fibrosis, and also activate autophagy in vivo. To explore the mechanism behind it, we detected the relationship among EVR, autophagy level, and TNF-α-induced EMT in HK2 cells. Our results showed that autophagy was upregulated upon mTOR pathway inhibition by EVR, which could significantly reduce expression of TNF-α-induced EMT. However, the inhibition of EVR on TNF-α-induced EMT was partly reversed following the addition of autophagy inhibitor chloroquine. In addition, we found that TNF-α activated EMT through protein kinase B (Akt) as well as nuclear factor kappa B (NF-κB) pathway according to the RNA sequencing, and EVR’s effect on the EMT was only associated with IκB-α stabilization instead of the Akt pathway. Together, our findings suggest that EVR may retard impaired autophagic flux and block NF-κB pathway activation, and thereby prevent progression of TNF-α-induced EMT and renal allograft interstitial fibrosis.
Collapse
Affiliation(s)
- Zeping Gui
- Department of Urology, the Second Affiliated Hospital With Nanjing Medical University, Nanjing, China
- Department of Urology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Chuanjian Suo
- Department of Urology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Jun Tao
- Department of Urology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Zijie Wang
- Department of Urology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Ming Zheng
- Department of Urology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Shuang Fei
- Department of Urology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Hao Chen
- Department of Urology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Li Sun
- Department of Urology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Zhijian Han
- Department of Urology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Xiaobing Ju
- Department of Urology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Hengcheng Zhang
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- *Correspondence: Ruoyun Tan, ; Min Gu, ; Hengcheng Zhang,
| | - Min Gu
- Department of Urology, the Second Affiliated Hospital With Nanjing Medical University, Nanjing, China
- *Correspondence: Ruoyun Tan, ; Min Gu, ; Hengcheng Zhang,
| | - Ruoyun Tan
- Department of Urology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
- *Correspondence: Ruoyun Tan, ; Min Gu, ; Hengcheng Zhang,
| |
Collapse
|
6
|
Poznyak AV, Sukhorukov VN, Zhuravlev A, Orekhov NA, Kalmykov V, Orekhov AN. Modulating mTOR Signaling as a Promising Therapeutic Strategy for Atherosclerosis. Int J Mol Sci 2022; 23:ijms23031153. [PMID: 35163076 PMCID: PMC8835022 DOI: 10.3390/ijms23031153] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/14/2022] Open
Abstract
For more than a decade, atherosclerosis has been one of the leading causes of death in developed countries. The issue of treatment and prevention of the disease is especially acute. Despite the huge amount of basic and clinical research, a significant number of gaps remain in our understanding of the pathogenesis of atherosclerosis, and only their closure will bring us closer to understanding the causes of the disease at the cellular and molecular levels and, accordingly, to the development of an effective treatment. One of the seemingly well-studied elements of atherogenesis is the mTOR signaling pathway. However, more and more new details are still being clarified. Therapeutic strategies associated with rapamycin have worked well in a number of different diseases, and there is every reason to believe that targeting components of the mTOR pathway may pay off in atherosclerosis as well.
Collapse
Affiliation(s)
- Anastasia V. Poznyak
- Skolkovo Innovative Center, Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, 121609 Moscow, Russia;
- Correspondence: (A.V.P.); (A.N.O.)
| | - Vasily N. Sukhorukov
- AP Avtsyn Research Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia; (V.N.S.); (A.Z.); (V.K.)
- National Medical Research Center of Cardiology, Institute of Experimental Cardiology, 15A 3-rd Cherepkovskaya Street, 121552 Moscow, Russia
| | - Alexander Zhuravlev
- AP Avtsyn Research Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia; (V.N.S.); (A.Z.); (V.K.)
| | - Nikolay A. Orekhov
- Skolkovo Innovative Center, Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, 121609 Moscow, Russia;
| | - Vladislav Kalmykov
- AP Avtsyn Research Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia; (V.N.S.); (A.Z.); (V.K.)
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Alexander N. Orekhov
- Skolkovo Innovative Center, Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, 121609 Moscow, Russia;
- AP Avtsyn Research Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia; (V.N.S.); (A.Z.); (V.K.)
