51
|
Selarka K, Shravage BV. Illuminating intercellular autophagy: A comprehensive review of cell non-autonomous autophagy. Biochem Biophys Res Commun 2024; 716:150024. [PMID: 38701555 DOI: 10.1016/j.bbrc.2024.150024] [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: 03/31/2024] [Accepted: 04/26/2024] [Indexed: 05/05/2024]
Abstract
Macro-autophagy (autophagy hereafter) is an evolutionarily conserved cellular process that has long been recognized as an intracellular mechanism for maintaining cellular homeostasis. It involves the formation of a membraned structure called the autophagosome, which carries cargo that includes toxic protein aggregates and dysfunctional organelles to the lysosome for degradation and recycling. Autophagy is primarily considered and studied as a cell-autonomous mechanism. However, recent studies have illuminated an underappreciated facet of autophagy, i.e., non-autonomously regulated autophagy. Non-autonomously regulated autophagy involves the degradation of autophagic components, including organelles, cargo, and signaling molecules, and is induced in neighboring cells by signals from primary adjacent or distant cells/tissues/organs. This review provides insight into the complex molecular mechanisms governing non-autonomously regulated autophagy, highlighting the dynamic interplay between cells within tissue/organ or distinct cell types in different tissues/organs. Emphasis is placed on modes of intercellular communication that include secreted molecules, including microRNAs, and their regulatory roles in orchestrating this phenomenon. Furthermore, we explore the multidimensional roles of non-autonomously regulated autophagy in various physiological contexts, spanning tissue development and aging, as well as its importance in diverse pathological conditions, including cancer and neurodegeneration. By studying the complexities of non-autonomously regulated autophagy, we hope to gain insights into the sophisticated intercellular dynamics within multicellular organisms, including mammals. These studies will uncover novel avenues for therapeutic intervention to modulate intercellular autophagic pathways in altered human physiology.
Collapse
Affiliation(s)
- Karan Selarka
- Developmental Biology Group, MACS-Agharkar Research Institute, Pune, India; Department of Biotechnology, Savitribai Phule Pune University, Pune, India
| | - Bhupendra V Shravage
- Developmental Biology Group, MACS-Agharkar Research Institute, Pune, India; Department of Biotechnology, Savitribai Phule Pune University, Pune, India; Department of Zoology, Savitribai Phule Pune University, Pune, India.
| |
Collapse
|
52
|
Nakamura Y, Sawai T, Kakiuchi K, Arawaka S. Neuronal activity promotes secretory autophagy for the extracellular release of α-synuclein. J Biol Chem 2024; 300:107419. [PMID: 38815862 PMCID: PMC11253543 DOI: 10.1016/j.jbc.2024.107419] [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: 11/07/2023] [Revised: 05/15/2024] [Accepted: 05/19/2024] [Indexed: 06/01/2024] Open
Abstract
Extracellular secretion is an essential mechanism for α-synuclein (α-syn) proteostasis. Although it has been reported that neuronal activity affects α-syn secretion, the underlying mechanisms remain unclear. Here, we investigated the autophagic processes that regulate the physiological release of α-syn in mouse primary cortical neurons and SH-SY5Y cells. Stimulating neuronal activity with glutamate or depolarization with high KCl enhanced α-syn secretion. This glutamate-induced α-syn secretion was blocked by a mixture of NMDA receptor antagonist AP5 and AMPA receptor antagonist NBQX, as well as by cytosolic Ca2+ chelator BAPTA-AM. Additionally, mTOR inhibitor rapamycin increased α-syn and p62/SQSTM1 (p62) secretion, and this effect of rapamycin was reduced in primary cortical neurons deficient in the autophagy regulator beclin 1 (derived from BECN1+/- mice). Glutamate-induced α-syn and p62 secretion was suppressed by the knockdown of ATG5, which is required for autophagosome formation. Glutamate increased LC3-II generation and decreased intracellular p62 levels, and the increase in LC3-II levels was blocked by BAPTA-AM. Moreover, glutamate promoted co-localization of α-syn with LC3-positive puncta, but not with LAMP1-positive structures in the neuronal somas. Glutamate-induced α-syn and p62 secretion were also reduced by the knockdown of RAB8A, which is required for autophagosome fusion with the plasma membrane. Collectively, these findings suggest that stimulating neuronal activity mediates autophagic α-syn secretion in a cytosolic Ca2+-dependent manner, and autophagosomes may participate in autophagic secretion by functioning as α-syn carriers.
Collapse
Affiliation(s)
- Yoshitsugu Nakamura
- Division of Neurology, Department of Internal Medicine IV, Osaka Medical and Pharmaceutical University Faculty of Medicine, Takatsuki, Osaka, Japan
| | - Taiki Sawai
- Division of Neurology, Department of Internal Medicine IV, Osaka Medical and Pharmaceutical University Faculty of Medicine, Takatsuki, Osaka, Japan
| | - Kensuke Kakiuchi
- Division of Neurology, Department of Internal Medicine IV, Osaka Medical and Pharmaceutical University Faculty of Medicine, Takatsuki, Osaka, Japan
| | - Shigeki Arawaka
- Division of Neurology, Department of Internal Medicine IV, Osaka Medical and Pharmaceutical University Faculty of Medicine, Takatsuki, Osaka, Japan.
| |
Collapse
|
53
|
Chai Y, Chen F, Li H, Sun X, Yang P, Xi Y. Mechanism of salidroside regulating autophagy based on network pharmacology and molecular docking. Anticancer Drugs 2024; 35:525-534. [PMID: 38502854 DOI: 10.1097/cad.0000000000001601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Salidroside is a natural product of phenols with a wide range of pharmacological functions, but whether it plays a role in regulating autophagy is unclear. We systematically investigated the regulatory effect and molecular mechanism of salidroside on autophagy through network pharmacology, which provided a theoretical basis for subsequent experimental research. First, the target genes of salidroside were obtained using the Chinese Medicine System Pharmacology Database and Analysis Platform, and the target genes were converted into standardized gene names using the Uniprot website. At the same time, autophagy-related genes were collected from GeneCards, and preliminary handling of data to obtain intersecting genes. Then, the String website was used to construct a protein-protein interaction network, and to perform the Gene Ontology functional annotation and Kyoto Encyclopedia of Genes and Genomes pathway analysis. To observe the specific molecular mechanism by which salidroside regulates autophagy, we constructed a drug component-target genes-autophagy network. Finally, we performed molecular docking to verify the possible binding conformation between salidroside and the candidate target. By searching the database and analyzing the data, we found that 113 target genes in salidroside interact with autophagy. Salidroside regulate autophagy in relation to a number of important oncogenes and signaling pathways. Molecular docking confirmed that salidroside has high affinity with mTOR, SIRT1, and AKT1. Through network pharmacology combined with molecular docking-validated research methods, we revealed the underlying mechanism of salidroside regulation of autophagy. This study not only provides new systematic insights into the underlying mechanism of salidroside in autophagy, but also provides new ideas for network approaches for autophagy-related research.
Collapse
Affiliation(s)
- Yihong Chai
- The First Clinical Medical College of Lanzhou University
| | - Feng Chen
- The First Clinical Medical College of Lanzhou University
| | - Hongxing Li
- The First Clinical Medical College of Lanzhou University
- Department of Obstetrics and Gynaecology
| | - Xiaohong Sun
- The First Clinical Medical College of Lanzhou University
| | - Panpan Yang
- The First Clinical Medical College of Lanzhou University
- Department of Obstetrics and Gynaecology
| | - YaMing Xi
- The First Clinical Medical College of Lanzhou University
- Department of Hematology, First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
54
|
Amin A, Perera ND, Tomas D, Cuic B, Radwan M, Hatters DM, Turner BJ, Shabanpoor F. Systemic administration of a novel Beclin 1-derived peptide significantly upregulates autophagy in the spinal motor neurons of autophagy reporter mice. Int J Pharm 2024; 659:124198. [PMID: 38816263 DOI: 10.1016/j.ijpharm.2024.124198] [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: 02/27/2024] [Revised: 04/09/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024]
Abstract
Autophagy, an intracellular degradation system, plays a vital role in protecting cells by clearing damaged organelles, pathogens, and protein aggregates. Autophagy upregulation through pharmacological interventions has gained significant attention as a potential therapeutic avenue for proteinopathies. Here, we report the development of an autophagy-inducing peptide (BCN4) derived from the Beclin 1 protein, the master regulator of autophagy. To deliver the BCN4 into cells and the central nervous system (CNS), it was conjugated to our previously developed cell and blood-brain barrier-penetrating peptide (CPP). CPP-BCN4 significantly upregulated autophagy and reduced protein aggregates in motor neuron (MN)-like cells. Moreover, its systemic administration in a reporter mouse model of autophagy resulted in a significant increase in autophagy activity in the spinal MNs. Therefore, this novel autophagy-inducing peptide with a demonstrated ability to upregulate autophagy in the CNS has significant potential for the treatment of various neurodegenerative diseases with protein aggregates as a characteristic feature.
Collapse
Affiliation(s)
- Azin Amin
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, VIC, Australia
| | - Nirma D Perera
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, VIC, Australia
| | - Doris Tomas
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, VIC, Australia
| | - Brittany Cuic
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, VIC, Australia
| | - Mona Radwan
- Walter and Eliza Hall Institute of Medical Research, University of Melbourne, Parkville 3010, VIC, Australia
| | - Danny M Hatters
- Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville 3010, VIC, Australia
| | - Bradley J Turner
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, VIC, Australia
| | - Fazel Shabanpoor
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, VIC, Australia; School of Chemistry, University of Melbourne, VIC 3010, Australia.
| |
Collapse
|
55
|
Amissah HA, Combs SE, Shevtsov M. Tumor Dormancy and Reactivation: The Role of Heat Shock Proteins. Cells 2024; 13:1087. [PMID: 38994941 PMCID: PMC11240553 DOI: 10.3390/cells13131087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
Tumors are a heterogeneous group of cell masses originating in various organs or tissues. The cellular composition of the tumor cell mass interacts in an intricate manner, influenced by humoral, genetic, molecular, and tumor microenvironment cues that dictate tumor growth or suppression. As a result, tumors undergo a period of a dormant state before their clinically discernible stage, which surpasses the clinical dormancy threshold. Moreover, as a genetically imprinted strategy, early-seeder cells, a distinct population of tumor cells, break off to dock nearby or extravasate into blood vessels to secondary tissues, where they form disseminated solitary dormant tumor cells with reversible capacity. Among the various mechanisms underlying the dormant tumor mass and dormant tumor cell formation, heat shock proteins (HSPs) might play one of the most important roles in how the dormancy program plays out. It is known that numerous aberrant cellular processes, such as malignant transformation, cancer cell stemness, tumor invasion, metastasis, angiogenesis, and signaling pathway maintenance, are influenced by the HSPs. An accumulating body of knowledge suggests that HSPs may be involved in the angiogenic switch, immune editing, and extracellular matrix (ECM) remodeling cascades, crucial genetically imprinted strategies important to the tumor dormancy initiation and dormancy maintenance program. In this review, we highlight the biological events that orchestrate the dormancy state and the body of work that has been conducted on the dynamics of HSPs in a tumor mass, as well as tumor cell dormancy and reactivation. Additionally, we propose a conceptual framework that could possibly underlie dormant tumor reactivation in metastatic relapse.
Collapse
Affiliation(s)
- Haneef Ahmed Amissah
- Institute of Life Sciences and Biomedicine, Department of Medical Biology and Medical Biology, FEFU Campus, Far Eastern Federal University, 690922 Vladivostok, Russia;
- Diagnostics Laboratory Department, Trauma and Specialist Hospital, CE-122-2486, Central Region, Winneba P.O. Box 326, Ghana
| | - Stephanie E. Combs
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany;
| | - Maxim Shevtsov
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany;
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia
| |
Collapse
|
56
|
Ayub A, Hasan MK, Mahmud Z, Hossain MS, Kabir Y. Dissecting the multifaceted roles of autophagy in cancer initiation, growth, and metastasis: from molecular mechanisms to therapeutic applications. Med Oncol 2024; 41:183. [PMID: 38902544 DOI: 10.1007/s12032-024-02417-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/28/2024] [Indexed: 06/22/2024]
Abstract
Autophagy is a cytoplasmic defense mechanism that cells use to break and reprocess their intracellular components. This utilization of autophagy is regarded as a savior in nutrient-deficient and other stressful conditions. Hence, autophagy keeps contact with and responds to miscellaneous cellular tensions and diverse pathways of signal transductions, such as growth signaling and cellular death. Importantly, autophagy is regarded as an effective tumor suppressor because regular autophagic breakdown is essential for cellular maintenance and minimizing cellular damage. However, paradoxically, autophagy has also been observed to promote the events of malignancies. This review discussed the dual role of autophagy in cancer, emphasizing its influence on tumor survival and progression. Possessing such a dual contribution to the malignant establishment, the prevention of autophagy can potentially advocate for the advancement of malignant transformation. In contrast, for the context of the instituted tumor, the agents of preventing autophagy potently inhibit the advancement of the tumor. Key regulators, including calpain 1, mTORC1, and AMPK, modulate autophagy in response to nutritional conditions and stress. Oncogenic mutations like RAS and B-RAF underscore autophagy's pivotal role in cancer development. The review also delves into autophagy's context-dependent roles in tumorigenesis, metastasis, and the tumor microenvironment (TME). It also discusses the therapeutic effectiveness of autophagy for several cancers. The recent implication of autophagy in the control of both innate and antibody-mediated immune systems made it a center of attention to evaluating its role concerning tumor antigens and treatments of cancer.
Collapse
Affiliation(s)
- Afia Ayub
- Department of Biochemistry and Molecular Biology, Tejgaon College, National University, Gazipur, 1704, Bangladesh
| | - Md Kamrul Hasan
- Department of Biochemistry and Molecular Biology, Tejgaon College, National University, Gazipur, 1704, Bangladesh.
