1
|
Huang J, Wang J. Selective protein degradation through chaperone‑mediated autophagy: Implications for cellular homeostasis and disease (Review). Mol Med Rep 2025; 31:13. [PMID: 39513615 PMCID: PMC11542157 DOI: 10.3892/mmr.2024.13378] [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: 03/19/2024] [Accepted: 06/03/2024] [Indexed: 11/15/2024] Open
Abstract
Cells rely on autophagy for the degradation and recycling of damaged proteins and organelles. Chaperone-mediated autophagy (CMA) is a selective process targeting proteins for degradation through the coordinated function of molecular chaperones and the lysosome‑associated membrane protein‑2A receptor (LAMP2A), pivotal in various cellular processes from signal transduction to the modulation of cellular responses under stress. In the present review, the intricate regulatory mechanisms of CMA were elucidated through multiple signaling pathways such as retinoic acid receptor (RAR)α, AMP‑activated protein kinase (AMPK), p38‑TEEB‑NLRP3, calcium signaling‑NFAT and PI3K/AKT, thereby expanding the current understanding of CMA regulation. A comprehensive exploration of CMA's versatile roles in cellular physiology were further provided, including its involvement in maintaining protein homeostasis, regulating ferroptosis, modulating metabolic diversity and influencing cell cycle and proliferation. Additionally, the impact of CMA on disease progression and therapeutic outcomes were highlighted, encompassing neurodegenerative disorders, cancer and various organ‑specific diseases. Therapeutic strategies targeting CMA, such as drug development and gene therapy were also proposed, providing valuable directions for future clinical research. By integrating recent research findings, the present review aimed to enhance the current understanding of cellular homeostasis processes and emphasize the potential of targeting CMA in therapeutic strategies for diseases marked by CMA dysfunction.
Collapse
Affiliation(s)
- Jiahui Huang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- College of Traditional Chinese Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Jiazhen Wang
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province and Education Ministry of People's Republic of China, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| |
Collapse
|
2
|
Hu HF, Fu JY, Han L, Gao GB, Zhang WX, Yu SM, Li N, Li YJ, Lu YF, Ding XF, Pan YL, Wang Y, He QY. The Antipsychotic Drug Aripiprazole Suppresses Colorectal Cancer by Targeting LAMP2a to Induce RNH1/miR-99a/mTOR-Mediated Autophagy and Apoptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2409498. [PMID: 39513392 DOI: 10.1002/advs.202409498] [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: 08/12/2024] [Revised: 10/24/2024] [Indexed: 11/15/2024]
Abstract
The mammalian target of rapamycin (mTOR) is a critical signaling hub for sustaining cancer survival. Targeting mTOR and inducing autophagic cell death downstream of it represent promising therapeutic strategies for cancer prevention. A US Food and Drug Administration-approved drug library containing 616 small molecules is used to screen anticancer drugs against colorectal cancer (CRC) cells that rely on mTOR. This led to the identification of an antipsychotic drug aripiprazole, which significantly induced mTOR inhibition and autophagic apoptosis in CRC, in vitro and in vivo. The use of drug affinity response target stability identified lysosome-associated membrane protein 2A (LAMP2a) as a direct target of aripiprazole. LAMP2a-deficient CRC cells are refractory to aripiprazole. High LAMP2a expression is associated with poor survival of patients with CRC and negatively correlated with expression of ribonuclease inhibitor 1 (RNH1), which is later confirmed as a novel substrate of LAMP2a. Mechanistically, aripiprazole bound to the Lys401-His404 of LAMP2a and repressed its activity, subsequently inactivating RNH1/miR-99a/mTOR signaling and inducing autophagy-mediated apoptosis, thereby suppressing tumorigenesis. Liposome-mediated delivery of aripiprazole in combination with fluorouracil elicited superior therapeutic benefits in CRC, as compared to single treatments, thereby highlighting that aripiprazole may be repurposed as a novel therapeutic agent for CRC treatment.
