1
|
Vafiadaki E, Kranias EG, Eliopoulos AG, Sanoudou D. The phospholamban R14del generates pathogenic aggregates by impairing autophagosome-lysosome fusion. Cell Mol Life Sci 2024; 81:450. [PMID: 39527246 PMCID: PMC11554986 DOI: 10.1007/s00018-024-05471-1] [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: 01/11/2024] [Revised: 08/30/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
Phospholamban (PLN) plays a crucial role in regulating sarcoplasmic reticulum (SR) Ca2+ cycling and cardiac contractility. Mutations within the PLN gene have been detected in patients with cardiomyopathy, with the heterozygous variant c.40_42delAGA (p.R14del) of PLN being the most prevalent. Investigations into the mechanisms underlying the pathology of PLN-R14del have revealed that cardiac cells from affected patients exhibit pathological aggregates containing PLN. Herein, we performed comprehensive molecular and cellular analyses to delineate the molecular aberrations associated with the formation of these aggregates. We determined that PLN aggregates contain autophagic proteins, indicating inefficient degradation via the autophagy pathway. Our findings demonstrate that the expression of PLN-R14del results in diminished autophagic flux due to impaired fusion between autophagosomes and lysosomes. Mechanistically, this defect is linked to aberrant recruitment of key membrane fusion proteins to autophagosomes, which is mediated in part by changes in Ca2+ homeostasis. Collectively, these results highlight a novel function of PLN-R14del in regulating autophagy, that may contribute to the formation of pathogenic aggregates in patients with cardiomyopathy. Prospective strategies tailored to ameliorate impaired autophagy may hold promise against PLN-R14del disease.
Collapse
Affiliation(s)
- Elizabeth Vafiadaki
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece.
| | - Evangelia G Kranias
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Aristides G Eliopoulos
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
- Department of Biology, School of Medicine, National and Kapodistrian University of Athens, Mikras Asias 75, Athens, 11527, Greece
- Center for New Biotechnologies and Precision Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - Despina Sanoudou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece.
- Center for New Biotechnologies and Precision Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece.
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece.
| |
Collapse
|
2
|
Mills TS, Kain B, Burchill MA, Danis E, Lucas ED, Culp-Hill R, Cowan CM, Schleicher WE, Patel SB, Tran BT, Cao R, Goodspeed A, Ferrara S, Bevers S, Jirón Tamburini BA, Roede JR, D'Alessandro A, King KY, Pietras EM. A distinct metabolic and epigenetic state drives trained immunity in HSC-derived macrophages from autoimmune mice. Cell Stem Cell 2024; 31:1630-1649.e8. [PMID: 39413777 PMCID: PMC11560650 DOI: 10.1016/j.stem.2024.09.010] [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: 03/06/2024] [Revised: 06/18/2024] [Accepted: 09/11/2024] [Indexed: 10/18/2024]
Abstract
Here, we investigate the contribution of long-term hematopoietic stem cells (HSCsLT) to trained immunity (TI) in the setting of chronic autoimmune disease. Using a mouse model of systemic lupus erythematosus (SLE), we show that bone marrow-derived macrophages (BMDMs) from autoimmune mice exhibit hallmark features of TI, including increased Mycobacterium avium killing and inflammatory cytokine production, which are mechanistically linked to increased glycolytic metabolism. We show that HSCs from autoimmune mice constitute a transplantable, long-term reservoir for macrophages that exhibit the functional properties of TI. However, these BMDMs exhibit reduced glycolytic activity and chromatin accessibility at metabolic genes while retaining elevated expression of TI-associated transcriptional regulators. Hence, HSC exposed to autoimmune inflammation can give rise to macrophages in which the functional and metabolic properties of TI are decoupled. Our data support a model in which TI is characterized by a spectrum of molecular and metabolic states driving augmented immune function.