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main St. W., Hamilton, L8S 4K1, Canada.
- Department of Public Health, North South University, Dhaka, Bangladesh.
| | - Zimam Mahmud
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| | - Md Sabbir Hossain
- Department of Biochemistry and Molecular Biology, Tejgaon College, National University, Gazipur, 1704, Bangladesh
| | - Yearul Kabir
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| |
Collapse
|
57
|
Zhao G, Wang Y, Fan Z, Xiong J, Ertas YN, Ashammakhi N, Wang J, Ma T. Nanomaterials in crossroad of autophagy control in human cancers: Amplification of cell death mechanisms. Cancer Lett 2024; 591:216860. [PMID: 38583650 DOI: 10.1016/j.canlet.2024.216860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/24/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
Cancer is the result of genetic abnormalities that cause normal cells to grow into neoplastic cells. Cancer is characterized by several distinct features, such as uncontrolled cell growth, extensive spreading to other parts of the body, and the ability to resist treatment. The scientists have stressed the development of nanostructures as novel therapeutic options in suppressing cancer, in response to the emergence of resistance to standard medicines. One of the specific mechanisms with dysregulation during cancer is autophagy. Nanomaterials have the ability to specifically carry medications and genes, and they can also enhance the responsiveness of tumor cells to standard therapy while promoting drug sensitivity. The primary mechanism in this process relies on autophagosomes and their fusion with lysosomes to break down the components of the cytoplasm. While autophagy was initially described as a form of cellular demise, it has been demonstrated to play a crucial role in controlling metastasis, proliferation, and treatment resistance in human malignancies. The pharmacokinetic profile of autophagy modulators is poor, despite their development for use in cancer therapy. Consequently, nanoparticles have been developed for the purpose of delivering medications and autophagy modulators selectively and specifically to the cancer process. Furthermore, several categories of nanoparticles have demonstrated the ability to regulate autophagy, which plays a crucial role in defining the biological characteristics and response to therapy of tumor cells.
Collapse
Affiliation(s)
- Gang Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yutao Wang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Dongcheng, Beijing, 100000, China
| | - Zhongru Fan
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Jian Xiong
- Department of Obstetrics and Gynaecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yavuz Nuri Ertas
- ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, 38039, Türkiye; Department of Biomedical Engineering, Erciyes University, Kayseri, 39039, Türkiye.
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ), Department of Biomedical Engineering, College of Engineering and Human Medicine, Michigan State University, East Lansing, MI, 48824, USA.
| | - Jianfeng Wang
- Department of Urology, First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Ting Ma
- Department of Hepatobiliary and Pancreatic Surgery, First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
58
|
Liu Y, Wang Y, Zhang J, Peng Q, Wang X, Xiao X, Shi K. Nanotherapeutics targeting autophagy regulation for improved cancer therapy. Acta Pharm Sin B 2024; 14:2447-2474. [PMID: 38828133 PMCID: PMC11143539 DOI: 10.1016/j.apsb.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/29/2023] [Accepted: 01/29/2024] [Indexed: 06/05/2024] Open
Abstract
The clinical efficacy of current cancer therapies falls short, and there is a pressing demand to integrate new targets with conventional therapies. Autophagy, a highly conserved self-degradation process, has received considerable attention as an emerging therapeutic target for cancer. With the rapid development of nanomedicine, nanomaterials have been widely utilized in cancer therapy due to their unrivaled delivery performance. Hence, considering the potential benefits of integrating autophagy and nanotechnology in cancer therapy, we outline the latest advances in autophagy-based nanotherapeutics. Based on a brief background related to autophagy and nanotherapeutics and their impact on tumor progression, the feasibility of autophagy-based nanotherapeutics for cancer treatment is demonstrated. Further, emerging nanotherapeutics developed to modulate autophagy are reviewed from the perspective of cell signaling pathways, including modulation of the mammalian target of rapamycin (mTOR) pathway, autophagy-related (ATG) and its complex expression, reactive oxygen species (ROS) and mitophagy, interference with autophagosome-lysosome fusion, and inhibition of hypoxia-mediated autophagy. In addition, combination therapies in which nano-autophagy modulation is combined with chemotherapy, phototherapy, and immunotherapy are also described. Finally, the prospects and challenges of autophagy-based nanotherapeutics for efficient cancer treatment are envisioned.
Collapse
Affiliation(s)
- Yunmeng Liu
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Yaxin Wang
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Jincheng Zhang
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Qikai Peng
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Xingdong Wang
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Xiyue Xiao
- College of Pharmacy, Nankai University, Tianjin 300350, China
| | - Kai Shi
- College of Pharmacy, Nankai University, Tianjin 300350, China
| |
Collapse
|
59
|
Yu Q, Ding J, Li S, Li Y. Autophagy in cancer immunotherapy: Perspective on immune evasion and cell death interactions. Cancer Lett 2024; 590:216856. [PMID: 38583651 DOI: 10.1016/j.canlet.2024.216856] [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: 03/04/2024] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
Both the innate and adaptive immune systems work together to produce immunity. Cancer immunotherapy is a novel approach to tumor suppression that has arisen in response to the ineffectiveness of traditional treatments like radiation and chemotherapy. On the other hand, immune evasion can diminish immunotherapy's efficacy. There has been a lot of focus in recent years on autophagy and other underlying mechanisms that impact the possibility of cancer immunotherapy. The primary feature of autophagy is the synthesis of autophagosomes, which engulf cytoplasmic components and destroy them by lysosomal degradation. The planned cell death mechanism known as autophagy can have opposite effects on carcinogenesis, either increasing or decreasing it. It is autophagy's job to maintain the balance and proper functioning of immune cells like B cells, T cells, and others. In addition, autophagy controls whether macrophages adopt the immunomodulatory M1 or M2 phenotype. The ability of autophagy to control the innate and adaptive immune systems is noteworthy. Interleukins and chemokines are immunological checkpoint chemicals that autophagy regulates. Reducing antigen presentation to induce immunological tolerance is another mechanism by which autophagy promotes cancer survival. Therefore, targeting autophagy is of importance for enhancing potential of cancer immunotherapy.
Collapse
Affiliation(s)
- Qiang Yu
- Department of Digestive Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Jiajun Ding
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Shisen Li
- Department of Digestive Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Yunlong Li
- Department of Digestive Surgery, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
60
|
Fu J, Lin J, Dai Z, Lin B, Zhang J. Hypoxia-associated autophagy flux dysregulation in human cancers. Cancer Lett 2024; 590:216823. [PMID: 38521197 DOI: 10.1016/j.canlet.2024.216823] [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: 02/16/2024] [Revised: 03/09/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
A general feature of cancer is hypoxia, determined as low oxygen levels. Low oxygen levels may cause cells to alter in ways that contribute to tumor growth and resistance to treatment. Hypoxia leads to variations in cancer cell metabolism, angiogenesis and metastasis. Furthermore, a hypoxic tumor microenvironment might induce immunosuppression. Moreover, hypoxia has the potential to impact cellular processes, such as autophagy. Autophagy refers to the catabolic process by which damaged organelles and toxic macromolecules are broken down. The abnormal activation of autophagy has been extensively recorded in human tumors and it serves as a regulator of cell growth, spread to other parts of the body, and resistance to treatment. There is a correlation between hypoxia and autophagy in human malignancies. Hypoxia can regulate the activity of AMPK, mTOR, Beclin-1, and ATGs to govern autophagy in human malignancies. Furthermore, HIF-1α, serving as an indicator of low oxygen levels, controls the process of autophagy. Hypoxia-induced autophagy has a crucial role in regulating the growth, spread, and resistance to treatment in human malignancies. Hypoxia-induced regulation of autophagy can impact other mechanisms of cell death, such as apoptosis. Chemoresistance and radioresistance have become significant challenges in recent years. Hypoxia-mediated autophagy plays a crucial role in determining the response to these therapeutic treatments.
Collapse
Affiliation(s)
- Jiding Fu
- Department of Intensive Care Unit, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Jie Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Zili Dai
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Baisheng Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China
| | - Jian Zhang
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, 510095, China.
| |
Collapse
|
61
|
Song Y, Na H, Lee SE, Kim YM, Moon J, Nam TW, Ji Y, Jin Y, Park JH, Cho SC, Lee J, Hwang D, Ha SJ, Park HW, Kim JB, Lee HW. Dysfunctional adipocytes promote tumor progression through YAP/TAZ-dependent cancer-associated adipocyte transformation. Nat Commun 2024; 15:4052. [PMID: 38744820 PMCID: PMC11094189 DOI: 10.1038/s41467-024-48179-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
Obesity has emerged as a prominent risk factor for the development of malignant tumors. However, the existing literature on the role of adipocytes in the tumor microenvironment (TME) to elucidate the correlation between obesity and cancer remains insufficient. Here, we aim to investigate the formation of cancer-associated adipocytes (CAAs) and their contribution to tumor growth using mouse models harboring dysfunctional adipocytes. Specifically, we employ adipocyte-specific BECN1 KO (BaKO) mice, which exhibit lipodystrophy due to dysfunctional adipocytes. Our results reveal the activation of YAP/TAZ signaling in both CAAs and BECN1-deficient adipocytes, inducing adipocyte dedifferentiation and formation of a malignant TME. The additional deletion of YAP/TAZ from BaKO mice significantly restores the lipodystrophy and inflammatory phenotypes, leading to tumor regression. Furthermore, mice fed a high-fat diet (HFD) exhibit decreased BECN1 and increased YAP/TAZ expression in their adipose tissues. Treatment with the YAP/TAZ inhibitor, verteporfin, suppresses tumor progression in BaKO and HFD-fed mice, highlighting its efficacy against mice with metabolic dysregulation. Overall, our findings provide insights into the key mediators of CAA and their significance in developing a TME, thereby suggesting a viable approach targeting adipocyte homeostasis to suppress cancer growth.
Collapse
Affiliation(s)
- Yaechan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Heeju Na
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung Eon Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - You Min Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jihyun Moon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Tae Wook Nam
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yul Ji
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Young Jin
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jae Hyung Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seok Chan Cho
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jaehoon Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Gemcro, Inc, Seoul, 03722, Republic of Korea
| | - Daehee Hwang
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jae Bum Kim
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
- Gemcro, Inc, Seoul, 03722, Republic of Korea.
| |
Collapse
|
62
|
Rahmati S, Moeinafshar A, Rezaei N. The multifaceted role of extracellular vesicles (EVs) in colorectal cancer: metastasis, immune suppression, therapy resistance, and autophagy crosstalk. J Transl Med 2024; 22:452. [PMID: 38741166 PMCID: PMC11092134 DOI: 10.1186/s12967-024-05267-8] [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: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer structures released by all cells and widely distributed in all biological fluids. EVs are implicated in diverse physiopathological processes by orchestrating cell-cell communication. Colorectal cancer (CRC) is one of the most common cancers worldwide, with metastasis being the leading cause of mortality in CRC patients. EVs contribute significantly to the advancement and spread of CRC by transferring their cargo, which includes lipids, proteins, RNAs, and DNAs, to neighboring or distant cells. Besides, they can serve as non-invasive diagnostic and prognostic biomarkers for early detection of CRC or be harnessed as effective carriers for delivering therapeutic agents. Autophagy is an essential cellular process that serves to remove damaged proteins and organelles by lysosomal degradation to maintain cellular homeostasis. Autophagy and EV release are coordinately activated in tumor cells and share common factors and regulatory mechanisms. Although the significance of autophagy and EVs in cancer is well established, the exact mechanism of their interplay in tumor development is obscure. This review focuses on examining the specific functions of EVs in various aspects of CRC, including progression, metastasis, immune regulation, and therapy resistance. Further, we overview emerging discoveries relevant to autophagy and EVs crosstalk in CRC.
Collapse
Affiliation(s)
- Soheil Rahmati
- Student Research Committee, Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Aysan Moeinafshar
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
63
|
Ding Y, Huang X, Ji T, Qi C, Gao X, Wei R. The emerging roles of miRNA-mediated autophagy in ovarian cancer. Cell Death Dis 2024; 15:314. [PMID: 38702325 PMCID: PMC11068799 DOI: 10.1038/s41419-024-06677-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 05/06/2024]
Abstract
Ovarian cancer is one of the common tumors of the female reproductive organs. It has a high mortality rate, is highly heterogeneous, and early detection and primary prevention are very complex. Autophagy is a cellular process in which cytoplasmic substrates are targeted for degradation in lysosomes through membrane structures called autophagosomes. The periodic elimination of damaged, aged, and redundant cellular molecules or organelles through the sequential translation between amino acids and proteins by two biological processes, protein synthesis, and autophagic protein degradation, helps maintain cellular homeostasis. A growing number of studies have found that autophagy plays a key regulatory role in ovarian cancer. Interestingly, microRNAs regulate gene expression at the posttranscriptional level and thus can regulate the development and progression of ovarian cancer through the regulation of autophagy in ovarian cancer. Certain miRNAs have recently emerged as important regulators of autophagy-related gene expression in cancer cells. Moreover, miRNA analysis studies have now identified a sea of aberrantly expressed miRNAs in ovarian cancer tissues that can affect autophagy in ovarian cancer cells. In addition, miRNAs in plasma and stromal cells in tumor patients can affect the expression of autophagy-related genes and can be used as biomarkers of ovarian cancer progression. This review focuses on the potential significance of miRNA-regulated autophagy in the diagnosis and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yamin Ding
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, China
| | - Xuan Huang
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, China
| | - Tuo Ji
- Institute of Clinical Oncology, The Second People's Hospital of Lianyungang City (Cancer Hospital of Lianyungang), Lianyungang, China
| | - Cong Qi
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, China
| | - Xuzhu Gao
- Institute of Clinical Oncology, The Second People's Hospital of Lianyungang City (Cancer Hospital of Lianyungang), Lianyungang, China.
| | - Rongbin Wei
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, China.
| |
Collapse
|
64
|
He T, Zou J, Sun K, Yang J. Global research status and frontiers on autophagy in hepatocellular carcinoma: a comprehensive bibliometric and visualized analysis. Int J Surg 2024; 110:2788-2802. [PMID: 38376850 PMCID: PMC11093451 DOI: 10.1097/js9.0000000000001202] [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: 11/08/2023] [Accepted: 02/04/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND An extensive body of research has explored the role of autophagy in hepatocellular carcinoma (HCC), revealing its critical involvement in the disease's pathogenesis, progression, and therapeutic targeting. However, there is a discernible deficit in quantitative, analytical studies concerning autophagy in the context of HCC. Accordingly, this investigation endeavored to meticulously assess the evolution of autophagy research, employing bibliometric citation analysis to offer a comprehensive evaluation of the findings in this field. METHODS The authors conducted a literature search on 2 August 2023, to extract relevant publications spanning from 2013 to 2022, indexed in the Science Citation Index-Expanded (SCIE) of the Web of Science Core Collection (WOSCC). Subsequently, the authors performed a bibliometric assessment of the compiled documents using visualization tools such as CiteSpace and VOSviewer. RESULTS The search yielded 734 publications penned by 4699 authors, encompassing contributions from 41 countries and 909 institutions, disseminated across 272 journals, and comprising 26 295 co-cited references from 2667 journals. Notably, China led in publication volume with 264 articles (amounting to 35.9%) and exhibited the most robust collaboration with the United States. The mechanisms underlying autophagy's influence on the emergence and advancement of HCC, as well as the implicated proteins and genes, have garnered significant attention. In recent years, investigations of targeting autophagy and the resistance to sorafenib have surfaced as pivotal themes and emerging frontiers in this domain. CONCLUSIONS This study rigorously collated and distilled the prevailing research narratives and novel insights on autophagy in HCC. The resultant synthesis provides a substantive foundation for medical professionals and researchers, as well as pivotal implications for future investigative endeavors in this arena.