Collapse
Affiliation(s)
- Hui-Fang Hu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
- The First Affiliated Hospital of Jinan University and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China
| | - Jia-Ying Fu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Lei Han
- Institute of Biomedicine and National Engineering Research Center of Genetic Medicine and Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, National Engineering Research Centre of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Gui-Bin Gao
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Wei-Xia Zhang
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Si-Ming Yu
- Institute of Biomedicine and National Engineering Research Center of Genetic Medicine and Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, National Engineering Research Centre of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Nan Li
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yang-Jia Li
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yi-Fan Lu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xiao-Feng Ding
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yun-Long Pan
- The First Affiliated Hospital of Jinan University and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China
| | - Yang Wang
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Qing-Yu He
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
- The First Affiliated Hospital of Jinan University and MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
3
|
Santos de Macedo BG, Albuquerque de Melo M, Pereira-Martins DA, Machado-Neto JA, Traina F. An updated outlook on autophagy mechanism and how it supports acute myeloid leukemia maintenance. Biochim Biophys Acta Rev Cancer 2024; 1879:189214. [PMID: 39515545 DOI: 10.1016/j.bbcan.2024.189214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
The gradual acquisition of genetic and epigenetic disturbances bestows malignant traits upon hematopoietic stem cells, subverting them into a founder and reservoir cell for de novo acute myeloid leukemia (AML) known as leukemic stem cells (LSC). Beyond its molecular heterogeneity, AML is also characterized by rewiring biological processes to support its onset and maintenance. LSC were observed to inherently and actively trigger mitochondrial turnover through selective autophagic removal such that impairing the process led to cell differentiation at the expense of its stemness. This review provides a current take on autophagy regulation mechanisms according to the current molecular characterization of the process; describes autophagy as a drug resistance mechanism, and a pivotal mechanism whereby LSC harmonize their strong reliance on mitochondrial respiration to obtain energy, and their necessity for lower internal oxidative stress to avoid exhaustion. Therefore, targeting autophagy presents a promising strategy to promote long-term remissions in AML.
Collapse
Affiliation(s)
- Brunno Gilberto Santos de Macedo
- Department of Medical Images, Hematology, and Oncology, Ribeirão Preto Medical School of University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Manuela Albuquerque de Melo
- Department of Medical Images, Hematology, and Oncology, Ribeirão Preto Medical School of University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - João Agostinho Machado-Neto
- Department of Medical Images, Hematology, and Oncology, Ribeirão Preto Medical School of University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Pharmacology, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Fabiola Traina
- Department of Medical Images, Hematology, and Oncology, Ribeirão Preto Medical School of University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
4
|
Barmaki H, Nourazarian A, Shademan B, Khaki-Khatibi F. The autophagy paradox: A new hypothesis in neurodegenerative disorders. Neurochem Int 2024; 179:105827. [PMID: 39111406 DOI: 10.1016/j.neuint.2024.105827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024]
Abstract
A recent study showed that while autophagy is usually tied to protein and organelle turnover, it can also play dual roles in neurodegenerative diseases. Traditionally, autophagy was seen as protective since it removes damaged proteins and organelles. but new data suggests autophagy can sometimes promote neuron death. and This review tackles autophagy's seemingly contradictory effects in neurodegeneration, or the "autophagy paradox. " It offers a framework for understanding autophagy in neurodegenerative research and the cellular processes involved. In short, our data uncovers a harmful autophagy role in certain situations, conflicting the view that it's always beneficial. We describe the distinct, disease-specific autophagy pathways functioning in various neurodegenerative diseases. Part two concerns potential therapeutic implications of manipulating autophagy and current strategies targeting the autophagic system, suggesting interesting areas for future research into tailored modulators. This could eventually enable activating or controlling specific autophagy pathways and aid in developing more effective treatments. Researchers believe more molecular-level research is needed so patient-tailored autophagy-modulating therapeutics can be developed given this dilemma. Moreover, research must translate faster into effective neurodegenerative disease treatment options. This article aims to provide a wholly new perspective on autophagy's classically described role in these severe diseases, challenging current dogma and opening new therapeutic avenue options.