Collapse
Affiliation(s)
- Taylor S Mills
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Immunology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Bailee Kain
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Matt A Burchill
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Etienne Danis
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Erin D Lucas
- Immunology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Courtney M Cowan
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Immunology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Wolfgang E Schleicher
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sweta B Patel
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Brandon T Tran
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ruoqiong Cao
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Andrew Goodspeed
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sarah Ferrara
- University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Shaun Bevers
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Beth A Jirón Tamburini
- Immunology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - James R Roede
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Katherine Y King
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Eric M Pietras
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
3
|
Wen W, Ertas YN, Erdem A, Zhang Y. Dysregulation of autophagy in gastric carcinoma: Pathways to tumor progression and resistance to therapy. Cancer Lett 2024; 591:216857. [PMID: 38583648 DOI: 10.1016/j.canlet.2024.216857] [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: 02/06/2024] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
The considerable death rates and lack of symptoms in early stages of gastric cancer (GC) make it a major health problem worldwide. One of the most prominent risk factors is infection with Helicobacter pylori. Many biological processes, including those linked with cell death, are disrupted in GC. The cellular "self-digestion" mechanism necessary for regular balance maintenance, autophagy, is at the center of this disturbance. Misregulation of autophagy, however, plays a role in the development of GC. In this review, we will examine how autophagy interacts with other cell death processes, such as apoptosis and ferroptosis, and how it affects the progression of GC. In addition to wonderful its role in the epithelial-mesenchymal transition, it is engaged in GC metastasis. The role of autophagy in GC in promoting drug resistance stands out. There is growing interest in modulating autophagy for GC treatment, with research focusing on natural compounds, small-molecule inhibitors, and nanoparticles. These approaches could lead to breakthroughs in GC therapy, offering new hope in the fight against this challenging disease.
Collapse
Affiliation(s)
- Wen Wen
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, China
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey.
| | - Ahmet Erdem
- 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; Department of Biomedical Engineering, Kocaeli University, Umuttepe Campus, Kocaeli, 41001 Turkey.
| | - Yao Zhang
- Department of Gynaecology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
4
|
Mukai H, Takanashi M, Ogawara KI, Maruyama M, Higaki K. Possible Regulation of P-Glycoprotein Function by Adrenergic Agonists II: Study with Isolated Rat Jejunal Sheets and Caco-2 Cell monolayers. J Pharm Sci 2024; 113:1209-1219. [PMID: 37984697 DOI: 10.1016/j.xphs.2023.11.010] [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: 09/26/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/22/2023]
Abstract
To clarify the regulation of drug absorption by the enteric nervous system, we investigated how adrenergic agonists (adrenaline (ADR), clonidine (CLO), dobutamine (DOB)) and dibutyryl cAMP (DBcAMP) affected P-glycoprotein (P-gp) function by utilizing isolated rat jejunal sheets and Caco-2 cell monolayers. ADR and CLO significantly decreased the secretory transport (Papptotal) of rhodamine-123 and tended to decrease the transport via P-gp (PappP-gp) and passive transport (Papppassive). In contrast, DBcAMP significantly increased and DOB tended to increase Papptotal and both tended to increase PappP-gpand Papppassive. Changes in P-gp expression on brush border membrane by adrenergic agonists and DBcAMP were significantly correlated with PappP-gp, while P-gp expression was not changed in whole cell homogenates, suggesting that the trafficking of P-gp would be responsible for its functional changes. Papppassive was inversely correlated with transmucosal or transepithelial electrical resistance, indicating that adrenergic agonists affected the paracellular permeability. Adrenergic agonists also changed cAMP levels, which were significantly correlated with PappP-gp. Furthermore, protein kinase A (PKA) or PKC inhibitor significantly decreased PappP-gp in Caco-2 cell monolayers, suggesting that they would partly contribute to the changes in P-gp activity. In conclusion, adrenergic agonists regulated P-gp function and paracellular permeability, which would be caused via adrenoceptor stimulation.