Collapse
Affiliation(s)
- Tao He
- Department of Hepatobiliary Surgery
| | - Jieyu Zou
- Department of Oncology, Chengdu Second People’s Hospital, Chengdu, Sichuan, People’s Republic of China
| | - Ke Sun
- Department of Hepatobiliary Surgery
| | | |
Collapse
|
65
|
Wang G, Jiang X, Torabian P, Yang Z. Investigating autophagy and intricate cellular mechanisms in hepatocellular carcinoma: Emphasis on cell death mechanism crosstalk. Cancer Lett 2024; 588:216744. [PMID: 38431037 DOI: 10.1016/j.canlet.2024.216744] [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: 12/14/2023] [Revised: 02/05/2024] [Accepted: 02/18/2024] [Indexed: 03/05/2024]
Abstract
Hepatocellular carcinoma (HCC) stands as a formidable global health challenge due to its prevalence, marked by high mortality and morbidity rates. This cancer type exhibits a multifaceted etiology, prominently linked to viral infections, non-alcoholic fatty liver disease, and genomic mutations. The inherent heterogeneity of HCC, coupled with its proclivity for developing drug resistance, presents formidable obstacles to effective therapeutic interventions. Autophagy, a fundamental catabolic process, plays a pivotal role in maintaining cellular homeostasis, responding to stressors such as nutrient deprivation. In the context of HCC, tumor cells exploit autophagy, either augmenting or impeding its activity, thereby influencing tumorigenesis. This comprehensive review underscores the dualistic role of autophagy in HCC, acting as both a pro-survival and pro-death mechanism, impacting the trajectory of tumorigenesis. The anti-carcinogenic potential of autophagy is evident in its ability to enhance apoptosis and ferroptosis in HCC cells. Pertinently, dysregulated autophagy fosters drug resistance in the carcinogenic context. Both genomic and epigenetic factors can regulate autophagy in HCC progression. Recognizing the paramount importance of autophagy in HCC progression, this review introduces pharmacological compounds capable of modulating autophagy-either inducing or inhibiting it, as promising avenues in HCC therapy.
Collapse
Affiliation(s)
- Gang Wang
- Department of Interventional, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, PR China
| | - Xiaodi Jiang
- Department of Infectious Disease, Shengjing Hospital of China Medical University, Shenyang, 110020, PR China
| | - Pedram Torabian
- Arnie Charbonneau Cancer Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4Z6, Canada; Department of Medical Sciences, University of Calgary, Calgary, AB, T2N 4Z6, Canada.
| | - Zhi Yang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, PR China.
| |
Collapse
|
66
|
Saddouk FZ, Kuzemczak A, Saito J, Greif DM. Endothelial HIFα/PDGF-B to smooth muscle Beclin1 signaling sustains pathological muscularization in pulmonary hypertension. JCI Insight 2024; 9:e162449. [PMID: 38652543 PMCID: PMC11141934 DOI: 10.1172/jci.insight.162449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
Mechanisms underlying maintenance of pathological vascular hypermuscularization are poorly delineated. Herein, we investigated retention of smooth muscle cells (SMCs) coating normally unmuscularized distal pulmonary arterioles in pulmonary hypertension (PH) mediated by chronic hypoxia with or without Sugen 5416, and reversal of this pathology. With hypoxia in mice or culture, lung endothelial cells (ECs) upregulated hypoxia-inducible factor 1α (HIF1-α) and HIF2-α, which induce platelet-derived growth factor B (PDGF-B), and these factors were reduced to normoxic levels with re-normoxia. Re-normoxia reversed hypoxia-induced pulmonary vascular remodeling, but with EC HIFα overexpression during re-normoxia, pathological changes persisted. Conversely, after establishment of distal muscularization and PH, EC-specific deletion of Hif1a, Hif2a, or Pdgfb induced reversal. In human idiopathic pulmonary artery hypertension, HIF1-α, HIF2-α, PDGF-B, and autophagy-mediating gene products, including Beclin1, were upregulated in pulmonary artery SMCs and/or lung lysates. Furthermore, in mice, hypoxia-induced EC-derived PDGF-B upregulated Beclin1 in distal arteriole SMCs, and after distal muscularization was established, re-normoxia, EC Pdgfb deletion, or treatment with STI571 (which inhibits PDGF receptors) downregulated SMC Beclin1 and other autophagy products. Finally, SMC-specific Becn1 deletion induced apoptosis, reversing distal muscularization and PH mediated by hypoxia with or without Sugen 5416. Thus, chronic hypoxia induction of the HIFα/PDGF-B axis in ECs is required for non-cell-autonomous Beclin1-mediated survival of pathological distal arteriole SMCs.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Arterioles/metabolism
- Arterioles/pathology
- Autophagy
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Beclin-1/metabolism
- Beclin-1/genetics
- Disease Models, Animal
- Endothelial Cells/metabolism
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/genetics
- Hypoxia/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Indoles
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Proto-Oncogene Proteins c-sis/metabolism
- Proto-Oncogene Proteins c-sis/genetics
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pyrroles
- Signal Transduction
- Vascular Remodeling
Collapse
Affiliation(s)
- Fatima Z. Saddouk
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and
- Department of Genetics, Yale University, New Haven, Connecticut, USA
| | - Andrew Kuzemczak
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and
- Department of Genetics, Yale University, New Haven, Connecticut, USA
| | - Junichi Saito
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and
- Department of Genetics, Yale University, New Haven, Connecticut, USA
| | - Daniel M. Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and
- Department of Genetics, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
67
|
Renna FJ, Gonzalez CD, Vaccaro MI. Decoding the Versatile Landscape of Autophagic Protein VMP1 in Cancer: A Comprehensive Review across Tissue Types and Regulatory Mechanisms. Int J Mol Sci 2024; 25:3758. [PMID: 38612567 PMCID: PMC11011780 DOI: 10.3390/ijms25073758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Autophagy, a catabolic process orchestrating the degradation of proteins and organelles within lysosomes, is pivotal for maintaining cellular homeostasis. However, its dual role in cancer involves preventing malignant transformation while fostering progression and therapy resistance. Vacuole Membrane Protein 1 (VMP1) is an essential autophagic protein whose expression, per se, triggers autophagy, being present in the whole autophagic flux. In pancreatic cancer, VMP1-whose expression is linked to the Kirsten Rat Sarcoma Virus (KRAS) oncogene-significantly contributes to disease promotion, progression, and chemotherapy resistance. This investigation extends to breast cancer, colon cancer, hepatocellular carcinoma, and more, highlighting VMP1's nuanced nature, contingent on specific tissue contexts. The examination of VMP1's interactions with micro-ribonucleic acids (miRNAs), including miR-21, miR-210, and miR-124, enhances our understanding of its regulatory network in cancer. Additionally, this article discusses VMP1 gene fusions, especially with ribosomal protein S6 kinase B1 (RPS6KB1), shedding light on potential implications for tumor malignancy. By deciphering the molecular mechanisms linking VMP1 to cancer progression, this exploration paves the way for innovative therapeutic strategies to disrupt these pathways and potentially improve treatment outcomes.
Collapse
Affiliation(s)
- Felipe J. Renna
- Instituto de Bioquimica y Medicina Molecular Prof Alberto Boveris (IBIMOL), CONICET, Universidad de Buenos Aires, Buenos Aires C1113AAC, Argentina;
| | - Claudio D. Gonzalez
- Instituto de Investigaciones, IUC, Medicina Traslacional, Hospital Universitario CEMIC, Buenos Aires C1431FWN, Argentina;
| | - Maria I. Vaccaro
- Instituto de Bioquimica y Medicina Molecular Prof Alberto Boveris (IBIMOL), CONICET, Universidad de Buenos Aires, Buenos Aires C1113AAC, Argentina;
- Instituto de Investigaciones, IUC, Medicina Traslacional, Hospital Universitario CEMIC, Buenos Aires C1431FWN, Argentina;
| |
Collapse
|
68
|
Tucker SK, Ghosal R, Swartz ME, Zhang S, Eberhart JK. Zebrafish raptor mutation inhibits the activity of mTORC1, inducing craniofacial defects due to autophagy-induced neural crest cell death. Development 2024; 151:dev202216. [PMID: 38512806 PMCID: PMC11006402 DOI: 10.1242/dev.202216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/26/2024] [Indexed: 03/23/2024]
Abstract
The mechanistic target of rapamycin (mTOR) coordinates metabolism and cell growth with environmental inputs. mTOR forms two functional complexes: mTORC1 and mTORC2. Proper development requires both complexes but mTORC1 has unique roles in numerous cellular processes, including cell growth, survival and autophagy. Here, we investigate the function of mTORC1 in craniofacial development. We created a zebrafish raptor mutant via CRISPR/Cas9, to specifically disrupt mTORC1. The entire craniofacial skeleton and eyes were reduced in size in mutants; however, overall body length and developmental timing were not affected. The craniofacial phenotype associates with decreased chondrocyte size and increased neural crest cell death. We found that autophagy is elevated in raptor mutants. Chemical inhibition of autophagy reduced cell death and improved craniofacial phenotypes in raptor mutants. Genetic inhibition of autophagy, via mutation of the autophagy gene atg7, improved facial phenotypes in atg7;raptor double mutants, relative to raptor single mutants. We conclude that finely regulated levels of autophagy, via mTORC1, are crucial for craniofacial development.
Collapse
Affiliation(s)
- Scott K. Tucker
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, TX 78712, USA
| | - Ritika Ghosal
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, TX 78712, USA
| | - Mary E. Swartz
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, TX 78712, USA
| | - Stephanie Zhang
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, TX 78712, USA
| | - Johann K. Eberhart
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, TX 78712, USA
| |
Collapse
|
69
|
Ke L, Lin X, Luo Y, Tao S, Yan C, He Y, Wu Y, Liu N, Qin Y. Autophagy core protein BECN1 is vital for spermatogenesis and male fertility in mice†. Biol Reprod 2024; 110:599-614. [PMID: 37975917 DOI: 10.1093/biolre/ioad160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 10/17/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023] Open
Abstract
Mammalian spermatogenesis is a highly complex multi-step biological process, and autophagy has been demonstrated to be involved in the process of spermatogenesis. Beclin-1/BECN1, a core autophagy factor, plays a critical role in many biological processes and diseases. However, its function in spermatogenesis remains largely unclear. In the present study, germ cell-specific Beclin 1 (Becn1) knockout mice were generated and were conducted to determine the role of Becn1 in spermatogenesis and fertility of mice. Results indicate that Becn1 deficiency leads to reduced sperm motility and quantity, partial failure of spermiation, actin network disruption, excessive residual cytoplasm, acrosome malformation, and aberrant mitochondrial accumulation of sperm, ultimately resulting in reduced fertility in male mice. Furthermore, inhibition of autophagy was observed in the testes of germ cell-specific Becn1 knockout mice, which may contribute to impaired spermiogenesis and reduced fertility. Collectively, our results reveal that Becn1 is essential for fertility and spermiogenesis in mice.
Collapse
Affiliation(s)
- Lu Ke
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xinyi Lin
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yuchuan Luo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Siming Tao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Chang Yan
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yifeilong He
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yingjie Wu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ning Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Yinghe Qin
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
70
|
Nakashima A, Furuta A, Yoshida-Kawaguchi M, Yamada K, Nunomura H, Morita K, Yasuda I, Yoneda S, Yamaki-Ushijima A, Shima T, Tsuda S. Immunological regulation and the role of autophagy in preeclampsia. Am J Reprod Immunol 2024; 91:e13835. [PMID: 38467995 DOI: 10.1111/aji.13835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/17/2024] [Accepted: 02/28/2024] [Indexed: 03/13/2024] Open
Abstract
Autophagy is a bulk degradation system that maintains cellular homeostasis by producing energy and/or recycling excess proteins. During early placentation, extravillous trophoblasts invade the decidua and uterine myometrium, facing maternal immune cells, which participate in the immune suppression of paternal and fetal antigens. Regulatory T cells will likely increase in response to a specific antigen before and during early pregnancy. Insufficient expansion of antigen-specific Treg cells, which possess the same T cell receptor, is associated with the pathophysiology of preeclampsia, suggesting sterile systemic inflammation. Autophagy is involved in reducing inflammation through the degradation of inflammasomes and in the differentiation and function of regulatory T cells. Autophagy dysregulation induces protein aggregation in trophoblasts, resulting in placental dysfunction. In this review, we discuss the role of regulatory T cells in normal pregnancies. In addition, we discuss the association between autophagy and regulatory T cells in the development of preeclampsia based on reports on the role of autophagy in autoimmune diseases.