Collapse
Affiliation(s)
- Haleh Barmaki
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran; Student Research Committee, Khoy University of Medical Sciences, Khoy, Iran.
| | - Behrouz Shademan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Khaki-Khatibi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
5
|
Duan H, Song S, Li R, Hu S, Zhuang S, Liu S, Li X, Gao W. Strategy for treating MAFLD: Electroacupuncture alleviates hepatic steatosis and fibrosis by enhancing AMPK mediated glycolipid metabolism and autophagy in T2DM rats. Diabetol Metab Syndr 2024; 16:218. [PMID: 39261952 PMCID: PMC11389443 DOI: 10.1186/s13098-024-01432-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 07/24/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Recent studies have highlighted type 2 diabetes (T2DM) as a significant risk factor for the development of metabolic dysfunction-associated fatty liver disease (MAFLD). This investigation aimed to assess electroacupuncture's (EA) impact on liver morphology and function in T2DM rats, furnishing experimental substantiation for its potential to stall MAFLD progression in T2DM. METHODS T2DM rats were induced by a high-fat diet and a single intraperitoneal injection of streptozotocin, and then randomly assigned to five groups: the T2DM group, the electroacupuncture group, the metformin group, combination group of electroacupuncture and metformin, combination group of electroacupuncture and Compound C. The control group received a standard diet alongside intraperitoneal citric acid - sodium citrate solution injections. After a 6-week intervention, the effects of each group on fasting blood glucose, lipids, liver function, morphology, lipid droplet infiltration, and fibrosis were evaluated. Techniques including Western blotting, qPCR, immunohistochemistry, and immunofluorescence were employed to gauge the expression of key molecules in AMPK-associated glycolipid metabolism, insulin signaling, autophagy, and fibrosis pathways. Additionally, transmission electron microscopy facilitated the observation of liver autophagy, lipid droplets, and fibrosis. RESULTS Our studies indicated that hyperglycemia, hyperlipidemia and IR promoted lipid accumulation, pathological and functional damage, and resulting in hepatic steatosis and fibrosis. Meanwhile, EA enhanced the activation of AMPK, which in turn improved glycolipid metabolism and autophagy through promoting the expression of PPARα/CPT1A and AMPK/mTOR pathway, inhibiting the expression of SREBP1c, PGC-1α/PCK2 and TGFβ1/Smad2/3 signaling pathway, ultimately exerting its effect on ameliorating hepatic steatosis and fibrosis in T2DM rats. The above effects of EA were consistent with metformin. The combination of EA and metformin had significant advantages in increasing hepatic AMPK expression, improving liver morphology, lipid droplet infiltration, fibrosis, and reducing serum ALT levels. In addition, the ameliorating effects of EA on the progression of MAFLD in T2DM rats were partly disrupted by Compound C, an inhibitor of AMPK. CONCLUSIONS EA upregulated hepatic AMPK expression, curtailing gluconeogenesis and lipogenesis while boosting fatty acid oxidation and autophagy levels. Consequently, it mitigated blood glucose, lipids, and insulin resistance in T2DM rats, thus impeding liver steatosis and fibrosis progression and retarding MAFLD advancement.