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| |
Collapse
|
7
|
Chen C, Wang S, Yu L, Mueller J, Fortunato F, Rausch V, Mueller S. H 2O 2-mediated autophagy during ethanol metabolism. Redox Biol 2021; 46:102081. [PMID: 34343907 PMCID: PMC8350071 DOI: 10.1016/j.redox.2021.102081] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/15/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Alcoholic liver disease (ALD) is the most common liver disease worldwide and its underlying molecular mechanisms are still poorly understood. Moreover, conflicting data have been reported on potentially protective autophagy, the exact role of ethanol-metabolizing enzymes and ROS. METHODS Expression of LC3B, CYP2E1, and NOX4 was studied in a mouse model of acute ethanol exposure by immunoblotting and immunohistochemistry. Autophagy was further studied in primary mouse hepatocytes and huh7 cells in response to ethanol and its major intermediator acetaldehyde. Experiments were carried out in cells overexpressing CYP2E1 and knock down of NOX4 using siRNA. The response to external H2O2 was studied by using the GOX/CAT system. Autophagic flux was monitored using the mRFP-GFP-LC3 plasmid, while rapamycin and chloroquine served as positive and negative controls. RESULTS Acute ethanol exposure of mice over 24 h significantly induced autophagy as measured by LC3B expression but also induced the ROS-generating CYP2E1 and NOX4 enzymes. Notably, ethanol but not its downstream metabolite acetaldehyde induced autophagy in primary mouse hepatocytes. In contrast, autophagy could only be induced in huh7 cells in the presence of overexpressed CYP2E1. In addition, overexpression of NOX4 also significantly increased autophagy, which could be blocked by siRNA mediated knock down. The antioxidant N-acetylcysteine (NAC) also efficiently blocked CYP2E1-and NOX4-mediated induction of autophagy. Finally, specific and non-toxic production of H2O2 by the GOX/CAT system as evidenced by elevated peroxiredoxin (Prx-2) also induced LC3B which was efficiently blocked by NAC. H2O2 strongly increased the autophagic flux as measured by mRFP-GFP-LC3 plasmid. CONCLUSION We here provide evidence that short-term ethanol exposure induces autophagy in hepatocytes both in vivo and in vitro through the generation of ROS. These data suggest that suppression of autophagy by ethanol is most likely due to longer alcohol exposure during chronic alcohol consumption with the accumulation of e.g. misfolded proteins.
Collapse
Affiliation(s)
- Cheng Chen
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Shijin Wang
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Linna Yu
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Johannes Mueller
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Franco Fortunato
- Department of Surgery, University of Heidelberg, Heidelberg, Germany
| | - Vanessa Rausch
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Sebastian Mueller
- Center for Alcohol Research and Salem Medical Center, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
8
|
Bansal S, Agrawal M, Mahendiratta S, Kumar S, Arora S, Joshi R, Prajapat M, Sarma P, Prakash A, Chopra K, Medhi B. Everolimus: A potential therapeutic agent targeting PI3K/Akt pathway in brain insulin system dysfunction and associated neurobehavioral deficits. Fundam Clin Pharmacol 2021; 35:1018-1031. [PMID: 33783880 DOI: 10.1111/fcp.12677] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND It is well accepted that PI3k/Akt signaling pathway is a potential therapeutic window which regulates metabolism and energy homeostasis within the brain, and is an important mediator of normal neuronal physiological functions. Dysregulation of this pathway results in impaired insulin signaling, learning and memory and neuronal survival. OBJECTIVES Elucidating the role of everolimus in intracerebroventricular (ICV) streptozotocin induced Insulin/IGF-1 dependent PI3K/Akt/mTOR pathway dysregulation and associated neurobehavioral deficits. METHODS Rats were administered with streptozotocin (3 mg/kg) intracerebroventricular, followed by administration of everolimus (1 mg/kg) orally for 21 days. After that, Morris water maze and passive avoidance tests were performed for assessment of memory. Animals were sacrificed to evaluate brain insulin pathway dysfunction, neurotrophic, apoptotic, inflammatory, and biochemical markers in rat brain. To elucidate the mechanism of action of everolimus, PI3K inhibitor, wortmannin was administered in the presence of everolimus in one group. RESULTS Streptozotocin administration resulted in a significant decrease of brain insulin, insulin growth factor-1 levels, and alterations in behavioral, neurotrophic (BDNF), inflammatory (TNF-α), apoptotic (NF-κB, Bcl2 and Bax) and biochemical (AChE and ChAT assay) parameters in comparison to sham group rats. Everolimus significantly mitigated the deleterious behavioral, biochemical, and molecular changes in rats having central insulin dysfunction. However, the protective effect of everolimus was completely abolished when it was administered in the presence of wortmannin. CONCLUSION Findings from the study reveal that mTOR inhibitors can be an important treatment strategy for neurobehavioral deficits occurring due to central insulin pathway dysfunction. Protective effect of drugs is via modulation of PI3K/Akt pathway.