Collapse
Affiliation(s)
- Akitoshi Nakashima
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Atsushi Furuta
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Mihoko Yoshida-Kawaguchi
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Kiyotaka Yamada
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Haruka Nunomura
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Keiko Morita
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Ippei Yasuda
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Satoshi Yoneda
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Akemi Yamaki-Ushijima
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Tomoko Shima
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Sayaka Tsuda
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
71
|
Chen Y, Tan X, Zhang W, Li Y, Deng X, Zeng J, Huang L, Ma X. Natural products targeting macroautophagy signaling in hepatocellular carcinoma therapy: Recent evidence and perspectives. Phytother Res 2024; 38:1623-1650. [PMID: 38302697 DOI: 10.1002/ptr.8103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/07/2023] [Accepted: 12/16/2023] [Indexed: 02/03/2024]
Abstract
Hepatocellular carcinoma (HCC), presently the second leading cause of global cancer-related mortality, continues to pose significant challenges in the realm of medical oncology, impacting both clinical drug selection and mechanistic research. Recent investigations have unveiled autophagy-related signaling as a promising avenue for HCC treatment. A growing body of research has highlighted the pivotal role of autophagy-modulating natural products in inhibiting HCC progression. In this context, we provide a concise overview of the fundamental autophagy mechanism and delineate the involvement of autophagic signaling pathways in HCC development. Additionally, we review pertinent studies demonstrating how natural products regulate autophagy to mitigate HCC. Our findings indicate that natural products exhibit cytotoxic effects through the induction of excessive autophagy, simultaneously impeding HCC cell proliferation by autophagy inhibition, thereby depriving HCC cells of essential energy. These effects have been associated with various signaling pathways, including PI3K/AKT, MAPK, AMPK, Wnt/β-catenin, Beclin-1, and ferroautophagy. These results underscore the considerable therapeutic potential of natural products in HCC treatment. However, it is important to note that the present study did not establish definitive thresholds for autophagy induction or inhibition by natural products. Further research in this domain is imperative to gain comprehensive insights into the dual role of autophagy, equipping us with a better understanding of this double-edged sword in HCC management.
Collapse
Affiliation(s)
- Yuan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiyue Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenwen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yubing Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinyu Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lihua Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
72
|
Ramakrishnan RK, Srivastava A, Rajan R, Abusnana S, Mussa BM. Investigating the role of an immediate early gene FOS as a potential regulator of autophagic response to hypoglycemia in embryonic hypothalamic neurons. Clin Transl Sci 2024; 17:e13749. [PMID: 38488430 PMCID: PMC10941580 DOI: 10.1111/cts.13749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 03/18/2024] Open
Abstract
Hypoglycemia-associated autonomic failure (HAAF) is a well-established complication of diabetes. Although HAAF has serious outcomes such as recurrent morbidity, coma, and death, the mechanisms of HAAF and its pathological components are largely unknown. Our previous studies have revealed that hypoglycemia is associated with the upregulation of an immediate early gene - FOS. In addition, it is documented that glucose deprivation activates neuronal autophagic activities. Therefore, the present study aimed to identify the role of FOS and one of the core components of the autophagy pathway, Beclin-1 (encoded by the BECN1 gene), in the regulation of autophagic mechanisms in embryonic hypothalamic neurons in response to hypoglycemic conditions. Embryonic Mouse Hypothalamic Cell Line N39 (mHypoE-N39 or N39) was cultured in reduced concentrations of glucose (2000, 900, 500, and 200 mg/L). Gene and protein expression, as well as immunofluorescence studies on autophagy were conducted under different reduced glucose concentrations in N39 hypothalamic neurons with and without FOS and BECN1 gene knockdowns (KD). The outcomes of the present study have demonstrated a significant increase in autophagosome formation and subsequent lysosomal degradation in the hypothalamic neurons in response to reduced glucose concentrations. This hypoglycemic response appears to be lowered to a similar extent in the FOS KD and BECN1 KD cells, albeit insignificantly from the negative control, is indicative of the involvement of FOS in the autophagic response of hypothalamic neurons to hypoglycemia. Moreover, the KD cells exhibited a change in morphology and reduced cell viability compared with the control cells. Our findings suggest that reduced FOS expression could potentially be associated with impaired autophagic activities that are dependent on BECN1, which could lead to decreased or blunted hypothalamic activation in response to hypoglycemia, and this, in turn, may contribute to the development of HAAF.
Collapse
Affiliation(s)
- Rakhee K. Ramakrishnan
- Research Institute for Medical and Health Sciences, College of Medicine, University of SharjahSharjahUnited Arab Emirates
| | - Ankita Srivastava
- Research Institute for Medical and Health Sciences, College of Medicine, University of SharjahSharjahUnited Arab Emirates
| | - Reeja Rajan
- Research Institute for Medical and Health Sciences, College of Medicine, University of SharjahSharjahUnited Arab Emirates
| | - Salah Abusnana
- Diabetes and Endocrinology DepartmentUniversity Hospital SharjahSharjahUnited Arab Emirates
- Clinical Science DepartmentCollege of Medicine, University of SharjahSharjahUnited Arab Emirates
| | - Bashair M. Mussa
- Basic Medical Science DepartmentCollege of Medicine, University of SharjahSharjahUnited Arab Emirates
| |
Collapse
|
73
|
Mathur A, Ritu, Chandra P, Das A. Autophagy: a necessary evil in cancer and inflammation. 3 Biotech 2024; 14:87. [PMID: 38390576 PMCID: PMC10879063 DOI: 10.1007/s13205-023-03864-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/21/2023] [Indexed: 02/24/2024] Open
Abstract
Autophagy, a highly regulated cellular process, assumes a dual role in the context of cancer. On the one hand, it functions as a crucial homeostatic pathway, responsible for degrading malfunctioning molecules and organelles, thereby maintaining cellular health. On the other hand, its involvement in cancer development and regression is multifaceted, contingent upon a myriad of factors. This review meticulously examines the intricacies of autophagy, from its molecular machinery orchestrated by Autophagy-Related Genes (ATG) initially discovered in yeast to the various modes of autophagy operative within cells. Beyond its foundational role in cellular maintenance, autophagy reveals context-specific functions in processes like angiogenesis and inflammation. Our analysis delves into how autophagy-related factors directly impact inflammation, underscoring their profound implications for cancer dynamics. Additionally, we extend our inquiry to explore autophagy's associations with cardiovascular conditions, neurodegenerative disorders, and autoimmune diseases, illuminating the broader medical relevance of this process. Furthermore, this review elucidates how autophagy contributes to sustaining hallmark cancer features, including stem cell maintenance, proliferation, angiogenesis, metastasis, and metabolic reprogramming. Autophagy emerges as a pivotal process that necessitates careful consideration in cancer treatment strategies. To this end, we investigate innovative approaches, ranging from enzyme-based therapies to MTOR inhibitors, lysosomal blockers, and nanoparticle-enabled interventions, all aimed at optimizing cancer treatment outcomes by targeting autophagy pathways. In summary, this comprehensive review provides a nuanced perspective on the intricate and context-dependent role of autophagy in cancer biology. Our exploration not only deepens our understanding of this fundamental process but also highlights its potential as a therapeutic target. By unraveling the complex interplay between autophagy and cancer, we pave the way for more precise and effective cancer treatments, promising better outcomes for patients.
Collapse
Affiliation(s)
- Amit Mathur
- Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi, 110042 India
| | - Ritu
- Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi, 110042 India
| | - Prakash Chandra
- Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi, 110042 India
| | - Asmita Das
- Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi, 110042 India
| |
Collapse
|
74
|
Kim R, Kin T, Beck WT. Impact of Complex Apoptotic Signaling Pathways on Cancer Cell Sensitivity to Therapy. Cancers (Basel) 2024; 16:984. [PMID: 38473345 DOI: 10.3390/cancers16050984] [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: 01/26/2024] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Anticancer drugs induce apoptotic and non-apoptotic cell death in various cancer types. The signaling pathways for anticancer drug-induced apoptotic cell death have been shown to differ between drug-sensitive and drug-resistant cells. In atypical multidrug-resistant leukemia cells, the c-Jun/activator protein 1 (AP-1)/p53 signaling pathway leading to apoptotic death is altered. Cancer cells treated with anticancer drugs undergo c-Jun/AP-1-mediated apoptotic death and are involved in c-Jun N-terminal kinase activation and growth arrest- and DNA damage-inducible gene 153 (Gadd153)/CCAAT/enhancer-binding protein homologous protein pathway induction, regardless of the p53 genotype. Gadd153 induction is associated with mitochondrial membrane permeabilization after anticancer drug treatment and involves a coupled endoplasmic reticulum stress response. The induction of apoptosis by anticancer drugs is mediated by the intrinsic pathway (cytochrome c, Cyt c) and subsequent activation of the caspase cascade via proapoptotic genes (e.g., Bax and Bcl-xS) and their interactions. Anticancer drug-induced apoptosis involves caspase-dependent and caspase-independent pathways and occurs via intrinsic and extrinsic pathways. The targeting of antiapoptotic genes such as Bcl-2 enhances anticancer drug efficacy. The modulation of apoptotic signaling by Bcl-xS transduction increases the sensitivity of multidrug resistance-related protein-overexpressing epidermoid carcinoma cells to anticancer drugs. The significance of autophagy in cancer therapy remains to be elucidated. In this review, we summarize current knowledge of cancer cell death-related signaling pathways and their alterations during anticancer drug treatment and discuss potential strategies to enhance treatment efficacy.
Collapse
Affiliation(s)
- Ryungsa Kim
- Department of Breast Surgery, Hiroshima Mark Clinic, 1-4-3F, 2-Chome Ohte-machi, Naka-ku, Hiroshima 730-0051, Japan
| | - Takanori Kin
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - William T Beck
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
75
|
Wang JZ, Paulus P, Niu Y, Zhu L, Morisseau C, Rawling T, Murray M, Hammock BD, Zhou F. The Role of Autophagy in Human Uveal Melanoma and the Development of Potential Disease Biomarkers and Novel Therapeutic Paradigms. Biomedicines 2024; 12:462. [PMID: 38398064 PMCID: PMC10886749 DOI: 10.3390/biomedicines12020462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Autophagy is a form of programmed cell degradation that enables the maintenance of homeostasis in response to extracellular stress stimuli. Autophagy is primarily activated by starvation and mediates the degradation, removal, or recycling of cell cytoplasm, organelles, and intracellular components in eukaryotic cells. Autophagy is also involved in the pathogenesis of human diseases, including several cancers. Human uveal melanoma (UM) is the primary intraocular malignancy in adults and has an extremely poor prognosis; at present there are no effective therapies. Several studies have suggested that autophagy is important in UM. By understanding the mechanisms of activation of autophagy in UM it may be possible to develop biomarkers to provide more definitive disease prognoses and to identify potential drug targets for the development of new therapeutic strategies. This article reviews the current information regarding autophagy in UM that could facilitate biomarker and drug development.
Collapse
Affiliation(s)
- Janney Z. Wang
- Molecular Drug Development Group, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Paus Paulus
- Molecular Drug Development Group, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Yihe Niu
- Molecular Drug Development Group, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Ling Zhu
- Save Sight Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Christophe Morisseau
- Department of Entomology and Nematology, UCD Comprehensive Cancer Center, University of California, Davis, CA 95616, USA (B.D.H.)
| | - Tristan Rawling
- School of Mathematical and Physical Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Michael Murray
- Molecular Drug Development Group, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Bruce D. Hammock
- Department of Entomology and Nematology, UCD Comprehensive Cancer Center, University of California, Davis, CA 95616, USA (B.D.H.)
| | - Fanfan Zhou
- Molecular Drug Development Group, Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
76
|
Kandouz M. Cell Death, by Any Other Name…. Cells 2024; 13:325. [PMID: 38391938 PMCID: PMC10886887 DOI: 10.3390/cells13040325] [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: 12/31/2023] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Studies trying to understand cell death, this ultimate biological process, can be traced back to a century ago. Yet, unlike many other fashionable research interests, research on cell death is more alive than ever. New modes of cell death are discovered in specific contexts, as are new molecular pathways. But what is "cell death", really? This question has not found a definitive answer yet. Nevertheless, part of the answer is irreversibility, whereby cells can no longer recover from stress or injury. Here, we identify the most distinctive features of different modes of cell death, focusing on the executive final stages. In addition to the final stages, these modes can differ in their triggering stimulus, thus referring to the initial stages. Within this framework, we use a few illustrative examples to examine how intercellular communication factors in the demise of cells. First, we discuss the interplay between cell-cell communication and cell death during a few steps in the early development of multicellular organisms. Next, we will discuss this interplay in a fully developed and functional tissue, the gut, which is among the most rapidly renewing tissues in the body and, therefore, makes extensive use of cell death. Furthermore, we will discuss how the balance between cell death and communication is modified during a pathological condition, i.e., colon tumorigenesis, and how it could shed light on resistance to cancer therapy. Finally, we briefly review data on the role of cell-cell communication modes in the propagation of cell death signals and how this has been considered as a potential therapeutic approach. Far from vainly trying to provide a comprehensive review, we launch an invitation to ponder over the significance of cell death diversity and how it provides multiple opportunities for the contribution of various modes of intercellular communication.