Collapse
Affiliation(s)
- Haoru Duan
- School of Acupuncture - Moxibustion, and Tuina, Beijing University of Chinese Medicine, Beijing, 100029, China
- Department of Acupuncture and Moxibustion, Chaoyang District Traditional Chinese Medicine Hospital, Beijing, 100026, China
| | - Shanshan Song
- School of Acupuncture - Moxibustion, and Tuina, Beijing University of Chinese Medicine, Beijing, 100029, China
- Department of Acupuncture and Moxibustion, China- Japan Friendship Hospital, Beijing, 100029, China
| | - Rui Li
- School of Acupuncture - Moxibustion, and Tuina, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Suqin Hu
- Department of Gastroenterology, Henan Province Hospital of Traditional Chinese Medicine, Henan University of Chinese Medicine, Henan, 450002, China
| | - Shuting Zhuang
- School of Acupuncture - Moxibustion, and Tuina, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Shaoyang Liu
- School of Acupuncture - Moxibustion, and Tuina, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaolu Li
- School of Acupuncture - Moxibustion, and Tuina, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wei Gao
- School of Acupuncture - Moxibustion, and Tuina, Beijing University of Chinese Medicine, Beijing, 100029, China
| |
Collapse
|
6
|
Oswalia J, Singh S, Gautam V, Arya R. Altered autophagic flux in GNE mutant cells of Indian origin: Potential drug target for GNE myopathy. Exp Cell Res 2024; 440:114118. [PMID: 38852763 DOI: 10.1016/j.yexcr.2024.114118] [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: 03/26/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Autophagy phenomenon in the cell maintains proteostasis balance by eliminating damaged organelles and protein aggregates. Imbalance in autophagic flux may cause accumulation of protein aggregates in various neurodegenerative disorders. Regulation of autophagy by either calcium or chaperone play a key role in the removal of protein aggregates from the cell. The neuromuscular rare genetic disorder, GNE Myopathy, is characterized by accumulation of rimmed vacuoles having protein aggregates of β-amyloid and tau that may result from altered autophagic flux. In the present study, the autophagic flux was deciphered in HEK cell-based model for GNE Myopathy harbouring GNE mutations of Indian origin. The refolding activity of HSP70 chaperone was found to be reduced in GNE mutant cells compared to wild type controls. The autophagic markers LC3II/I ratio was altered with increased number of autophagosome formation in GNE mutant cells compared to wild type cells. The cytosolic calcium levels were also increased in GNE mutant cells of Indian origin. Interestingly, treatment of GNE mutant cells with HSP70 activator, BGP-15, restored the expression and refolding activity of HSP70 along with autophagosome formation. Treatment with calcium chelator, BAPTA-AM restored the cytoplasmic calcium levels and autophagosome formation but not LC3II/I ratio significantly. Our study provides insights towards GNE mutation specific response for autophagy regulation and opens up a therapeutic advancement area in calcium signalling and HSP70 function for GNE related Myopathy.
Collapse
Affiliation(s)
- Jyoti Oswalia
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Shagun Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Vaishali Gautam
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Ranjana Arya
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
7
|
Guo X, Kong Y, Kwon TH, Li C, Wang W. Autophagy and regulation of aquaporins in the kidneys. Kidney Res Clin Pract 2023; 42:676-685. [PMID: 37098672 DOI: 10.23876/j.krcp.22.247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/20/2022] [Indexed: 04/27/2023] Open
Abstract
Aquaporins (AQPs) are water channel proteins that facilitate the transport of water molecules across cell membranes. To date, seven AQPs have been found to be expressed in mammal kidneys. The cellular localization and regulation of the transport properties of AQPs in the kidney have been widely investigated. Autophagy is known as a highly conserved lysosomal pathway, which degrades cytoplasmic components. Through basal autophagy, kidney cells maintain their functions and structure. As a part of the adaptive responses of the kidney, autophagy may be altered in response to stress conditions. Recent studies revealed that autophagic degradation of AQP2 in the kidney collecting ducts leads to impaired urine concentration in animal models with polyuria. Therefore, the modulation of autophagy could be a therapeutic approach to treat water balance disorders. However, as autophagy is either protective or deleterious, it is crucial to establish an optimal condition and therapeutic window where autophagy induction or inhibition could yield beneficial effects. Further studies are needed to understand both the regulation of autophagy and the interaction between AQPs and autophagy in the kidneys in renal diseases, including nephrogenic diabetes insipidus.