Collapse
Affiliation(s)
- Seema Bansal
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Madhunika Agrawal
- Department of Pharmacology, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Saniya Mahendiratta
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Subodh Kumar
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Shiyana Arora
- Department of Pharmacology, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Rupa Joshi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Manisha Prajapat
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Phulen Sarma
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ajay Prakash
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Kanwaljit Chopra
- Department of Pharmacology, University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
9
|
Marques ARA, Ramos C, Machado-Oliveira G, Vieira OV. Lysosome (Dys)function in Atherosclerosis-A Big Weight on the Shoulders of a Small Organelle. Front Cell Dev Biol 2021; 9:658995. [PMID: 33855029 PMCID: PMC8039146 DOI: 10.3389/fcell.2021.658995] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/11/2021] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is a progressive insidious chronic disease that underlies most of the cardiovascular pathologies, including myocardial infarction and ischemic stroke. The malfunctioning of the lysosomal compartment has a central role in the etiology and pathogenesis of atherosclerosis. Lysosomes are the degradative organelles of mammalian cells and process endogenous and exogenous substrates in a very efficient manner. Dysfunction of these organelles and consequent inefficient degradation of modified low-density lipoproteins (LDL) and apoptotic cells in atherosclerotic lesions have, therefore, numerous deleterious consequences for cellular homeostasis and disease progression. Lysosome dysfunction has been mostly studied in the context of the inherited lysosomal storage disorders (LSDs). However, over the last years it has become increasingly evident that the consequences of this phenomenon are more far-reaching, also influencing the progression of multiple acquired human pathologies, such as neurodegenerative diseases, cancer, and cardiovascular diseases (CVDs). During the formation of atherosclerotic plaques, the lysosomal compartment of the various cells constituting the arterial wall is under severe stress, due to the tremendous amounts of lipoproteins being processed by these cells. The uncontrolled uptake of modified lipoproteins by arterial phagocytic cells, namely macrophages and vascular smooth muscle cells (VSMCs), is the initial step that triggers the pathogenic cascade culminating in the formation of atheroma. These cells become pathogenic "foam cells," which are characterized by dysfunctional lipid-laden lysosomes. Here, we summarize the current knowledge regarding the origin and impact of the malfunctioning of the lysosomal compartment in plaque cells. We further analyze how the field of LSD research may contribute with some insights to the study of CVDs, particularly how therapeutic approaches that target the lysosomes in LSDs could be applied to hamper atherosclerosis progression and associated mortality.
Collapse
Affiliation(s)
- André R A Marques
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Cristiano Ramos
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Gisela Machado-Oliveira
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Otília V Vieira
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
10
|
Elliott E, Bailey O, Waldron FM, Hardingham GE, Chandran S, Gregory JM. Therapeutic Targeting of Proteostasis in Amyotrophic Lateral Sclerosis-a Systematic Review and Meta-Analysis of Preclinical Research. Front Neurosci 2020; 14:511. [PMID: 32523508 PMCID: PMC7261930 DOI: 10.3389/fnins.2020.00511] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/24/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Amyotrophic lateral sclerosis (ALS) is a rapidly progressive fatal neurodegenerative condition. There are no effective treatments. The only globally licensed medication, that prolongs life by 2–3 months, was approved by the FDA in 1995. One reason for the absence of effective treatments is disease heterogeneity noting that ALS is clinically heterogeneous and can be considered to exist on a neuropathological spectrum with frontotemporal dementia. Despite this significant clinical heterogeneity, protein misfolding has been identified as a unifying pathological feature in these cases. Based on this shared pathophysiology, we carried out a systematic review and meta-analysis to assess the therapeutic efficacy of compounds that specifically target protein misfolding in preclinical studies of both ALS and FTD. Methods: Three databases: (i) PubMed, (ii) MEDLINE, and (iii) EMBASE were searched. All studies comparing the effect of treatments targeting protein misfolding in pre-clinical ALS or FTD models to a control group were retrieved. Results: Systematic review identified 70 pre-clinical studies investigating the effects of therapies targeting protein misfolding on survival. Meta-analysis revealed that targeting protein misfolding did significantly improve survival compared to untreated controls (p < 0.001, df = 68, α = 0.05, CI 1.05–1.16), with no evidence of heterogeneity between studies (I2 = 0%). Further subgroup analyses, evaluating the effect of timing of these interventions, showed that, only treating prior to symptom onset (n = 33), significantly improved survival (p < 0.001, df = 31, α = 0.05, CI 1.08–1.29), although this likely reflects the inadequate sample size of later time points. Furthermore, arimoclomol was found to significantly reduce secondary outcome measures including: (i) histological outcomes, (ii) behavioral outcomes, and (iii) biochemical outcomes (p < 0.005). Conclusions: This analysis supports the hypothesis that protein misfolding plays an important role in the pathogenesis of ALS and FTD and that targeting protein misfolding, at least in pre-clinical models, can significantly improve survival, especially if such an intervention is administered prior to symptom onset.