Collapse
Affiliation(s)
- Mustapha Kandouz
- Department of Pathology, School of Medicine, Wayne State University, 540 East Canfield Avenue, Detroit, MI 48201, USA;
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
77
|
Bruno A, Milillo C, Anaclerio F, Buccolini C, Dell’Elice A, Angilletta I, Gatta M, Ballerini P, Antonucci I. Perinatal Tissue-Derived Stem Cells: An Emerging Therapeutic Strategy for Challenging Neurodegenerative Diseases. Int J Mol Sci 2024; 25:976. [PMID: 38256050 PMCID: PMC10815412 DOI: 10.3390/ijms25020976] [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: 12/05/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Over the past 20 years, stem cell therapy has been considered a promising option for treating numerous disorders, in particular, neurodegenerative disorders. Stem cells exert neuroprotective and neurodegenerative benefits through different mechanisms, such as the secretion of neurotrophic factors, cell replacement, the activation of endogenous stem cells, and decreased neuroinflammation. Several sources of stem cells have been proposed for transplantation and the restoration of damaged tissue. Over recent decades, intensive research has focused on gestational stem cells considered a novel resource for cell transplantation therapy. The present review provides an update on the recent preclinical/clinical applications of gestational stem cells for the treatment of protein-misfolding diseases including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and amyotrophic lateral sclerosis (ALS). However, further studies should be encouraged to translate this promising therapeutic approach into the clinical setting.
Collapse
Affiliation(s)
- Annalisa Bruno
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Innovative Technologies in Medicine & Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Cristina Milillo
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Psychological, Health and Territorial Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Federico Anaclerio
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Psychological, Health and Territorial Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Carlotta Buccolini
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Psychological, Health and Territorial Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Anastasia Dell’Elice
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Psychological, Health and Territorial Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Ilaria Angilletta
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Psychological, Health and Territorial Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Marco Gatta
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Innovative Technologies in Medicine & Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Patrizia Ballerini
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Innovative Technologies in Medicine & Dentistry, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Ivana Antonucci
- Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.B.); (C.M.); (C.B.); (A.D.); (I.A.)
- Department of Psychological, Health and Territorial Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
78
|
Mishra A, Giuliani G, Longo VD. Nutrition and dietary restrictions in cancer prevention. Biochim Biophys Acta Rev Cancer 2024; 1879:189063. [PMID: 38147966 DOI: 10.1016/j.bbcan.2023.189063] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/15/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023]
Abstract
The composition and pattern of dietary intake have emerged as key factors influencing aging, regeneration, and consequently, healthspan and lifespan. Cancer is one of the major diseases more tightly linked with aging, and age-related mortality. Although the role of nutrition in cancer incidence is generally well established, we are far from a consensus on how diet influences tumour development in different tissues. In this review, we will discuss how diet and dietary restrictions affect cancer risk and the molecular mechanisms potentially responsible for their effects. We will cover calorie restriction, intermittent fasting, prolonged fasting, fasting-mimicking diet, time-restricted eating, ketogenic diet, high protein diet, Mediterranean diet, and the vegan and vegetarian diets.
Collapse
Affiliation(s)
- Amrendra Mishra
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Giacomo Giuliani
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Valter D Longo
- Longevity Institute and Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA; IFOM, FIRC Institute of Molecular Oncology, Via Adamello, 16, 20139 Milano, Italy.
| |
Collapse
|
79
|
Mobinikhaledi M, Faridzadeh A, Farkhondeh T, Pourhanifeh MH, Samarghandian S. The Roles of Autophagy-related miRNAs in Gynecologic Tumors: A Review of Current Knowledge for Possible Targeted Therapy. Curr Mol Med 2024; 24:1269-1281. [PMID: 39300715 DOI: 10.2174/0115665240263059231002093454] [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: 05/18/2023] [Revised: 08/12/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2024]
Abstract
Gynecological cancers are the leading cause of malignancy-related death and disability in the world. These cancers are diagnosed at end stages, and unfortunately, the standard therapeutic strategies available for the treatment of affected women [including chemotherapy, radiotherapy and surgery] are not safe and effective enough. Moreover, the unwanted side-effects lowering the patients' life quality is another problem for these therapies. Therefore, researchers should search for better alternative/complementary treatments. The involvement of autophagy in the pathogenesis of various cancers has been demonstrated. Recently, a novel crosstalk between microRNAs, small non-coding RNAs with important regulatory functions, and autophagy machinery has been highlighted. In this review, we indicate the importance of this interaction for targeted therapy in the treatment of cancers including gynecological cancers, with a focus on underlying mechanisms.
Collapse
Affiliation(s)
- Mahya Mobinikhaledi
- Department of Pediatrics, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tahereh Farkhondeh
- Department of Toxicology and Pharmacology, School of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
80
|
Beilankouhi EAV, Valilo M, Dastmalchi N, Teimourian S, Safaralizadeh R. The Function of Autophagy in the Initiation, and Development of Breast Cancer. Curr Med Chem 2024; 31:2974-2990. [PMID: 37138421 DOI: 10.2174/0929867330666230503145319] [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: 12/09/2020] [Revised: 02/26/2021] [Accepted: 03/15/2021] [Indexed: 05/05/2023]
Abstract
Autophagy is a significant catabolic procedure that increases in stressful conditions. This mechanism is mostly triggered after damage to the organelles, the presence of unnatural proteins, and nutrient recycling in reaction to these stresses. One of the key points in this article is that cleaning and preserving damaged organelles and accumulated molecules through autophagy in normal cells helps prevent cancer. Since dysfunction of autophagy is associated with various diseases, including cancer, it has a dual function in tumor suppression and expansion. It has newly become clear that the regulation of autophagy can be used for the treatment of breast cancer, which has a promising effect of increasing the efficiency of anticancer treatment in a tissue- and cell-type-specific manner by affecting the fundamental molecular mechanisms. Regulation of autophagy and its function in tumorigenesis is a vital part of modern anticancer techniques. This study discusses the current advances related to the mechanisms that describe essential modulators of autophagy involved in the metastasis of cancers and the development of new breast cancer treatments.
Collapse
Affiliation(s)
| | - Mohammad Valilo
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Narges Dastmalchi
- Department of Biology, University College of Nabi Akram, Tabriz, Iran
| | - Shahram Teimourian
- Department of Medical Genetics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
81
|
Pour PM, Nouri Z, Ghasemi D, Sajadimajd S, Farzaei MH. Cytotoxic Impact of Naringenin-Loaded Solid Lipid Nanoparticles on RIN5F Pancreatic β Cells via Autophagy Blockage. RECENT ADVANCES IN DRUG DELIVERY AND FORMULATION 2024; 18:304-314. [PMID: 39356101 DOI: 10.2174/0126673878297658240804192222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 10/03/2024]
Abstract
BACKGROUND Autophagy plays a crucial role in modulating the proliferation of cancer diseases. However, the application of Naringenin (Nar), a compound with potential benefits against these diseases, has been limited due to its poor solubility and bioavailability. OBJECTIVE This study aimed to develop solid lipid nanoparticles (Nar-SLNs) loaded with Nar to enhance their therapeutic impact. METHODS In vitro experiments using Rin-5F cells exposed to Nar and Nar-SLNs were carried out to investigate the protective effects of Nar and its nanoformulation against the pancreatic cancer cell line of Rin-5F. RESULTS Treatment with Nar and Nar-SLN led to an increase in autophagic markers (Akt, LC3, Beclin1, and ATG genes) and a decrease in the level of miR-21. Both Nar and Nar-SLN treatments inhibited cell proliferation and reduced the expression of autophagic markers. Notably, Nar-SLNs exhibited greater efficacy compared to free Nar. CONCLUSION These findings suggest that SLNs effectively enhance the cytotoxic impact of Nar, making Nar-SLNs a promising candidate for suppressing or preventing Rin-5F cell growth.
Collapse
Affiliation(s)
- Pardis Mohammadi Pour
- Department of Pharmacognosy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Nouri
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Dariush Ghasemi
- Kimia Andisheh Teb Medical and Molecular Laboratory Research Co., Tehran, Iran
| | - Soraya Sajadimajd
- Department of Biology, School of Sciences, Razi University, Kermanshah, Iran
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
82
|
Ihlamur M, Akgul B, Zengin Y, Korkut ŞV, Kelleci K, Abamor EŞ. The mTOR Signaling Pathway and mTOR Inhibitors in Cancer: Next-generation Inhibitors and Approaches. Curr Mol Med 2024; 24:478-494. [PMID: 37165594 DOI: 10.2174/1566524023666230509161645] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 05/12/2023]
Abstract
mTOR is a serine/threonine kinase that plays various roles in cell growth, proliferation, and metabolism. mTOR signaling in cancer becomes irregular. Therefore, drugs targeting mTOR have been developed. Although mTOR inhibitors rapamycin and rapamycin rapalogs (everolimus, rapamycin, temsirolimus, deforolimus, etc.) and new generation mTOR inhibitors (Rapalink, Dual PI3K/mTOR inhibitors, etc.) are used in cancer treatments, mTOR resistance mechanisms may inhibit the efficacy of these drugs. Therefore, new inhibition approaches are developed. Although these new inhibition approaches have not been widely investigated in cancer treatment, the use of nanoparticles has been evaluated as a new treatment option in a few types of cancer. This review outlines the functions of mTOR in the cancer process, its resistance mechanisms, and the efficiency of mTOR inhibitors in cancer treatment. Furthermore, it discusses the next-generation mTOR inhibitors and inhibition strategies created using nanoparticles. Since mTOR resistance mechanisms prevent the effects of mTOR inhibitors used in cancer treatments, new inhibition strategies should be developed. Inhibition approaches are created using nanoparticles, and one of them offers a promising treatment option with evidence supporting its effectiveness.
Collapse
Affiliation(s)
- Murat Ihlamur
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Istanbul, Turkey
- Department of Electronics and Automation, Biruni University, Istanbul, Turkey
| | - Busra Akgul
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Istanbul, Turkey
| | - Yağmur Zengin
- Biomedical Engineering Institute, Department of Biomedical Engineering, Bogazici University, Istanbul, Turkey
| | - Şenay Vural Korkut
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Yildiz Technical University, Istanbul, Turkey
| | - Kübra Kelleci
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Istanbul, Turkey
- Department of Medical Services and Techniques, Beykoz University, Istanbul, Turkey
| | - Emrah Şefik Abamor
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Istanbul, Turkey
| |
Collapse
|
83
|
Cai J, Wang R, Chen Y, Zhang C, Fu L, Fan C. LncRNA FIRRE regulated endometrial cancer radiotherapy sensitivity via the miR-199b-5p/SIRT1/BECN1 axis-mediated autophagy. Genomics 2024; 116:110750. [PMID: 38052260 DOI: 10.1016/j.ygeno.2023.110750] [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: 05/10/2023] [Revised: 11/13/2023] [Accepted: 11/27/2023] [Indexed: 12/07/2023]
Abstract
BACKGROUND Endometrial cancer (EC) poses a serious threat to women's health. Radiotherapy has been widely used for EC treatment. However, the mechanism of FIRRE in EC development and radioresistance remains unknown. METHODS MTT and colony formation assays determined cell proliferation. The degree of autophagy was tested by the measurement of autophagy-related genes and immunofluorescence staining of LC3. Molecular interactions were demonstrated via luciferase reporter assay, RIP, and Co-IP. The FIRRE role's was analyzed by in vivo xenograft tumor model. RESULTS FIRRE and SIRT1 were upregulated in EC tumor tissues, whereas miR-199b-5p was reduced. FIRRE knockdown increased EC cell radiotherapy sensitivity by sponging miR-199b-5p and inhibiting autophagy. SIRT1 was targeted and negatively regulated by miR-199b-5p. SIRT1 could otherwise deacetylate BECN1 protein and participate in FIRRE-mediated autophagy. Silencing FIRRE increased sensitivity of EC radiotherapy in vivo. CONCLUSION FIRRE reduced EC cell radiotherapy sensitivity by stimulating autophagy via miR-199b-5p/SIRT1/BECN1 axis.
Collapse
Affiliation(s)
- Junhong Cai
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University/Hainan General Hospital, Haikou 570311, Hainan Province, PR China.
| | - Ru Wang
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University/Hainan General Hospital, Haikou 570311, Hainan Province, PR China
| | - Yaxiong Chen
- Department of Radiotherapy Center, Hainan Affiliated Hospital of Hainan Medical University/Hainan General Hospital, Haikou 570311, Hainan Province, PR China
| | - Chen Zhang
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University/Hainan General Hospital, Haikou 570311, Hainan Province, PR China
| | - Lanyan Fu
- Department of Gynecology, Hainan Affiliated Hospital of Hainan Medical University/Hainan General Hospital, Haikou 570311, Hainan Province, PR China
| | - Cunfu Fan
- Department of Pathology, Hainan Affiliated Hospital of Hainan Medical University/Hainan General Hospital, Haikou 570311, Hainan Province, PR China
| |
Collapse
|
84
|
Pandya K, Singh N. In silico study reveals unconventional interactions between MDC1 of DDR and Beclin-1 of autophagy. Mol Divers 2023; 27:2789-2802. [PMID: 36482226 DOI: 10.1007/s11030-022-10579-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022]
Abstract
DNA damage response (DDR) and autophagy are concerned with maintaining cellular homeostasis and dysregulation of these two pathways lead to pathologic conditions including tumorigenesis. Autophagy is activated as a protective mechanism during DDR which is indicative of their functional cooperativity but the molecular mechanism leading to the convergence of these two pathways during genotoxic stress remains elusive. In this study, through in silico analysis, we have shown an interaction between the Mediator of DNA damage checkpoint 1 (MDC1), an important DDR-associated protein, and Beclin-1, an autophagy inducer. MDC1 is an adaptor or scaffold protein known to regulate DDR, apoptosis, and cell cycle progression. While, Beclin-1 is involved in autophagosome nucleation and exhibits affinity for binding to Fork-head-associated domain (FHA) containing proteins. The FHA domain is commonly conserved in DDR-related proteins including MDC1. Through molecular docking, we have predicted the modeled complex between the MDC1 FHA domain and the Beclin-1 Coiled coil domain (CCD). The docking complex was modeled using ClusPro2.0, based on the crystal structure for the dimerized MDC1 FHA domain and Beclin-1 CCD. The complex stability and binding affinities were assessed using a Ramachandran plot, MD simulation, MM/GBSA, and PRODIGY webserver. Finally, the hot-spot residues at the interface were determined using computational alanine scanning by the DrugScorePPI webserver. Our analysis unveils significant interaction between MDC1 and Beclin-1, involving hydrogen bonds, non-bonded contacts, and salt bridges and indicates MDC1 possibly recruits Beclin-1 to the DSBs, as a consequence of which Beclin-1 is able to modulate DDR.