Collapse
Affiliation(s)
- Hironori Mukai
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan; Production Department, Odawara Central Factory, Nippon Shinyaku Co., Ltd., 676-1 Kuwahara, Odawara, Kanagawa 250-0861, Japan
| | - Masashi Takanashi
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan; Central Hyogo Area, Hanshin Dispensing Pharmacy, I & H Co., Ltd., 1-18 Ohmasu-cho, Ashiya, Hyogo 659-0066, Japan
| | - Ken-Ichi Ogawara
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1, Motoyamakita, Higashinada-ku, Kobe, Hyogo 658-8558, Japan
| | - Masato Maruyama
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Kazutaka Higaki
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| |
Collapse
|
5
|
Yang X, Weng Q, Li X, Lu K, Wang L, Song K, Zhang C, Rahimnejad S. High water temperature raised the requirements of methionine for spotted seabass (Lateolabrax maculatus). FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:23-40. [PMID: 36322361 DOI: 10.1007/s10695-022-01136-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/17/2022] [Indexed: 06/16/2023]
Abstract
This study evaluated the effects of dietary methionine level and rearing water temperature on growth, antioxidant capacity, methionine metabolism, and hepatocyte autophagy in spotted seabass (Lateolabrax maculatus). A factorial design was used with six methionine levels [0.64, 0.85, 1.11, 1.33, 1.58, and 1.76%] and two temperatures [moderate temperature (MT): 27 ℃, and high temperature (HT): 33 ℃]. The results revealed the significant effects of both dietary methionine level and water temperature on weight gain (WG) and feed efficiency (FE), and their interaction effect was found on WG (P < 0.05). In both water temperatures tested, fish WG increased with increasing methionine level up to 1.11% and decreased thereafter. The groups of fish reared at MT exhibited dramatically higher WG and FE than those kept at HT while an opposite trend was observed for feed intake. Liver antioxidant indices including reduced glutathione and malondialdehyde (MDA) concentrations, and catalase and superoxide dismutase (SOD) activities remarkably increased in the HT group compared to the MT group. Moreover, the lowest MDA concentration and the highest SOD activity were recorded at methionine levels between 1.11% and 0.85%, respectively, regardless of water temperatures. Expression of methionine metabolism-related key enzyme genes (mat2b, cbs, ms, and bhmt) in the liver was increased at moderate methionine levels, and higher expression levels were detected at MT compared to HT with the exception of ms gene relative expression. Relative expression of hepatocyte autophagy-related genes (pink1, atg5, mul1, foxo3) and hsp70 was upregulated by increasing methionine level up to a certain level and decreased thereafter and increasing water temperature led to significantly enhanced expression of hsp70. In summary, HT induced heat stress and reduced fish growth, and an appropriate dietary methionine level improved the antioxidant capacity and stress resistance of fish. A second-order polynomial regression analysis based on the WG suggested that the optimal dietary methionine level for maximum growth of spotted seabass is 1.22% of the diet at 27 ℃ and 1.26% of the diet at 33 ℃, then 1.37 g and 1.68 g dietary methionine intake is required for 100 g weight gain at 27 ℃ or 33 ℃, respectively.
Collapse
Affiliation(s)
- Xin Yang
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, People's Republic of China
| | - Qinjiang Weng
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, People's Republic of China
| | - Xueshan Li
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, People's Republic of China
| | - Kangle Lu
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, People's Republic of China
| | - Ling Wang
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, People's Republic of China
| | - Kai Song
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, People's Republic of China.
| | - Chunxiao Zhang
- Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Fisheries College, Jimei University, Xiamen, 361021, People's Republic of China.
| | - Samad Rahimnejad
- Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, University of South Bohemia in Ceske Budejovice, Zatisi 728/II, 389 25, Vodnany, Czech Republic
| |
Collapse
|
6
|
Nguyen TH, Nguyen TM, Ngoc DTM, You T, Park MK, Lee CH. Unraveling the Janus-Faced Role of Autophagy in Hepatocellular Carcinoma: Implications for Therapeutic Interventions. Int J Mol Sci 2023; 24:16255. [PMID: 38003445 PMCID: PMC10671265 DOI: 10.3390/ijms242216255] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/02/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
This review aims to provide a comprehensive understanding of the molecular mechanisms underlying autophagy and mitophagy in hepatocellular carcinoma (HCC). Autophagy is an essential cellular process in maintaining cell homeostasis. Still, its dysregulation is associated with the development of liver diseases, including HCC, which is one of leading causes of cancer-related death worldwide. We focus on elucidating the dual role of autophagy in HCC, both in tumor initiation and progression, and highlighting the complex nature involved in the disease. In addition, we present a detailed analysis of a small subset of autophagy- and mitophagy-related molecules, revealing their specific functions during tumorigenesis and the progression of HCC cells. By understanding these mechanisms, we aim to provide valuable insights into potential therapeutic strategies to manipulate autophagy effectively. The goal is to improve the therapeutic response of liver cancer cells and overcome drug resistance, providing new avenues for improved treatment options for HCC patients. Overall, this review serves as a valuable resource for researchers and clinicians interested in the complex role of autophagy in HCC and its potential as a target for innovative therapies aimed to combat this devastating disease.