Collapse
Affiliation(s)
- Elizabeth Elliott
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.,Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom.,The Anne Rowling Regenerative Neurology Clinic, The University of Edinburgh, Edinburgh, United Kingdom.,The Euan MacDonald Centre, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Neuroscience, The University of Edinburgh, Edinburgh, United Kingdom.,MRC Edinburgh Brain Bank, Academic Department of Neuropathology, The University of Edinburgh, Edinburgh, United Kingdom
| | - Olivia Bailey
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom.,The Anne Rowling Regenerative Neurology Clinic, The University of Edinburgh, Edinburgh, United Kingdom.,The Euan MacDonald Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Fergal M Waldron
- Ashworth Laboratories, Institute of Evolutionary Biology and Centre for Immunity Infection and Evolution, University of Edinburgh, Edinburgh, United Kingdom
| | - Giles E Hardingham
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.,The Euan MacDonald Centre, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Siddharthan Chandran
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.,Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom.,The Anne Rowling Regenerative Neurology Clinic, The University of Edinburgh, Edinburgh, United Kingdom.,The Euan MacDonald Centre, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Neuroscience, The University of Edinburgh, Edinburgh, United Kingdom.,Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom.,Centre for Brain Development and Repair, inStem, Bangalore, India.,MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Jenna M Gregory
- UK Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.,Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom.,The Anne Rowling Regenerative Neurology Clinic, The University of Edinburgh, Edinburgh, United Kingdom.,The Euan MacDonald Centre, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Neuroscience, The University of Edinburgh, Edinburgh, United Kingdom.,MRC Edinburgh Brain Bank, Academic Department of Neuropathology, The University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Pathology, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
11
|
De Munck DG, De Meyer GR, Martinet W. Autophagy as an emerging therapeutic target for age-related vascular pathologies. Expert Opin Ther Targets 2020; 24:131-145. [PMID: 31985292 DOI: 10.1080/14728222.2020.1723079] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: The incidence of age-related vascular diseases such as arterial stiffness, hypertension and atherosclerosis, is rising dramatically and is substantially impacting healthcare systems. Mounting evidence suggests that there is an important role for autophagy in maintaining (cardio)vascular health. Impaired vascular autophagy has been linked to arterial aging and the initiation of vascular disease.Areas covered: The function and implications of autophagy in vascular smooth muscle cells and endothelial cells are discussed in healthy blood vessels and arterial disease. Furthermore, we discuss current treatment options for vascular disease and their links with autophagy. A literature search was conducted in PubMed up to October 2019.Expert opinion: Although the therapeutic potential of inducing autophagy in age-related vascular pathologies is considerable, several issues should be addressed before autophagy induction can be clinically used to treat vascular disease. These issues include uncertainty regarding the most effective drug target as well as the lack of potency and selectivity of autophagy inducing drugs. Moreover, drug tolerance or autophagy mediated cell death have been reported as possible adverse effects. Special attention is required for determining the cause of autophagy deficiency to optimize the treatment strategy.
Collapse
Affiliation(s)
- Dorien G De Munck
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Guido Ry De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
12
|
Accumulation of PNPLA3 on lipid droplets is the basis of associated hepatic steatosis. Proc Natl Acad Sci U S A 2019; 116:9521-9526. [PMID: 31019090 PMCID: PMC6511016 DOI: 10.1073/pnas.1901974116] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Fatty liver disease (FLD) is a disorder in which accumulation of triglycerides (TGs) in the liver can lead to inflammation, fibrosis, and cirrhosis. Previously, we identified a variant (I148M) in patatin-like phospholipase domain-containing protein 3 (PNPLA3) that is strongly associated with FLD, but the mechanistic basis for the association remains elusive. Although PNPLA3 has TG hydrolase activity in vitro, inactivation or overexpression of the WT protein in mice does not cause steatosis. In contrast, expression of two catalytically defective forms of PNPLA3 (I148M or S47A) in sucrose-fed mice causes accumulation of both PNPLA3 and TGs on hepatic lipid droplets (LDs). To determine if amassing PNPLA3 on LDs is a cause or consequence of steatosis, we engineered a synthetic isoform of PNPLA3 that uncouples protein accumulation from loss of enzymatic activity. Expression of a ubiquitylation-resistant form of PNPLA3 in mice caused accumulation of PNPLA3 on hepatic LDs and development of FLD. Lowering PNPLA3 levels by either shRNA knockdown or proteolysis-targeting chimera (PROTAC)-mediated degradation reduced liver TG content in mice overexpressing PNPLA3(148M). Taken together, our results show that the steatosis associated with PNPLA3(148M) is caused by accumulation of PNPLA3 on LDs.