Collapse
Affiliation(s)
- Kavya Pandya
- Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India
| | - Neeru Singh
- Department of Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar, India.
| |
Collapse
|
85
|
Mehanna S, Arakawa S, Imasaka M, Chen W, Nakanishi Y, Nishiura H, Shimizu S, Ohmuraya M. Beclin1 is essential for the pancreas development. Dev Biol 2023; 504:113-119. [PMID: 37739117 DOI: 10.1016/j.ydbio.2023.09.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
Beclin1 (Becn1) is a multifunctional protein involved in autophagy regulation, membrane trafficking, and tumor suppression. In this study, we examined the roles of Becn1 in the pancreas development by generating mice with conditional deletion of Becn1 in the pancreas using pancreatic transcriptional factor 1a (Ptf1a)-Cre mice (Becn1f/f; Ptf1aCre/+). Surprisingly, loss of Becn1 in the pancreas resulted in severe pancreatic developmental defects, leading to insufficient exocrine and endocrine pancreatic function. Approximately half of Becn1f/f; Ptf1aCre/+ mice died immediately after birth. However, duodenum and neural tissue development were almost normal, indicating that pancreatic insufficiency was the cause of death. These findings demonstrated a novel role for Becn1 in pancreas morphogenesis, differentiation, and growth, and suggested that loss of this factor leaded to pancreatic agenesis at birth.
Collapse
Affiliation(s)
- Sally Mehanna
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Satoko Arakawa
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo, Tokyo, 113-8510, Japan
| | - Mai Imasaka
- Department of Genetics, Hyogo Medical University, Nishinomiya, Hyogo, 663-8501, Japan
| | - Wenting Chen
- Department of Genetics, Hyogo Medical University, Nishinomiya, Hyogo, 663-8501, Japan
| | - Yuto Nakanishi
- Department of Genetics, Hyogo Medical University, Nishinomiya, Hyogo, 663-8501, Japan
| | - Hiroshi Nishiura
- Division of Functional Pathology, Department of Pathology, Hyogo Medical University, Nishinomiya, Hyogo, 663-8501, Japan
| | - Shigeomi Shimizu
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo, Tokyo, 113-8510, Japan
| | - Masaki Ohmuraya
- Department of Genetics, Hyogo Medical University, Nishinomiya, Hyogo, 663-8501, Japan.
| |
Collapse
|
86
|
Gustafson DL, Viola LO, Towers CG, Das S, Duval DL, Van Eaton KM. Sensitivity of osteosarcoma cell lines to autophagy inhibition as determined by pharmacologic and genetic manipulation. Vet Comp Oncol 2023; 21:726-738. [PMID: 37724007 PMCID: PMC11470750 DOI: 10.1111/vco.12937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/05/2023] [Accepted: 09/05/2023] [Indexed: 09/20/2023]
Abstract
Pharmacologic inhibition of autophagy can be achieved using lysosomotropic agents such as hydroxychloroquine (HCQ) that interfere with fusion of the autophagosome to the lysosome thus preventing completion of the recycling process. The goal of the present study is to determine the sensitivity of eight canine (cOSA) and four human (hOSA) osteosarcoma tumour cell lines to antiproliferative and cytotoxic effects of lysosomal autophagy inhibitors, and to compare these results to the autophagy-dependence measured using a CRISPR/Cas9 live-cell imaging assay in OSA and other tumour cell lines. Antiproliferative and cytotoxic response to HCQ and Lys05 was determined using live cell imaging and YOYO-1 staining. CRISPR/Cas9 live cell imaging screen was done using species specific guide RNA's and transfection of reagents into cells. Response to autophagy core genes was compared to response to an essential (PCNA) and non-essential (FOXO3A) gene. cOSA and hOSA cell lines showed similar antiproliferative and cytotoxic responses to HCQ and Lys05 with median lethal dose (Dm ) values ranging from 4.6-15.8 μM and 2.1-5.1 μM for measures of anti-proliferative response, respectively. A relationship was observed between antiproliferative responses to HCQ and Lys05 and VPS34 CRISPR score with Dm values correlating with VPS34 response (r = 0.968 and 0.887) in a species independent manner. The results show that a subset of cOSA and hOSA cell lines are autophagy-dependent and sensitive to HCQ at pharmacologically-relevant exposures.
Collapse
Affiliation(s)
- Daniel L. Gustafson
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
- Developmental Therapeutics Program, University of Colorado Cancer Center, Aurora, Colorado, USA
| | - Lindsey O. Viola
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| | - Christina G. Towers
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Sciences, La Jolla, California, USA
| | - Sunetra Das
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| | - Dawn L. Duval
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
- Developmental Therapeutics Program, University of Colorado Cancer Center, Aurora, Colorado, USA
| | - Kristen M. Van Eaton
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, USA
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
87
|
Ye J, Zhang J, Zhu Y, Wang L, Jiang X, Liu B, He G. Targeting autophagy and beyond: Deconvoluting the complexity of Beclin-1 from biological function to cancer therapy. Acta Pharm Sin B 2023; 13:4688-4714. [PMID: 38045051 PMCID: PMC10692397 DOI: 10.1016/j.apsb.2023.08.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/05/2023] [Accepted: 08/02/2023] [Indexed: 12/05/2023] Open
Abstract
Beclin-1 is the firstly-identified mammalian protein of the autophagy machinery, which functions as a molecular scaffold for the assembly of PI3KC3 (class III phosphatidylinositol 3 kinase) complex, thus controlling autophagy induction and other cellular trafficking events. Notably, there is mounting evidence establishing the implications of Beclin-1 in diverse tumorigenesis processes, including tumor suppression and progression as well as resistance to cancer therapeutics and CSC (cancer stem-like cell) maintenance. More importantly, Beclin-1 has been confirmed as a potential target for the treatment of multiple cancers. In this review, we provide a comprehensive survey of the structure, functions, and regulations of Beclin-1, and we discuss recent advances in understanding the controversial roles of Beclin-1 in oncology. Moreover, we focus on summarizing the targeted Beclin-1-regulating strategies in cancer therapy, providing novel insights into a promising strategy for regulating Beclin-1 to improve cancer therapeutics in the future.
Collapse
Affiliation(s)
- Jing Ye
- Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Zhang
- Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanghui Zhu
- Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lian Wang
- Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease Related Molecular Network, Chengdu 610041, China
| | - Xian Jiang
- Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Liu
- Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gu He
- Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology (CIII), Frontiers Science Center for Disease Related Molecular Network, Chengdu 610041, China
| |
Collapse
|
88
|
Lee S, Son JY, Lee J, Cheong H. Unraveling the Intricacies of Autophagy and Mitophagy: Implications in Cancer Biology. Cells 2023; 12:2742. [PMID: 38067169 PMCID: PMC10706449 DOI: 10.3390/cells12232742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Autophagy is an essential lysosome-mediated degradation pathway that maintains cellular homeostasis and viability in response to various intra- and extracellular stresses. Mitophagy is a type of autophagy that is involved in the intricate removal of dysfunctional mitochondria during conditions of metabolic stress. In this review, we describe the multifaceted roles of autophagy and mitophagy in normal physiology and the field of cancer biology. Autophagy and mitophagy exhibit dual context-dependent roles in cancer development, acting as tumor suppressors and promoters. We also discuss the important role of autophagy and mitophagy within the cancer microenvironment and how autophagy and mitophagy influence tumor host-cell interactions to overcome metabolic deficiencies and sustain the activity of cancer-associated fibroblasts (CAFs) in a stromal environment. Finally, we explore the dynamic interplay between autophagy and the immune response in tumors, indicating their potential as immunomodulatory targets in cancer therapy. As the field of autophagy and mitophagy continues to evolve, this comprehensive review provides insights into their important roles in cancer and cancer microenvironment.
Collapse
Affiliation(s)
- Sunmi Lee
- Branch of Molecular Cancer Biology, Division of Cancer Biology, Research Institute, National Cancer Center, Goyang-si 10408, Republic of Korea; (S.L.); (J.-Y.S.)
| | - Ji-Yoon Son
- Branch of Molecular Cancer Biology, Division of Cancer Biology, Research Institute, National Cancer Center, Goyang-si 10408, Republic of Korea; (S.L.); (J.-Y.S.)
| | - Jinkyung Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science & Policy, National Cancer Center, Goyang-si 10408, Republic of Korea;
| | - Heesun Cheong
- Branch of Molecular Cancer Biology, Division of Cancer Biology, Research Institute, National Cancer Center, Goyang-si 10408, Republic of Korea; (S.L.); (J.-Y.S.)
- Department of Cancer Biomedical Science, Graduate School of Cancer Science & Policy, National Cancer Center, Goyang-si 10408, Republic of Korea;
| |
Collapse
|
89
|
Zhang L, Luo L, Zheng Q, Wang W, Huang Z, Hu J, Chen Y. Emodin derivative E35 and its combination with autophagy inhibitor against acute myeloid leukemia cells in vitro and in vivo. Exp Cell Res 2023; 432:113780. [PMID: 37742725 DOI: 10.1016/j.yexcr.2023.113780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 09/06/2023] [Accepted: 09/10/2023] [Indexed: 09/26/2023]
Abstract
Acute myeloid leukemia (AML) is an aggressive hematopoietic malignancy with poor prognosis and high recurrence rate. The discovery of more effective therapeutic strategies for AML plays a crucial role. The present work showed that E35, a novel derivative of emodin, significantly inhibited cell proliferation and induced autophagy and apoptosis in AML cells. Treatment with E35 markedly induced Beclin-1, LC3-II, cleaved Caspase-9 and PARP, and suppressed mitogen-activated protein kinase (MAPK) pathway. E35 exposure evoked autophagic activity prior to apoptosis induction, and autophagy inhibition by 3-methyladenine (3-MA) dramatically increased E35-induced apoptosis in both AML cell lines and patient-derived AML cells. Nevertheless, study on AML xenograft model showed that the combination E35 with 3-MA exhibited much more inhibitory effects on leukemia cell growth in vivo. No obvious adverse reactions occurred in the xenograft animals administered E35 alone or its cotreatment with 3-MA. These findings suggest that E35 could exert anti-leukemia effects, and that the combination of E35 and autophagy inhibitor might prove a more highly efficient strategy for AML treatment.
Collapse
Affiliation(s)
- Li Zhang
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of Hematology, The First Affiliated Hospital of Xiamen University, Xiamen, 361011, China
| | - Liping Luo
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Qiaoyun Zheng
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Wenfeng Wang
- Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Zhongyang Huang
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Jianda Hu
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| | - Yingyu Chen
- Department of Hematology, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| |
Collapse
|
90
|
Guil-Luna S, Sanchez-Montero MT, Rodríguez-Ariza A. S-Nitrosylation at the intersection of metabolism and autophagy: Implications for cancer. Biochim Biophys Acta Rev Cancer 2023; 1878:189012. [PMID: 37918453 DOI: 10.1016/j.bbcan.2023.189012] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/26/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023]
Abstract
Metabolic plasticity, which determines tumour growth and metastasis, is now understood to be a flexible and context-specific process in cancer metabolism. One of the major pathways contributing to metabolic adaptations in eucaryotic cells is autophagy, a cellular degradation and recycling process that is activated during periods of starvation or stress to maintain metabolite and biosynthetic intermediate levels. Consequently, there is a close association between the metabolic adaptive capacity of tumour cells and autophagy-related pathways in cancer. Additionally, nitric oxide regulates protein function and signalling through S-nitrosylation, a post-translational modification that can also impact metabolism and autophagy. The primary objective of this review is to provide an up-to-date overview of the role of S-nitrosylation at the intersection of metabolism and autophagy in cancer. First, we will outline the involvement of S-nitrosylation in the metabolic adaptations that occur in tumours. Then, we will discuss the multifaceted role of autophagy in cancer, the interplay between metabolism and autophagy during tumour progression, and the contribution of S-nitrosylation to autophagic dysregulation in cancer. Finally, we will present insights into relevant therapeutic aspects and discuss prospects for the future.
Collapse
Affiliation(s)
- Silvia Guil-Luna
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Cancer Network Biomedical Research Center (CIBERONC), Madrid, Spain; Department of Comparative Anatomy and Pathology, Faculty of Veterinary Medicine of Córdoba, University of Córdoba, Córdoba, Spain
| | | | - Antonio Rodríguez-Ariza
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain; Cancer Network Biomedical Research Center (CIBERONC), Madrid, Spain; Medical Oncology Department, Reina Sofía University Hospital, Córdoba, Spain.
| |
Collapse
|
91
|
Torres-López L, Dobrovinskaya O. Dissecting the Role of Autophagy-Related Proteins in Cancer Metabolism and Plasticity. Cells 2023; 12:2486. [PMID: 37887330 PMCID: PMC10605719 DOI: 10.3390/cells12202486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023] Open
Abstract
Modulation of autophagy as an anticancer strategy has been widely studied and evaluated in several cell models. However, little attention has been paid to the metabolic changes that occur in a cancer cell when autophagy is inhibited or induced. In this review, we describe how the expression and regulation of various autophagy-related (ATGs) genes and proteins are associated with cancer progression and cancer plasticity. We present a comprehensive review of how deregulation of ATGs affects cancer cell metabolism, where inhibition of autophagy is mainly reflected in the enhancement of the Warburg effect. The importance of metabolic changes, which largely depend on the cancer type and form part of a cancer cell's escape strategy after autophagy modulation, is emphasized. Consequently, pharmacological strategies based on a dual inhibition of metabolic and autophagy pathways emerged and are reviewed critically here.