Collapse
Affiliation(s)
- Thi Ha Nguyen
- College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Tuan Minh Nguyen
- College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | | | - Taesik You
- College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| | - Mi Kyung Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy National Cance Center, Goyang 10408, Republic of Korea
- Department of Bio-Healthcare, Hwasung Medi-Science University, Hwaseong-si 18274, Republic of Korea
| | - Chang Hoon Lee
- College of Pharmacy, Dongguk University, Seoul 04620, Republic of Korea
| |
Collapse
|
7
|
Xiao S, Peng K, Li C, Long Y, Yu Q. The role of sphingosine-1-phosphate in autophagy and related disorders. Cell Death Discov 2023; 9:380. [PMID: 37852968 PMCID: PMC10584985 DOI: 10.1038/s41420-023-01681-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/03/2023] [Accepted: 10/12/2023] [Indexed: 10/20/2023] Open
Abstract
S1P, also referred to as sphingosine-1-phosphate, is a lipid molecule with bioactive properties involved in numerous cellular processes such as cell growth, movement, programmed cell death, self-degradation, cell specialization, aging, and immune system reactions. Autophagy is a meticulously controlled mechanism in which cells repurpose their elements to maintain cellular balance. There are five stages in autophagy: initiation, nucleation, elongation and maturation, fusion, and degradation. New research has provided insight into the complex connection between S1P and autophagy, uncovering their interaction in both normal and abnormal circumstances. Gaining knowledge about the regulatory mechanism of S1P signaling on autophagy can offer a valuable understanding of its function in well-being and illness, potentially leading to innovative therapeutic concepts for diverse ailments. Hence, this review analyzes the essential stages in mammalian autophagy, with a specific emphasis on recent research exploring the control of each stage by S1P. Additionally, it sheds light on the roles of S1P-induced autophagy in various disorders.
Collapse
Affiliation(s)
- Siqi Xiao
- Department of Gastroenterology & Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Jiefang Avenue 1095#, Wuhan City, Hubei Province, 430030, P.R. China
| | - Kaixin Peng
- Department of Gastroenterology & Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Jiefang Avenue 1095#, Wuhan City, Hubei Province, 430030, P.R. China
| | - Congxin Li
- Department of Gastroenterology & Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Jiefang Avenue 1095#, Wuhan City, Hubei Province, 430030, P.R. China
| | - Yuanyuan Long
- Department of Gastroenterology & Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Jiefang Avenue 1095#, Wuhan City, Hubei Province, 430030, P.R. China
| | - Qin Yu
- Department of Gastroenterology & Hepatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Jiefang Avenue 1095#, Wuhan City, Hubei Province, 430030, P.R. China.