Collapse
|
13
|
Kurdi A, Roth L, Van der Veken B, Van Dam D, De Deyn PP, De Doncker M, Neels H, De Meyer GR, Martinet W. Everolimus depletes plaque macrophages, abolishes intraplaque neovascularization and improves survival in mice with advanced atherosclerosis. Vascul Pharmacol 2019; 113:70-76. [DOI: 10.1016/j.vph.2018.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/31/2018] [Accepted: 12/23/2018] [Indexed: 01/12/2023]
|
14
|
Dolcetta D, Dominici R. The local mammalian target of rapamycin (mTOR) modulation: a promising strategy to counteract neurodegeneration. Neural Regen Res 2019; 14:1711-1712. [PMID: 31169184 PMCID: PMC6585559 DOI: 10.4103/1673-5374.257524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
| | - Roberto Dominici
- Clinical Chemistry Laboratory, Magenta Hospital, ASST Ovest Milanese, Milan, Italy
| |
Collapse
|
15
|
Pupyshev AB, Korolenko TA, Tikhonova MA. Effects and Mechanisms of Rapamycin Action on Experimental Neurodegeneration. NEUROCHEM J+ 2018. [DOI: 10.1134/s1819712418030108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
16
|
Cassano T, Magini A, Giovagnoli S, Polchi A, Calcagnini S, Pace L, Lavecchia MA, Scuderi C, Bronzuoli MR, Ruggeri L, Gentileschi MP, Romano A, Gaetani S, De Marco F, Emiliani C, Dolcetta D. Early intrathecal infusion of everolimus restores cognitive function and mood in a murine model of Alzheimer's disease. Exp Neurol 2018; 311:88-105. [PMID: 30243986 DOI: 10.1016/j.expneurol.2018.09.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/13/2018] [Accepted: 09/17/2018] [Indexed: 01/05/2023]
Abstract
The discovery that mammalian target of rapamycin (mTOR) inhibition increases lifespan in mice and restores/delays many aging phenotypes has led to the identification of a novel potential therapeutic target for the treatment of Alzheimer's disease (AD). Among mTOR inhibitors, everolimus, which has been developed to improve the pharmacokinetic characteristics of rapamycin, has been extensively profiled in preclinical and clinical studies as anticancer and immunosuppressive agent, but no information is available about its potential effects on neurodegenerative disorders. Using a reliable mouse model of AD (3 × Tg-AD mice), we explored whether short-term treatment with everolimus injected directly into the brain by osmotic pumps was able to modify AD-like pathology with low impact on peripheral organs. We first established in non-transgenic mice the stability of everolimus at 37 °C in comparison with rapamycin and, then, evaluated its pharmacokinetics and pharmacodynamics profiles through either a single peripheral (i.p.) or central (i.c.v.) route of administration. Finally, 6-month-old (symptomatic phase) 3 × Tg-AD mice were treated with continuous infusion of either vehicle or everolimus (0.167 μg/μl/day, i.c.v.) using the osmotic pumps. Four weeks after the beginning of infusion, we tested our hypothesis following an integrated approach, including behavioral (tests for cognitive and depressive-like alterations), biochemical and immunohistochemical analyses. Everolimus (i) showed higher stability than rapamycin at 37 °C, (ii) poorly crossed the blood-brain barrier after i.p. injection, (iii) was slowly metabolized in the brain due to a longer t1/2 in the brain compared to blood, and (iv) was more effective in the CNS when administered centrally compared to a peripheral route. Moreover, the everolimus-induced mTOR inhibition reduced human APP/Aβ and human tau levels and improved cognitive function and depressive-like phenotype in the 3 × Tg-AD mice. The intrathecal infusion of everolimus may be effective to treat early stages of AD-pathology through a short and cyclic administration regimen, with short-term outcomes and a low impact on peripheral organs.
Collapse
Affiliation(s)
- Tommaso Cassano
- Department of Clinical and Experimental Medicine, Medical School, University of Foggia, 71100 Foggia, Italy.
| | - Alessandro Magini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06126 Perugia, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy
| | - Alice Polchi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06126 Perugia, Italy
| | - Silvio Calcagnini
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Lorenzo Pace
- Department of Clinical and Experimental Medicine, Medical School, University of Foggia, 71100 Foggia, Italy
| | - Michele Angelo Lavecchia
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Caterina Scuderi
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Maria Rosanna Bronzuoli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Loredana Ruggeri
- Division of Hematology and Clinical Immunology and Bone Marrow Transplant Program, Department of Medicine, University of Perugia, 06132 Perugia, Italy
| | - Maria Pia Gentileschi
- UOSD SAFU, RiDAIT Dept, The Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Adele Romano
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Silvana Gaetani
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, 00185 Rome, Italy
| | - Federico De Marco
- UOSD SAFU, RiDAIT Dept, The Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06126 Perugia, Italy
| | - Diego Dolcetta
- UOSD SAFU, RiDAIT Dept, The Regina Elena National Cancer Institute, 00144 Rome, Italy.