Collapse
Affiliation(s)
- Liliana Torres-López
- Laboratory of Immunology and Ionic Transport Regulation, Biomedical Research Centre, University of Colima, Av. 25 de Julio #965, Villas de San Sebastián, Colima 28045, Mexico;
| | | |
Collapse
|
92
|
Liu Y, Chen L, Meng F, Zhang T, Luo J, Chen S, Shi H, Liu B, Lv Z. The Effect of Temperature on the Embryo Development of Cephalopod Sepiella japonica Suggests Crosstalk between Autophagy and Apoptosis. Int J Mol Sci 2023; 24:15365. [PMID: 37895043 PMCID: PMC10607546 DOI: 10.3390/ijms242015365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Temperature is a crucial environmental factor that affects embryonic development, particularly for marine organisms with long embryonic development periods. However, the sensitive period of embryonic development and the role of autophagy/apoptosis in temperature regulation in cephalopods remain unclear. In this study, we cultured embryos of Sepiella japonica, a typical species in the local area of the East China Sea, at different incubation temperatures (18 °C, 23 °C, and 28 °C) to investigate various developmental aspects, including morphological and histological characteristics, mortality rates, the duration of embryonic development, and expression patterns of autophagy-related genes (LC3, BECN1, Inx4) and apoptosis marker genes (Cas3, p53) at 25 developmental stages. Our findings indicate that embryos in the high-temperature (28 °C) group had significantly higher mortality and embryonic malformation rates than those in the low-temperature (18 °C) group. Furthermore, high temperature (28 °C) shortened the duration of embryonic development by 7 days compared to the optimal temperature (23 °C), while low temperature (18 °C) caused a delay of 9 days. Therefore, embryos of S. japonica were more intolerant to high temperatures (28 °C), emphasizing the critical importance of maintaining an appropriate incubation temperature (approximately 23 °C). Additionally, our study observed, for the first time, that the Early blastula, Blastopore closure, and Optic vesicle to Caudal end stages were the most sensitive stages. During these periods, abnormalities in the expression of autophagy-related and apoptosis-related genes were associated with higher rates of mortality and malformations, highlighting the strong correlation and potential interaction between autophagy and apoptosis in embryonic development under varying temperature conditions.
Collapse
Affiliation(s)
- Yifan Liu
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan 316022, China; (Y.L.); (F.M.)
| | - Long Chen
- Marine Science and Technical College, Zhejiang Ocean University, Zhoushan 316022, China; (L.C.); (J.L.); (S.C.)
| | - Fang Meng
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan 316022, China; (Y.L.); (F.M.)
| | - Tao Zhang
- Zhejiang Province Key Lab of Mariculture and Enhancement, Marine Fisheries Research Institute of Zhejiang, Zhoushan 316021, China; (T.Z.); (H.S.)
| | - Jun Luo
- Marine Science and Technical College, Zhejiang Ocean University, Zhoushan 316022, China; (L.C.); (J.L.); (S.C.)
| | - Shuang Chen
- Marine Science and Technical College, Zhejiang Ocean University, Zhoushan 316022, China; (L.C.); (J.L.); (S.C.)
| | - Huilai Shi
- Zhejiang Province Key Lab of Mariculture and Enhancement, Marine Fisheries Research Institute of Zhejiang, Zhoushan 316021, China; (T.Z.); (H.S.)
| | - Bingjian Liu
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan 316022, China; (Y.L.); (F.M.)
- Marine Science and Technical College, Zhejiang Ocean University, Zhoushan 316022, China; (L.C.); (J.L.); (S.C.)
| | - Zhenming Lv
- National Engineering Research Center for Marine Aquaculture, Zhejiang Ocean University, Zhoushan 316022, China; (Y.L.); (F.M.)
- Marine Science and Technical College, Zhejiang Ocean University, Zhoushan 316022, China; (L.C.); (J.L.); (S.C.)
| |
Collapse
|
93
|
García-Pérez BE, Pérez-Torres C, Baltierra-Uribe SL, Castillo-Cruz J, Castrejón-Jiménez NS. Autophagy as a Target for Non-Immune Intrinsic Functions of Programmed Cell Death-Ligand 1 in Cancer. Int J Mol Sci 2023; 24:15016. [PMID: 37834467 PMCID: PMC10573536 DOI: 10.3390/ijms241915016] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/27/2023] [Accepted: 10/07/2023] [Indexed: 10/15/2023] Open
Abstract
Autophagy is a catabolic process that is essential to the maintenance of homeostasis through the cellular recycling of damaged organelles or misfolded proteins, which sustains energy balance. Additionally, autophagy plays a dual role in modulating the development and progression of cancer and inducing a survival strategy in tumoral cells. Programmed cell death-ligand 1 (PD-L1) modulates the immune response and is responsible for maintaining self-tolerance. Because tumor cells exploit the PD-L1-PD-1 interaction to subvert the immune response, immunotherapy has been developed based on the use of PD-L1-blocking antibodies. Recent evidence has suggested a bidirectional regulation between autophagy and PD-L1 molecule expression in tumor cells. Moreover, the research into the intrinsic properties of PD-L1 has highlighted new functions that are advantageous to tumor cells. The relationship between autophagy and PD-L1 is complex and still not fully understood; its effects can be context-dependent and might differ between tumoral cells. This review refines our understanding of the non-immune intrinsic functions of PD-L1 and its potential influence on autophagy, how these could allow the survival of tumor cells, and what this means for the efficacy of anti-PD-L1 therapeutic strategies.
Collapse
Affiliation(s)
- Blanca Estela García-Pérez
- Departmento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Col. Santo Tomás, Alcaldía Miguel Hidalgo, Mexico City 11340, Mexico
| | - Christian Pérez-Torres
- Departmento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Col. Santo Tomás, Alcaldía Miguel Hidalgo, Mexico City 11340, Mexico
| | - Shantal Lizbeth Baltierra-Uribe
- Departmento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Col. Santo Tomás, Alcaldía Miguel Hidalgo, Mexico City 11340, Mexico
| | - Juan Castillo-Cruz
- Departmento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Col. Santo Tomás, Alcaldía Miguel Hidalgo, Mexico City 11340, Mexico
- Departmento de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Prolongación de Carpio y Plan de Ayala S/N, Col. Santo Tomás, Alcaldía Miguel Hidalgo, Mexico City 11340, Mexico
| | - Nayeli Shantal Castrejón-Jiménez
- Área Académica de Medicina Veterinaria y Zootecnia, Instituto de Ciencias Agropecuarias, Universidad Autónoma del Estado de Hidalgo, Av. Universidad km. 1. Exhacienda de Aquetzalpa A.P. 32, Tulancingo 43600, Mexico
| |
Collapse
|
94
|
Chueh KS, Lu JH, Juan TJ, Chuang SM, Juan YS. The Molecular Mechanism and Therapeutic Application of Autophagy for Urological Disease. Int J Mol Sci 2023; 24:14887. [PMID: 37834333 PMCID: PMC10573233 DOI: 10.3390/ijms241914887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Autophagy is a lysosomal degradation process known as autophagic flux, involving the engulfment of damaged proteins and organelles by double-membrane autophagosomes. It comprises microautophagy, chaperone-mediated autophagy (CMA), and macroautophagy. Macroautophagy consists of three stages: induction, autophagosome formation, and autolysosome formation. Atg8-family proteins are valuable for tracking autophagic structures and have been widely utilized for monitoring autophagy. The conversion of LC3 to its lipidated form, LC3-II, served as an indicator of autophagy. Autophagy is implicated in human pathophysiology, such as neurodegeneration, cancer, and immune disorders. Moreover, autophagy impacts urological diseases, such as interstitial cystitis /bladder pain syndrome (IC/BPS), ketamine-induced ulcerative cystitis (KIC), chemotherapy-induced cystitis (CIC), radiation cystitis (RC), erectile dysfunction (ED), bladder outlet obstruction (BOO), prostate cancer, bladder cancer, renal cancer, testicular cancer, and penile cancer. Autophagy plays a dual role in the management of urologic diseases, and the identification of potential biomarkers associated with autophagy is a crucial step towards a deeper understanding of its role in these diseases. Methods for monitoring autophagy include TEM, Western blot, immunofluorescence, flow cytometry, and genetic tools. Autophagosome and autolysosome structures are discerned via TEM. Western blot, immunofluorescence, northern blot, and RT-PCR assess protein/mRNA levels. Luciferase assay tracks flux; GFP-LC3 transgenic mice aid study. Knockdown methods (miRNA and RNAi) offer insights. This article extensively examines autophagy's molecular mechanism, pharmacological regulation, and therapeutic application involvement in urological diseases.
Collapse
Affiliation(s)
- Kuang-Shun Chueh
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Road, San-min District, Kaohsiung 80708, Taiwan;
- Department of Urology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
- Department of Urology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Jian-He Lu
- Center for Agricultural, Forestry, Fishery, Livestock and Aquaculture Carbon Emission Inventory and Emerging Compounds (CAFEC), General Research Service Center, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan;
| | - Tai-Jui Juan
- Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan;
- Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
| | - Shu-Mien Chuang
- Department of Urology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Yung-Shun Juan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, No. 100, Shih-Chuan 1st Road, San-min District, Kaohsiung 80708, Taiwan;
- Department of Urology, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| |
Collapse
|
95
|
Bierlein M, Charles J, Polisuk-Balfour T, Bretscher H, Rice M, Zvonar J, Pohl D, Winslow L, Wasie B, Deurloo S, Van Wert J, Williams B, Ankney G, Harmon Z, Dann E, Azuz A, Guzman-Vargas A, Kuhns E, Neufeld TP, O’Connor MB, Amissah F, Zhu CC. Autophagy impairment and lifespan reduction caused by Atg1 RNAi or Atg18 RNAi expression in adult fruit flies (Drosophila melanogaster). Genetics 2023; 225:iyad154. [PMID: 37594076 PMCID: PMC11491525 DOI: 10.1093/genetics/iyad154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/03/2023] [Indexed: 08/19/2023] Open
Abstract
Autophagy, an autophagosome and lysosome-based eukaryotic cellular degradation system, has previously been implicated in lifespan regulation in different animal models. In this report, we show that expression of the RNAi transgenes targeting the transcripts of the key autophagy genes Atg1 or Atg18 in adult fly muscle or glia does not affect the overall levels of autophagosomes in those tissues and does not change the lifespan of the tested flies but the lifespan reduction phenotype has become apparent when Atg1 RNAi or Atg18 RNAi is expressed ubiquitously in adult flies or after autophagy is eradicated through the knockdown of Atg1 or Atg18 in adult fly adipocytes. Lifespan reduction was also observed when Atg1 or Atg18 was knocked down in adult fly enteroblasts and midgut stem cells. Overexpression of wild-type Atg1 in adult fly muscle or adipocytes reduces the lifespan and causes accumulation of high levels of ubiquitinated protein aggregates in muscles. Our research data have highlighted the important functions of the key autophagy genes in adult fly adipocytes, enteroblasts, and midgut stem cells and their undetermined roles in adult fly muscle and glia for lifespan regulation.
Collapse
Affiliation(s)
- Mariah Bierlein
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Joseph Charles
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | | | - Heidi Bretscher
- Department of Genetics, Cell Biology, and Developmental Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Micaela Rice
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Jacklyn Zvonar
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Drake Pohl
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Lindsey Winslow
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Brennah Wasie
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Sara Deurloo
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Jordan Van Wert
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Britney Williams
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Gabrielle Ankney
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Zachary Harmon
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Erica Dann
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Anna Azuz
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Alex Guzman-Vargas
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Elizabeth Kuhns
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| | - Thomas P Neufeld
- Department of Genetics, Cell Biology, and Developmental Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael B O’Connor
- Department of Genetics, Cell Biology, and Developmental Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Felix Amissah
- School of Pharmacy, Ferris State University, Big Rapids, MI 49307, USA
| | - Changqi C Zhu
- Department of Biological Sciences, Ferris State University, Big Rapids, MI 49307, USA
| |
Collapse
|
96
|
Ahmadi S, Saberivand A, Jalili C, Asadpour R, Khordadmehr M, Saberivand M. Hydroalcoholic extract of Taraxacum officinale induces apoptosis and autophagy in 4T1 breast cancer cells. VETERINARY RESEARCH FORUM : AN INTERNATIONAL QUARTERLY JOURNAL 2023; 14:507-513. [PMID: 37814658 PMCID: PMC10560329 DOI: 10.30466/vrf.2023.1985987.3726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 01/26/2023] [Indexed: 10/11/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and deadly breast cancer sub-type with limited therapeutic options. Dandelion (Taraxacum officinale) exhibiting extensive anti-cancer activity is reported to be effective against TNBC; however, its anti-tumor effect mechanisms have not been fully elucidated. The purpose of this study was to determine the anti-cancer activity of hydroalcoholic extract of dandelion (HADE) on 4T1 cells, and the mechanism of HADE-induced cell death. The effect of HADE on cell viability was assessed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and lactate dehydrogenase assays. Apoptotic cell death was monitored by flow cytometry. The DNA fragmentation was evaluated by Acridine orange/Ethidium bromide (AO/EB) staining. Nitric oxide (NO) level was detected using Griess assay. The effects of HADE on Atg-7, Beclin-1, Bcl2, Bax and p53 genes were investigated by real-time reverse transcription-polymerase chain reaction. The results showed that HADE inhibited cell growth and proliferation in a dose- and time-dependent manner. The HADE induced 4T1 breast cancer cell death via apoptosis and autophagy. The DNA fragmentation was improved as the concentration of HADE increased. The NO secretion was declined with increasing concentration of HADE. Gene expression analysis confirmed HADE-induced apoptosis and autophagy in cancer cells. The Bax, Bax/Bcl-2 ratio, p53, Beclin-1 and Atg-7 over-expression as well as Bcl-2 down-regulation were also evident in treated cancer cells.