Collapse
Affiliation(s)
- Xiangdong Guo
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yonglun Kong
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Chunling Li
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Weidong Wang
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
8
|
Sklifasovskaya AP, Blagonravov M, Ryabinina A, Goryachev V, Syatkin S, Chibisov S, Akhmetova K, Prokofiev D, Agostinelli E. The role of heat shock proteins in the pathogenesis of heart failure (Review). Int J Mol Med 2023; 52:106. [PMID: 37772383 PMCID: PMC10558216 DOI: 10.3892/ijmm.2023.5309] [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: 07/11/2023] [Accepted: 09/11/2023] [Indexed: 09/30/2023] Open
Abstract
The influence of heat shock proteins (HSPs) on protein quality control systems in cardiomyocytes is currently under investigation. The effect of HSPs on the regulated cell death of cardiomyocytes (CMCs) is of great importance, since they play a major role in the implementation of compensatory and adaptive mechanisms in the event of cardiac damage. HSPs mediate a number of mechanisms that activate the apoptotic cascade, playing both pro‑ and anti‑apoptotic roles depending on their location in the cell. Another type of cell death, autophagy, can in some cases lead to cell death, while in other situations it acts as a cell survival mechanism. The present review considered the characteristics of the expression of HSPs of different molecular weights in CMCs in myocardial damage caused by heart failure, as well as their role in the realization of certain types of regulated cell death.
Collapse
Affiliation(s)
| | | | - Anna Ryabinina
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | | | - Sergey Syatkin
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Sergey Chibisov
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Karina Akhmetova
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Daniil Prokofiev
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Enzo Agostinelli
- Department of Sensory Organs, Faculty of Medicine and Dentistry, Sapienza University of Rome, University Hospital Policlinico Umberto I, I-00161 Rome, Italy
- International Polyamines Foundation, ETS-ONLUS, I-00159 Rome, Italy
| |
Collapse
|
9
|
Raee P, Tan SC, Najafi S, Zandsalimi F, Low TY, Aghamiri S, Fazeli E, Aghapour M, Mofarahe ZS, Heidari MH, Fathabadi FF, Abdi F, Asouri M, Ahmadi AA, Ghanbarian H. Autophagy, a critical element in the aging male reproductive disorders and prostate cancer: a therapeutic point of view. Reprod Biol Endocrinol 2023; 21:88. [PMID: 37749573 PMCID: PMC10521554 DOI: 10.1186/s12958-023-01134-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/01/2023] [Indexed: 09/27/2023] Open
Abstract
Autophagy is a highly conserved, lysosome-dependent biological mechanism involved in the degradation and recycling of cellular components. There is growing evidence that autophagy is related to male reproductive biology, particularly spermatogenic and endocrinologic processes closely associated with male sexual and reproductive health. In recent decades, problems such as decreasing sperm count, erectile dysfunction, and infertility have worsened. In addition, reproductive health is closely related to overall health and comorbidity in aging men. In this review, we will outline the role of autophagy as a new player in aging male reproductive dysfunction and prostate cancer. We first provide an overview of the mechanisms of autophagy and its role in regulating male reproductive cells. We then focus on the link between autophagy and aging-related diseases. This is followed by a discussion of therapeutic strategies targeting autophagy before we end with limitations of current studies and suggestions for future developments in the field.
Collapse
Affiliation(s)
- Pourya Raee
- Student Research Committee, Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 19395-4719, Iran
| | - Farshid Zandsalimi
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Teck Yew Low
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Shahin Aghamiri
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Fazeli
- Mehr Fertility Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mahyar Aghapour
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany
| | - Zahra Shams Mofarahe
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Heidari
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Fadaei Fathabadi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farid Abdi
- Department of Chemical Engineering, Science and Research branch, Islamic Azad University, Tehran, Iran
| | - Mohsen Asouri
- North Research Center, Pasteur Institute of Iran, Amol, Iran
| | | | - Hossein Ghanbarian
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 19395-4719, Iran.