| |
Collapse
|
17
|
Wu X, Yu J, Yan J, Dai J, Si L, Chi Z, Sheng X, Cui C, Ma M, Tang H, Xu T, Yu H, Kong Y, Guo J. PI3K/AKT/mTOR pathway inhibitors inhibit the growth of melanoma cells with mTOR H2189Y mutations in vitro. Cancer Biol Ther 2018; 19:584-589. [PMID: 29708815 DOI: 10.1080/15384047.2018.1435221] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
mTOR is an important therapeutic target in many types of cancers. In melanoma, the mTOR nonsynonymous mutation rate is up to 10.4%. However, mTOR inhibitors have shown limited effects in clinical trials of melanoma. Because mTOR mutations are distributed, not selecting patients with specific mTOR mutations may be the main reason for therapeutic failures. Our previous research found that mutations in the mTOR P2213S and S2215Y kinase domains resulted in gain-of-function and were sensitive to specific inhibitors. The purpose of this study was to test the effect of heterozygous/homozygous H2189Y mutations on downstream pathways and sensitivity to inhibitors. mTOR kinase activity was analyzed by western blot and a K-LISA™ mTOR activity kit. The sensitivity of melanoma cells to inhibitors was tested by a proliferation assay. The expression of downstream pathway proteins was also analyzed by western blot. The results showed that heterozygous/homozygous H2189Y mutations were gain-of-function. The heterozygous H2189Y mutation was sensitive to the AKT inhibitor, AZD5363, and the phosphoinositide 3-kinase inhibitor, LY294002. The homozygous H2189Y mutation was sensitive to the mTOR inhibitor, everolimus, and the AKT inhibitors AZD5363 and MK-2206 2HCL,and the phosphoinositide 3-kinase inhibitor, LY294002. These results indicated that homozygous mutations in the kinase domain have a greater effect on protein function than heterozygous mutations. The mTOR kinase domain may play an important role in mTOR kinase activity and may be the target of selective inhibitors. Our study can facilitate the selection of appropriate inhibitors for patients in clinical trials.
Collapse
Affiliation(s)
- Xiaowen Wu
- a Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma , Peking University Cancer Hospital & Institute , Beijing , China
| | - Jiayi Yu
- a Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma , Peking University Cancer Hospital & Institute , Beijing , China
| | - Junya Yan
- a Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma , Peking University Cancer Hospital & Institute , Beijing , China
| | - Jie Dai
- a Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma , Peking University Cancer Hospital & Institute , Beijing , China
| | - Lu Si
- a Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma , Peking University Cancer Hospital & Institute , Beijing , China
| | - Zhihong Chi
- a Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma , Peking University Cancer Hospital & Institute , Beijing , China
| | - Xinan Sheng
- a Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma , Peking University Cancer Hospital & Institute , Beijing , China
| | - Chuanliang Cui
- a Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma , Peking University Cancer Hospital & Institute , Beijing , China
| | - Meng Ma
- a Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma , Peking University Cancer Hospital & Institute , Beijing , China
| | - Huan Tang
- a Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma , Peking University Cancer Hospital & Institute , Beijing , China
| | - Tianxiao Xu
- a Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma , Peking University Cancer Hospital & Institute , Beijing , China
| | - Huan Yu
- a Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma , Peking University Cancer Hospital & Institute , Beijing , China
| | - Yan Kong
- a Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma , Peking University Cancer Hospital & Institute , Beijing , China
| | - Jun Guo
- a Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Renal Cancer and Melanoma , Peking University Cancer Hospital & Institute , Beijing , China
| |
Collapse
|
18
|
Defective Autophagy in Atherosclerosis: To Die or to Senesce? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7687083. [PMID: 29682164 PMCID: PMC5846382 DOI: 10.1155/2018/7687083] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/29/2017] [Accepted: 01/18/2018] [Indexed: 11/17/2022]
Abstract
Autophagy is a subcellular process that plays an important role in the degradation of proteins and damaged organelles such as mitochondria (a process termed “mitophagy”) via lysosomes. It is crucial for regulating protein and mitochondrial quality control and maintaining cellular homeostasis, whereas dysregulation of autophagy has been implicated in a wide range of diseases including atherosclerosis. Recent evidence has shown that the autophagic process becomes dysfunctional during the progression of atherosclerosis, regardless of whether there are many autophagy-stimulating factors (e.g., reactive oxygen species, oxidized lipids, and cytokines) present within the atherosclerotic plaque. This review highlights the recent insights into the causes and consequences of defective autophagy in atherosclerosis, with a special focus on the role of autophagy and mitophagy in plaque macrophages, vascular smooth muscle cells (VSMCs), and endothelial cells (ECs). It has been shown that defective autophagy can promote apoptosis in macrophages but that it accelerates premature senescence in VSMCs. In the ECs, defective autophagy promotes both apoptosis and senescence. We will discuss the discrepancy between these three cell types in their response to autophagy deficiency and underline the cell type-dependent role of autophagy, which may have important implications for the efficacy of autophagy-targeted treatments for atherosclerosis.