Collapse
Affiliation(s)
- Sharareh Ahmadi
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Adel Saberivand
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Cyrus Jalili
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Asadpour
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Monire Khordadmehr
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Maryam Saberivand
- Connective Tissue Diseases Research Center, Tabriz University of Medical Science, Tabriz, Iran
| |
Collapse
|
97
|
Rodriguez M, Owens F, Perry M, Stone N, Soler Y, Almohtadi R, Zhao Y, Batrakova EV, El-Hage N. Implication of the Autophagy-Related Protein Beclin1 in the Regulation of EcoHIV Replication and Inflammatory Responses. Viruses 2023; 15:1923. [PMID: 37766329 PMCID: PMC10537636 DOI: 10.3390/v15091923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/05/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
The protein Beclin1 (BECN1, a mammalian homologue of ATG6 in yeast) plays an important role in the initiation and the normal process of autophagy in cells. Moreover, we and others have shown that Beclin1 plays an important role in viral replication and the innate immune signaling pathways. We previously used the cationic polymer polyethyleneimine (PEI) conjugated to mannose (Man) as a non-viral tool for the delivery of a small interfering (si) Beclin1-PEI-Man nanoplex, which specifically targets mannose receptor-expressing glia (microglia and astrocytes) in the brain when administered intranasally to conventional mice. To expand our previous reports, first we used C57BL/6J mice infected with EcoHIV and exposed them to combined antiretroviral therapy (cART). We show that EcoHIV enters the mouse brain, while intranasal delivery of the nanocomplex significantly reduces the secretion of HIV-induced inflammatory molecules and downregulates the expression of the transcription factor nuclear factor (NF)-kB. Since a spectrum of neurocognitive and motor problems can develop in people living with HIV (PLWH) despite suppressive antiretroviral therapy, we subsequently measured the role of Beclin1 in locomotor activities using EcoHIV-infected BECN1 knockout mice exposed to cART. Viral replication and cytokine secretion were reduced in the postmortem brains recovered from EcoHIV-infected Becn1+/- mice when compared to EcoHIV-infected Becn1+/+ mice, although the impairment in locomotor activities based on muscle strength were comparable. This further highlights the importance of Beclin1 in the regulation of HIV replication and in viral-induced cytokine secretion but not in HIV-induced locomotor impairments. Moreover, the cause of HIV-induced locomotor impairments remains speculative, as we show that this may not be entirely due to viral load and/or HIV-induced inflammatory cytokines.
Collapse
Affiliation(s)
- Myosotys Rodriguez
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (M.R.); (F.O.); (M.P.); (N.S.); (Y.S.); (R.A.)
| | - Florida Owens
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (M.R.); (F.O.); (M.P.); (N.S.); (Y.S.); (R.A.)
| | - Marissa Perry
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (M.R.); (F.O.); (M.P.); (N.S.); (Y.S.); (R.A.)
| | - Nicole Stone
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (M.R.); (F.O.); (M.P.); (N.S.); (Y.S.); (R.A.)
| | - Yemmy Soler
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (M.R.); (F.O.); (M.P.); (N.S.); (Y.S.); (R.A.)
| | - Rianna Almohtadi
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (M.R.); (F.O.); (M.P.); (N.S.); (Y.S.); (R.A.)
| | - Yuling Zhao
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.Z.); (E.V.B.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Elena V. Batrakova
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.Z.); (E.V.B.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Nazira El-Hage
- Department of Immunology and Nanomedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (M.R.); (F.O.); (M.P.); (N.S.); (Y.S.); (R.A.)
| |
Collapse
|
98
|
Jin J, Zhu KS, Tang SM, Xiang Y, Mao MY, Hong XD, Chen AF, Zhang XD, Lu H, Chen ZL, Wu JF, Pan SF, Zhu SH. Trehalose promotes functional recovery of keratinocytes under oxidative stress and wound healing via ATG5/ATG7. Burns 2023; 49:1382-1391. [PMID: 36759218 DOI: 10.1016/j.burns.2022.11.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 11/07/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022]
Abstract
Wounds are in a stressed state, which precludes healing. Trehalose is a stress metabolite that protects cells under stress. Here, we explored whether trehalose reduces stress-induced wound tissue damage. A stress model was prepared by exposing human keratinocytes to hydrogen peroxide (H2O2), followed by trehalose treatment. Trehalose effects on expression of the autophagy-related proteins ATG5 and ATG7 and cell proliferation and migration were evaluated. For in vivo verification, a wound model was established in Sprague-Dawley rats, to measure the effects of trehalose wound-healing rate and reactive oxygen species (ROS) content. Histological changes during wound healing and trehalose's effects on ATG5 and ATG7 expression, necrosis, and apoptosis were examined·H2O2 stress increased ATG5 and ATG7 expression in vitro, but this was insufficient to prevent stress-induced damage. Trehalose further increased ATG5/ATG7 levels, which restored proliferation and increased migration by depolymerizing the cytoskeleton. However, trehalose did not exert these effects after ATG5 and ATG7 knockout. In vivo, the ROS content was higher in the wound tissue than in normal skin. Trehalose increased ATG5/ATG7 expression in wound tissue keratinocytes, reduced necrosis, depolymerized the cytoskeleton, and promoted cell migration, thereby promoting wound healing.
Collapse
Affiliation(s)
- Jian Jin
- Department of Burn Surgery, Changhai Hospital, The Second Military Medical University, Baoshan District, Shanghai, China; Shanghai Depeac Biotechnology Co., Ltd., Yangpu District, Shanghai, China
| | - Kai-Si Zhu
- Department of Anesthesiology, Changhai Hospital, The Second Military Medical University, Baoshan District, Shanghai, China
| | - Shu-Min Tang
- Department of Emergency, Changhai Hospital, The Second Military Medical University, Baoshan District, Shanghai, China
| | - Yang Xiang
- Department of Burn Surgery, Changhai Hospital, The Second Military Medical University, Baoshan District, Shanghai, China
| | - Ming-Yi Mao
- Shanghai Depeac Biotechnology Co., Ltd., Yangpu District, Shanghai, China
| | - Xu-Dong Hong
- Department of Burn & Plastic Surgery, 903rd Hospital of PLA (The Original 117th Hospital of People's Liberation Army), Xihu District, Hangzhou, China
| | - Ai-Fen Chen
- Department of Burn & Plastic Surgery, 903rd Hospital of PLA (The Original 117th Hospital of People's Liberation Army), Xihu District, Hangzhou, China
| | - Xu-Dong Zhang
- Department of Burn & Plastic Surgery, 903rd Hospital of PLA (The Original 117th Hospital of People's Liberation Army), Xihu District, Hangzhou, China
| | - Hao Lu
- Shanghai Depeac Biotechnology Co., Ltd., Yangpu District, Shanghai, China
| | - Zheng-Li Chen
- Shanghai Depeac Biotechnology Co., Ltd., Yangpu District, Shanghai, China
| | - Jin-Fang Wu
- Department of Burn & Plastic Surgery, 903rd Hospital of PLA (The Original 117th Hospital of People's Liberation Army), Xihu District, Hangzhou, China
| | - Sun-Feng Pan
- Department of Burns and Plastic Surgery, Zhejiang Chinese Medical University Affiliated Jiaxing TCM Hospital, Zhejiang Province, China.
| | - Shi-Hui Zhu
- Shanghai Depeac Biotechnology Co., Ltd., Yangpu District, Shanghai, China.
| |
Collapse
|
99
|
Hong T, Wen J, Mei L, Li R, Zhou J, Li J, Xiao XH. Insulin Inhibits Autophagy by Inhibiting the Binding of FoXO1 to the Promoter Region of GABARAPL1. Horm Metab Res 2023; 55:634-641. [PMID: 37380030 PMCID: PMC10484641 DOI: 10.1055/a-2119-3301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/28/2023] [Indexed: 06/30/2023]
Abstract
Hyperinsulinemia and insulin resistance in T2D have a potent suppressive effect on hepatic autophagy, however, the underlying mechanisms remain unclear. To explore the effect of insulin on hepatic autophagy and its possible signaling pathways, HL-7702 cells were treated with insulin with or without insulin signaling inhibitors. The interaction between insulin and the promoter region of GABARAPL1 was assessed through luciferase assay and EMSA. There were significant dose-dependent decreases in the number of intracellular autophagosomes and the protein levels of GABARAPL1 and beclin1 in insulin-treated HL-7702 cells. Insulin signaling inhibitors reversed the inhibitory effect of insulin on rapamycin-induced autophagy and autophagy-related gene upregulation. Insulin blocks the binding of FoxO1 to putative insulin response elements in GABARAPL1 gene promoter, leading to the repressed transcription of GABARAPL1 gene and the suppression of hepatic autophagy. Our study identified GABARAPL1 as a novel target of insulin in suppressing hepatic autophagy.
Collapse
Affiliation(s)
- Tao Hong
- The Second Affiliated Hospital, Department of Endocrinology and
Metabolism, University of South China Medical College, Hengyang,
China
| | - Jie Wen
- The First Affiliated Hospital, Department of Endocrinology and
Metabolism, University of South China Medical College, Hengyang,
China
| | - Lang Mei
- The First Affiliated Hospital, Department of Endocrinology and
Metabolism, University of South China Medical College, Hengyang,
China
| | - Ruixiang Li
- The First Affiliated Hospital, Department of Endocrinology and
Metabolism, University of South China Medical College, Hengyang,
China
| | - Junlin Zhou
- The First Affiliated Hospital, Health Management Center, University of
South China Medical College, Hengyang, China
| | - Jiaoyang Li
- The First Affiliated Hospital, Department of Endocrinology and
Metabolism, University of South China Medical College, Hengyang,
China
| | - Xin-Hua Xiao
- The First Affiliated Hospital, Department of Endocrinology and
Metabolism, University of South China Medical College, Hengyang,
China
| |
Collapse
|
100
|
Zhang X, Shan G, Li N, Chen J, Ji C, Li X, Jiang L, Lee TKW, Keng VW, Zhao Y. An autophagy-inducing stapled peptide induces mitochondria dysfunction and triggers autotic cell death in triple-negative breast cancer. Cell Death Discov 2023; 9:303. [PMID: 37598181 PMCID: PMC10439894 DOI: 10.1038/s41420-023-01600-0] [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: 11/25/2022] [Revised: 07/06/2023] [Accepted: 08/10/2023] [Indexed: 08/21/2023] Open
Abstract
Autophagy is a lysosome-dependent bulk degradation process essential for cell viability but excessive autophagy leads to a unique form of cell death termed autosis. Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer with notable defect in its autophagy process. In previous studies, we developed stapled peptides that specifically targeted the essential autophagy protein Beclin 1 to induce autophagy and promote endolysosomal trafficking. Here we show that one lead peptide Tat-SP4 induced mild increase of autophagy in TNBC cells but showed potent anti-proliferative effect that could not be rescued by inhibitors of programmed cell death pathways. The cell death induced by Tat-SP4 showed typical features of autosis including sustained adherence to the substrate surface, rupture of plasma membrane and effective rescue by digoxin, a cardioglycoside that blocks the Na+/K+ ATPase. Tat-SP4 also induced prominent mitochondria dysfunction including loss of mitochondria membrane potential, elevated mitochondria reactive oxygen species and reduced oxidative phosphorylation. The anti-proliferative effect of Tat-SP4 was confirmed in a TNBC xenograft model. Our study uncovers three notable aspects of autosis. Firstly, autosis can be triggered by moderate increase in autophagy if such increase exceeds the endogenous capacity of the host cells. Secondly, mitochondria may play an essential role in autosis with dysregulated autophagy leading to mitochondria dysfunction to trigger autosis. Lastly, intrinsic autophagy deficiency and quiescent mitochondria bioenergetic profile likely render TNBC cells particularly susceptible to autosis. Our designed peptides like Tat-SP4 may serve as potential therapeutic candidates against TNBC by targeting this vulnerability.
Collapse
Affiliation(s)
- Xiaozhe Zhang
- Department of Applied Biology and Chemical Technology, State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, 999077, Hong Kong, P. R. China
| | - Gao Shan
- Department of Applied Biology and Chemical Technology, State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, 999077, Hong Kong, P. R. China
| | - Na Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, 518057, Shenzhen, P. R. China
| | - Jingyi Chen
- Department of Applied Biology and Chemical Technology, State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, 999077, Hong Kong, P. R. China
| | - Changyang Ji
- School of Life Sciences, Centre for Cell & Developmental Biology, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Xiaoxiao Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, 518057, Shenzhen, P. R. China
| | - Liwen Jiang
- School of Life Sciences, Centre for Cell & Developmental Biology, State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Terence Kin Wah Lee
- Department of Applied Biology and Chemical Technology, State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, 999077, Hong Kong, P. R. China
| | - Vincent W Keng
- Department of Applied Biology and Chemical Technology, State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, 999077, Hong Kong, P. R. China
| | - Yanxiang Zhao
- Department of Applied Biology and Chemical Technology, State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Kowloon, 999077, Hong Kong, P. R. China.
- The Hong Kong Polytechnic University Shenzhen Research Institute, 518057, Shenzhen, P. R. China.
| |
Collapse
|