| |
Collapse
|
8
|
de Wet S, Theart R, Loos B. Cogs in the autophagic machine-equipped to combat dementia-prone neurodegenerative diseases. Front Mol Neurosci 2023; 16:1225227. [PMID: 37720551 PMCID: PMC10500130 DOI: 10.3389/fnmol.2023.1225227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/31/2023] [Indexed: 09/19/2023] Open
Abstract
Neurodegenerative diseases are often characterized by hydrophobic inclusion bodies, and it may be the case that the aggregate-prone proteins that comprise these inclusion bodies are in fact the cause of neurotoxicity. Indeed, the appearance of protein aggregates leads to a proteostatic imbalance that causes various interruptions in physiological cellular processes, including lysosomal and mitochondrial dysfunction, as well as break down in calcium homeostasis. Oftentimes the approach to counteract proteotoxicity is taken to merely upregulate autophagy, measured by an increase in autophagosomes, without a deeper assessment of contributors toward effective turnover through autophagy. There are various ways in which autophagy is regulated ranging from the mammalian target of rapamycin (mTOR) to acetylation status of proteins. Healthy mitochondria and the intracellular energetic charge they preserve are key for the acidification status of lysosomes and thus ensuring effective clearance of components through the autophagy pathway. Both mitochondria and lysosomes have been shown to bear functional protein complexes that aid in the regulation of autophagy. Indeed, it may be the case that minimizing the proteins associated with the respective neurodegenerative pathology may be of greater importance than addressing molecularly their resulting inclusion bodies. It is in this context that this review will dissect the autophagy signaling pathway, its control and the manner in which it is molecularly and functionally connected with the mitochondrial and lysosomal system, as well as provide a summary of the role of autophagy dysfunction in driving neurodegenerative disease as a means to better position the potential of rapamycin-mediated bioactivities to control autophagy favorably.
Collapse
Affiliation(s)
- Sholto de Wet
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Rensu Theart
- Department of Electric and Electronic Engineering, Stellenbosch University, Stellenbosch, South Africa
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
9
|
Srivastava M, Bhukya PL, Barman MK, Bhise N, Lole KS. Modulation of cellular autophagy by genotype 1 hepatitis E virus ORF3 protein. J Gen Virol 2023; 104. [PMID: 36809248 DOI: 10.1099/jgv.0.001824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Hepatitis E virus (HEV) egresses from infected hepatocytes as quasienveloped particles containing open reading frame 3 (ORF3) protein. HEV ORF3 (small phosphoprotein) interacts with host proteins to establish a favourable environment for virus replication. It is a functional viroporin that plays an important role during virus release. Our study provides evidence that pORF3 plays a pivotal role in inducing Beclin1-mediated autophagy that helps HEV-1 replication as well as its exit from cells. The ORF3 interacts with host proteins involved in regulation of transcriptional activity, immune response, cellular and molecular processes, and modulation of autophagy, by interacting with proteins, DAPK1, ATG2B, ATG16L2 and also several histone deacetylases (HDACs). For autophagy induction, the ORF3 utilizes non-canonical NF-κB2 pathway and sequesters p52NF-κB and HDAC2 to upregulate DAPK1 expression, leading to enhanced Beclin1 phosphorylation. By sequestering several HDACs, HEV may prevent histone deacetylation to maintain overall cellular transcription intact to promote cell survival. Our findings highlight a novel crosstalk between cell survival pathways participating in ORF3-mediated autophagy.
Collapse
Affiliation(s)
| | - Prudhvi Lal Bhukya
- Division of Hepatitis, National Institute of Virology, Pune, India
- ICMR-National Animal Resource Facility for Biomedical Research, Hyderabad, India
| | | | - Neha Bhise
- Division of Hepatitis, National Institute of Virology, Pune, India
| | - Kavita S Lole
- Division of Hepatitis, National Institute of Virology, Pune, India
| |
Collapse
|
10
|
Liu B, Zhang Y, Ren H, Yao Q, Ba J, Luan J, Zhao P, Qin Z, Qi Z. mTOR signaling regulates Zika virus replication bidirectionally through autophagy and protein translation. J Med Virol 2023; 95:e28422. [PMID: 36546404 DOI: 10.1002/jmv.28422] [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: 10/07/2022] [Revised: 12/10/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
Zika virus (ZIKV) reemerged in 2016 and attracted much more attention worldwide. To date, the limited knowledge of ZIKV interactions with host cells in the early stages of infection impedes the prevention of viral epidemics and the treatment of ZIKV disease. The mammalian target of rapamycin (mTOR) signaling pathway plays an essential role in the regulation of autophagy and protein synthesis during multiple viral infections. This study aimed to investigate the functional role of mTOR signaling in ZIKV replication in human umbilical vein endothelial cells. Immunoblotting demonstrated that ZIKV infection inhibited mTORC1 signaling, enhancing autophagy but obstructing protein translation. Drugs or siRNA for interfering with mTOR signaling molecules were utilized to demonstrate that AKT/TSC2/mTORC1 signaling was involved in ZIKV infection and that autophagy promoted ZIKV production, but viral protein expression was regulated by mTORC1 signaling. Moreover, confocal microscopy indicated a robust correlation between autophagy and viral RNA transcription. This study clarifies the dual functions of mTOR signaling during ZIKV infection and provides theoretical support for developing potential anti-ZIKV drugs based on mTOR signaling molecules and deeper insights to better understand the mechanism between ZIKV and host cells.