Collapse
|
19
|
Hei C, Liu P, Yang X, Niu J, Li PA. Inhibition of mTOR signaling Confers Protection against Cerebral Ischemic Injury in Acute Hyperglycemic Rats. Int J Biol Sci 2017; 13:878-887. [PMID: 28808420 PMCID: PMC5555105 DOI: 10.7150/ijbs.18976] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 01/28/2017] [Indexed: 01/04/2023] Open
Abstract
Hyperglycemia is known to exacerbate neuronal death resulted from cerebral ischemia. The mechanisms are not fully understood. The mammalian target of rapamycin (mTOR) pathway regulates cell growth, division and apoptosis. Recent studies suggest that activation of mTOR may mediate ischemic brain damage. The objective of the present experiment is to explore whether mTOR mediates ischemic brain damage in acute hyperglycemic animals. Rats were subjected to 10 min of forebrain ischemia under euglycemic, hyperglycemic and rapamycin-treated hyperglycemic conditions. The rat brain samples were collected from the cortex and hippocampi after 3h and 16h of reperfusion. The results showed that hyperglycemia significantly increased neuronal death in the cortex and hippocampus and the exacerbation effect of hyperglycemia was associated with further activation of mTOR under control and/or ischemic conditions. Inhibition of mTOR with rapamycin ameliorated the damage and suppressed hyperglycemia-elevated p-MTOR, p-P70S6K and p-S6. In addition, hyperglycemia per se increased the levels of cytosolic cytochrome c and autophagy marker LC3-II, while rapamycin alleviated these alterations. It is concluded that activation of mTOR signaling may play a detrimental role in mediating the aggravating effect of hyperglycemia on cerebral ischemia.
Collapse
Affiliation(s)
- Changchun Hei
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region and Department Human Anatomy, Histology and Embryology, Ningxia Medical University, Yinchuan 750004, China.,Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| | - Ping Liu
- Department of Endocrinology, General Hospital of Ningxia Medical University, Yinchuan 750004, China.,Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| | - Xiao Yang
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA.,Neuroscience Center, General Hospital of Ningxia Medical University, and Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Yinchuan 750004, China
| | - Jianguo Niu
- Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region and Department Human Anatomy, Histology and Embryology, Ningxia Medical University, Yinchuan 750004, China
| | - P Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA
| |
Collapse
|
20
|
Martinet W, Roth L, De Meyer GRY. Standard Immunohistochemical Assays to Assess Autophagy in Mammalian Tissue. Cells 2017; 6:E17. [PMID: 28665306 PMCID: PMC5617963 DOI: 10.3390/cells6030017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 06/20/2017] [Accepted: 06/26/2017] [Indexed: 12/31/2022] Open
Abstract
Autophagy is a highly conserved lysosomal degradation pathway with major impact on diverse human pathologies. Despite the development of different methodologies to detect autophagy both in vitro and in vivo, monitoring autophagy in tissue via immunohistochemical techniques is hampered due to the lack of biomarkers. Immunohistochemical detection of a punctate pattern of ATG8/MAP1LC3 proteins is currently the most frequently used approach to detect autophagy in situ, but it depends on a highly sensitive detection method and is prone to misinterpretation. Moreover, reliable MAP1LC3 immunohistochemical staining requires correct tissue processing and high-quality, isoform-specific antibodies. Immunohistochemical analysis of other autophagy-related protein targets such as SQSTM1, ubiquitin, ATG5 or lysosomal proteins is not recommended as marker for autophagic activity in tissue for multiple reasons including aspecific labeling of cellular structures and a lack of differential protein expression during autophagy initiation. To better understand the role of autophagy in human disease, novel biomarkers for visualization of the autophagic process with standard histology techniques are urgently needed.
Collapse
Affiliation(s)
- Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium.
| | - Lynn Roth
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium.
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium.
| |
Collapse
|
21
|
Kurdi A, Cleenewerck M, Vangestel C, Lyssens S, Declercq W, Timmermans JP, Stroobants S, Augustyns K, De Meyer GRY, Van Der Veken P, Martinet W. ATG4B inhibitors with a benzotropolone core structure block autophagy and augment efficiency of chemotherapy in mice. Biochem Pharmacol 2017. [PMID: 28642033 DOI: 10.1016/j.bcp.2017.06.119] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Autophagy is a cell survival mechanism hijacked by advanced tumors to endure a rough microenvironment. Late autophagy inhibitors such as (hydroxy)chloroquine have been used clinically to halt tumor progression with modest success. However, given the toxic nature of these compounds and their lack of specificity, novel targets should be considered. We recently identified a benzotropolone derivative that significantly inhibited the essential autophagy protein ATG4B. Therefore, we synthesized and tested additional benzotropolone compounds to identify a promising ATG4B inhibitor that impairs autophagy both in vitro and in vivo. A compound library containing 27 molecules with a benzotropolone backbone was synthesized and screened for inhibition of recombinant ATG4B. Depending on the benzotropolone compound, inhibition of recombinant ATG4B ranged from 3 to 82%. Active compounds were evaluated in cellular assays to confirm inhibition of ATG4B and suppression of autophagy. Seven compounds inhibited processing of the autophagy protein LC3 and autophagosome formation. Compound UAMC-2526 was selected for further in vivo use because of its fair plasma stability. This compound abolished autophagy both in nutrient-deprived GFP-LC3 mice and in CD1-/- Foxn1nu mice bearing HT29 colorectal tumor xenografts. Moreover, addition of UAMC-2526 to the chemotherapy drug oxaliplatin significantly improved inhibition of tumor growth. Our data indicate that suppression of autophagy via ATG4B inhibition is a feasible strategy to augment existing chemotherapy efficacy and to halt tumor progression.