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Albano GD, Montalbano AM, Gagliardo R, Profita M. Autophagy/Mitophagy in Airway Diseases: Impact of Oxidative Stress on Epithelial Cells. Biomolecules 2023; 13:1217. [PMID: 37627282 PMCID: PMC10452925 DOI: 10.3390/biom13081217] [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: 06/08/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Autophagy is the key process by which the cell degrades parts of itself within the lysosomes. It maintains cell survival and homeostasis by removing molecules (particularly proteins), subcellular organelles, damaged cytoplasmic macromolecules, and by recycling the degradation products. The selective removal or degradation of mitochondria is a particular type of autophagy called mitophagy. Various forms of cellular stress (oxidative stress (OS), hypoxia, pathogen infections) affect autophagy by inducing free radicals and reactive oxygen species (ROS) formation to promote the antioxidant response. Dysfunctional mechanisms of autophagy have been found in different respiratory diseases such as chronic obstructive lung disease (COPD) and asthma, involving epithelial cells. Several existing clinically approved drugs may modulate autophagy to varying extents. However, these drugs are nonspecific and not currently utilized to manipulate autophagy in airway diseases. In this review, we provide an overview of different autophagic pathways with particular attention on the dysfunctional mechanisms of autophagy in the epithelial cells during asthma and COPD. Our aim is to further deepen and disclose the research in this direction to stimulate the develop of new and selective drugs to regulate autophagy for asthma and COPD treatment.
Collapse
Affiliation(s)
- Giusy Daniela Albano
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), Section of Palermo, Via Ugo La Malfa 153, 90146 Palermo, Italy; (A.M.M.); (R.G.); (M.P.)
| | | | | | | |
Collapse
|
11
|
Liu J, Wang L, He H, Liu Y, Jiang Y, Yang J. The Complex Role of Chaperone-Mediated Autophagy in Cancer Diseases. Biomedicines 2023; 11:2050. [PMID: 37509689 PMCID: PMC10377530 DOI: 10.3390/biomedicines11072050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Chaperone-mediated autophagy (CMA) is a process that rapidly degrades proteins labeled with KFERQ-like motifs within cells via lysosomes to terminate their cellular functioning. Meanwhile, CMA plays an essential role in various biological processes correlated with cell proliferation and apoptosis. Previous studies have shown that CMA was initially found to be procancer in cancer cells, while some theories suggest that it may have an inhibitory effect on the progression of cancer in untransformed cells. Therefore, the complex relationship between CMA and cancer has aroused great interest in the application of CMA activity regulation in cancer therapy. Here, we describe the basic information related to CMA and introduce the physiological functions of CMA, the dual role of CMA in different cancer contexts, and its related research progress. Further study on the mechanism of CMA in tumor development may provide novel insights for tumor therapy targeting CMA. This review aims to summarize and discuss the complex mechanisms of CMA in cancer and related potential strategies for cancer therapy.