Collapse
Affiliation(s)
- Ammar Kurdi
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Matthias Cleenewerck
- Laboratory of Medicinal Chemistry (UAMC), Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Christel Vangestel
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp, Belgium
| | - Sophie Lyssens
- Laboratory of Medicinal Chemistry (UAMC), Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Wim Declercq
- VIB Inflammation Research Center, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Sigrid Stroobants
- Molecular Imaging Center Antwerp (MICA), University of Antwerp, Antwerp, Belgium
| | - Koen Augustyns
- Laboratory of Medicinal Chemistry (UAMC), Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Pieter Van Der Veken
- Laboratory of Medicinal Chemistry (UAMC), Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
22
|
Moulis M, Vindis C. Methods for Measuring Autophagy in Mice. Cells 2017; 6:cells6020014. [PMID: 28594368 PMCID: PMC5492018 DOI: 10.3390/cells6020014] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/02/2017] [Accepted: 06/03/2017] [Indexed: 12/22/2022] Open
Abstract
Autophagy is a dynamic intracellular process that mediates the degradation of damaged cytoplasmic components by the lysosome. This process plays important roles in maintaining normal cellular homeostasis and energy balance. Measuring autophagy activity is critical and although the determination of autophagic flux in isolated cells is well documented, there is a need to have reliable and quantitative assays to evaluate autophagy in whole organisms. Because mouse models have been precious in establishing the functional significance of autophagy under physiological or pathological conditions, we present in this chapter a compendium of the current available methods to measure autophagy in mice, and discuss their advantages and limitations.
Collapse
Affiliation(s)
- Manon Moulis
- INSERM, UMR-1048, Institute of Metabolic and Cardiovascular Diseases and University Paul Sabatier, F-31342 Toulouse, France.
| | - Cécile Vindis
- INSERM, UMR-1048, Institute of Metabolic and Cardiovascular Diseases and University Paul Sabatier, F-31342 Toulouse, France.
| |
Collapse
|
23
|
Kurdi A, De Doncker M, Leloup A, Neels H, Timmermans JP, Lemmens K, Apers S, De Meyer GRY, Martinet W. Continuous administration of the mTORC1 inhibitor everolimus induces tolerance and decreases autophagy in mice. Br J Pharmacol 2016; 173:3359-3371. [PMID: 27638766 DOI: 10.1111/bph.13626] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/31/2016] [Accepted: 09/05/2016] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND PURPOSE Everolimus is an allosteric inhibitor of the mechanistic target of rapamycin complex 1 (mTORC1) widely known for its potent autophagy stimulating properties. Because everolimus shows poor solubility and stability in aqueous solutions, long-term in vivo administration in preclinical models is challenging. The aim of the present study was to evaluate the effects of short-term and long-term everolimus administration on mTORC1 inhibition and autophagy induction in mice. EXPERIMENTAL APPROACH We developed a vehicle in which everolimus was solubilized and stable at 37°C for at least 1 month. Using osmotic minipumps, GFP microtubule-associated protein light chain 3 transgenic mice were treated continuously either with vehicle or everolimus (1.5 mg·kg-1 per day) for 3 or 28 days. Alternatively, a regimen consisting of intermittent everolimus administration (every other day) for 56 days by oral gavage was used. Autophagy markers and mTORC1 activation status were investigated in the liver. KEY RESULTS As expected, everolimus inhibited mTORC1 and stimulated autophagy in the liver after 3 days of treatment. However, continuous administration for 28 days resulted in hyperactivation of the Akt1-mTORC1 pathway accompanied by a remarkable decrease in autophagy markers. Everolimus given intermittently for 56 days partially rescued mTORC1 sensitivity to the drug but without inducing autophagy. The failure to induce autophagy following long-term everolimus administration was due to uncoupling of the mTORC1 substrate unc-51 like autophagy activating kinase 1. CONCLUSIONS AND IMPLICATIONS Our data encourage the use of intermittent everolimus regimens to prevent tolerance and to extend its activity.
Collapse
Affiliation(s)
- Ammar Kurdi
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Arthur Leloup
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Hugo Neels
- Laboratory for TDM and Toxicology, ZNA Stuivenberg, Antwerp, Belgium.,Toxicological Centre, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Katrien Lemmens
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sandra Apers
- Natural Products and Food Research and Analysis (NatuRA), Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|