Collapse
Affiliation(s)
- Jing Liu
- Department of Basic Medicine, School of Medicine, Hunan Normal University, Changsha 410013, China
| | - Lijuan Wang
- Department of Anesthesiology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Hua He
- Department of Basic Medicine, School of Medicine, Hunan Normal University, Changsha 410013, China
| | - Yueying Liu
- Department of Basic Medicine, School of Medicine, Hunan Normal University, Changsha 410013, China
| | - Yiqun Jiang
- Department of Basic Medicine, School of Medicine, Hunan Normal University, Changsha 410013, China
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha 410013, China
| | - Jinfeng Yang
- Department of Anesthesiology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
| |
Collapse
|
12
|
Bourebaba L, Serwotka-Suszczak A, Pielok A, Sikora M, Mularczyk M, Marycz K. The PTP1B inhibitor MSI-1436 ameliorates liver insulin sensitivity by modulating autophagy, ER stress and systemic inflammation in Equine metabolic syndrome affected horses. Front Endocrinol (Lausanne) 2023; 14:1149610. [PMID: 37020593 PMCID: PMC10067883 DOI: 10.3389/fendo.2023.1149610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 03/08/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND Equine metabolic syndrome (EMS) is a multifactorial pathology gathering insulin resistance, low-grade inflammation and past or chronic laminitis. Among the several molecular mechanisms underlying EMS pathogenesis, increased negative insulin signalling regulation mediated by protein tyrosine phosphatase 1 B (PTP1B) has emerged as a critical axis in the development of liver insulin resistance and general metabolic distress associated to increased ER stress, inflammation and disrupted autophagy. Thus, the use of PTP1B selective inhibitors such as MSI-1436 might be considered as a golden therapeutic tool for the proper management of EMS and associated conditions. Therefore, the present investigation aimed at verifying the clinical efficacy of MSI-1436 systemic administration on liver metabolic balance, insulin sensitivity and inflammatory status in EMS affected horses. Moreover, the impact of MSI-1436 treatment on liver autophagy machinery and associated ER stress in liver tissue has been analysed. METHODS Liver explants isolated from healthy and EMS horses have been treated with MSI-1436 prior to gene and protein expression analysis of main markers mediating ER stress, mitophagy and autophagy. Furthermore, EMS horses have been intravenously treated with a single dose of MSI-1436, and evaluated for their metabolic and inflammatory status. RESULTS Clinical application of MSI-1436 to EMS horses restored proper adiponectin levels and attenuated the typical hyperinsulinemia and hyperglycemia. Moreover, administration of MSI-1436 further reduced the circulating levels of key pro-inflammatory mediators including IL-1β, TNF-α and TGF-β and triggered the Tregs cells activation. At the molecular level, PTP1B inhibition resulted in a noticeable mitigation of liver ER stress, improvement of mitochondrial dynamics and consequently, a regulation of autophagic response. Similarly, short-term ex vivo treatment of EMS liver explants with trodusquemine (MSI-1436) substantially enhanced autophagy by upregulating the levels of HSC70 and Beclin-1 at both mRNA and protein level. Moreover, the PTP1B inhibitor potentiated mitophagy and associated expression of MFN2 and PINK1. Interestingly, inhibition of PTP1B resulted in potent attenuation of ER stress key mediators' expression namely, CHOP, ATF6, HSPA5 and XBP1. CONCLUSION Presented findings shed for the first time promising new insights in the development of an MSI-1436-based therapy for proper equine metabolic syndrome intervention and may additionally find potential translational application to human metabolic syndrome treatment.
Collapse
Affiliation(s)
- Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
- International Institute of Translational Medicine, Wisznia Mała, Poland
| | - Anna Serwotka-Suszczak
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Ariadna Pielok
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Mateusz Sikora
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Malwina Mularczyk
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
- International Institute of Translational Medicine, Wisznia Mała, Poland
| | - Krzysztof Marycz
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- *Correspondence: Krzysztof Marycz,
| |
Collapse
|
13
|
Carinci M, Palumbo L, Pellielo G, Agyapong ED, Morciano G, Patergnani S, Giorgi C, Pinton P, Rimessi A. The Multifaceted Roles of Autophagy in Infectious, Obstructive, and Malignant Airway Diseases. Biomedicines 2022; 10:biomedicines10081944. [PMID: 36009490 PMCID: PMC9405571 DOI: 10.3390/biomedicines10081944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Autophagy is a highly conserved dynamic process by which cells deliver their contents to lysosomes for degradation, thus ensuring cell homeostasis. In response to environmental stress, the induction of autophagy is crucial for cell survival. The dysregulation of this degradative process has been implicated in a wide range of pathologies, including lung diseases, representing a relevant potential target with significant clinical outcomes. During lung disease progression and infections, autophagy may exert both protective and harmful effects on cells. In this review, we will explore the implications of autophagy and its selective forms in several lung infections, such as SARS-CoV-2, Respiratory Syncytial Virus (RSV) and Mycobacterium tuberculosis (Mtb) infections, and different lung diseases such as Cystic Fibrosis (CF), Chronic Obstructive Pulmonary Disease (COPD), and Malignant Mesothelioma (MM).
Collapse
Affiliation(s)
- Marianna Carinci
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Laura Palumbo
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Giulia Pellielo
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Esther Densu Agyapong
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Giampaolo Morciano
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Simone Patergnani
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center of Research for Innovative Therapies in Cystic Fibrosis, University of Ferrara, Via Fossato di Mortara, 70, 44121 Ferrara, Italy
| | - Alessandro Rimessi
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center of Research for Innovative Therapies in Cystic Fibrosis, University of Ferrara, Via Fossato di Mortara, 70, 44121 Ferrara, Italy
- Correspondence:
| |
Collapse
|
14
|
Chaperone-Mediated Autophagy in Neurodegenerative Diseases: Molecular Mechanisms and Pharmacological Opportunities. Cells 2022; 11:cells11142250. [PMID: 35883693 PMCID: PMC9323300 DOI: 10.3390/cells11142250] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 11/23/2022] Open
Abstract
Chaperone-mediated autophagy (CMA) is a protein degradation mechanism through lysosomes. By targeting the KFERQ motif of the substrate, CMA is responsible for the degradation of about 30% of cytosolic proteins, including a series of proteins associated with neurodegenerative diseases (NDs). The fact that decreased activity of CMA is observed in NDs, and ND-associated mutant proteins, including alpha-synuclein and Tau, directly impair CMA activity reveals a possible vicious cycle of CMA impairment and pathogenic protein accumulation in ND development. Given the intrinsic connection between CMA dysfunction and ND, enhancement of CMA has been regarded as a strategy to counteract ND. Indeed, genetic and pharmacological approaches to modulate CMA have been shown to promote the degradation of ND-associated proteins and alleviate ND phenotypes in multiple ND models. This review summarizes the current knowledge on the mechanism of CMA with a focus on its relationship with NDs and discusses the therapeutic potential of CMA modulation for ND.
Collapse
|
15
|
Vitale E, Perveen S, Rossin D, Lo Iacono M, Rastaldo R, Giachino C. Role of Chaperone-Mediated Autophagy in Ageing Biology and Rejuvenation of Stem Cells. Front Cell Dev Biol 2022; 10:912470. [PMID: 35837330 PMCID: PMC9273769 DOI: 10.3389/fcell.2022.912470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/07/2022] [Indexed: 11/17/2022] Open
Abstract
What lies at the basis of the mechanisms that regulate the maintenance and self-renewal of pluripotent stem cells is still an open question. The control of stemness derives from a fine regulation between transcriptional and metabolic factors. In the last years, an emerging topic has concerned the involvement of Chaperone-Mediated Autophagy (CMA) as a key mechanism in stem cell pluripotency control acting as a bridge between epigenetic, transcriptional and differentiation regulation. This review aims to clarify this new and not yet well-explored horizon discussing the recent studies regarding the CMA impact on embryonic, mesenchymal, and haematopoietic stem cells. The review will discuss how CMA influences embryonic stem cell activity promoting self-renewal or differentiation, its involvement in maintaining haematopoietic stem cell function by increasing their functionality during the normal ageing process and its effects on mesenchymal stem cells, in which modulation of CMA regulates immunosuppressive and differentiation properties. Finally, the importance of these new discoveries and their relevance for regenerative medicine applications, from transplantation to cell rejuvenation, will be addressed.
Collapse
|
16
|
Shahverdi M, Hajiasgharzadeh K, Sorkhabi AD, Jafarlou M, Shojaee M, Jalili Tabrizi N, Alizadeh N, Santarpia M, Brunetti O, Safarpour H, Silvestris N, Baradaran B. The regulatory role of autophagy-related miRNAs in lung cancer drug resistance. Biomed Pharmacother 2022; 148:112735. [DOI: 10.1016/j.biopha.2022.112735] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 12/13/2022] Open
|