Collapse
Affiliation(s)
- Bin Liu
- Department of Microbiology, Naval Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, China.,Naval Medical Center, Naval Medical University, Shanghai, China
| | - Yahui Zhang
- Department of Cardiology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Hao Ren
- Department of Microbiology, Naval Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, China
| | - Qiufeng Yao
- Department of Microbiology, Naval Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, China
| | - Jianbo Ba
- Naval Medical Center, Naval Medical University, Shanghai, China
| | - Jie Luan
- Naval Medical Center, Naval Medical University, Shanghai, China
| | - Ping Zhao
- Department of Microbiology, Naval Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, China
| | - Zhaoling Qin
- Department of Microbiology, Naval Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, China
| | - Zhongtian Qi
- Department of Microbiology, Naval Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai, China
| |
Collapse
|
11
|
Aguilera MO, Robledo E, Melani M, Wappner P, Colombo MI. FKBP8 is a novel molecule that participates in the regulation of the autophagic pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119212. [PMID: 35090967 DOI: 10.1016/j.bbamcr.2022.119212] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 12/28/2021] [Accepted: 01/01/2022] [Indexed: 06/14/2023]
Abstract
Autophagy is a homeostatic process by which misfolded proteins, organelles and cytoplasmic material are engulfed in autophagosomal vesicles and degraded through a lisosomal pathway. FKBP8 is a member of the FK506-binding proteins family (FKBP) usually found in mitochondria and the endoplasmic reticulum. This protein plays a critical role in cell functions such as protein trafficking and folding. In the present report we demonstrate that the depletion of FKBP8 abrogated autophagy activation induced by starvation, whereas the overexpression of this protein triggered the autophagy cascade. We found that FKBP8 co-localizes with ATG14L and BECN1, both members of the VPS34 lipid kinase complex, which regulates the initial steps in the autophagosome formation process. We have also demonstrated that FKBP8 is necessary for VPS34 activity. Our findings indicate that the regulatory function of FKBP8 in the autophagy process depends of its transmembrane domain. Surprisingly, this protein was not found in autophagosomal vesicles, which reinforces the notion that the FKBP8 only participates in the initial steps of the autophagosome formation process. Taken together, our data provide evidence that FKBP8 modulates the early steps of the autophagosome formation event by interacting with the VPS34 lipid kinase complex. SUMMARY: In this article, the protein FKBP38 is reported to be a novel modulator of the initial steps of the autophagic pathway, specifically in starvation-induced autophagy. FKBP38 interacts with the VPS34 lipid kinase complex, with the transmembrane domain of FKBP38 being critical for its biological function.
Collapse
Affiliation(s)
- Milton Osmar Aguilera
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina; Laboratorio de Mecanismos Moleculares Implicados en el Tráfico Vesicular y la Autofagia, Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina; Microbiología, Parasitología e Inmunología, Facultad de Odontología, Universidad Nacional de Cuyo, Mendoza, Argentina.
| | - Esteban Robledo
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina; Laboratorio de Mecanismos Moleculares Implicados en el Tráfico Vesicular y la Autofagia, Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina
| | - Mariana Melani
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina; Instituto Leloir, Buenos Aires, Argentina; Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pablo Wappner
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina; Instituto Leloir, Buenos Aires, Argentina
| | - María Isabel Colombo
- Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Buenos Aires, Argentina; Laboratorio de Mecanismos Moleculares Implicados en el Tráfico Vesicular y la Autofagia, Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina.
| |
Collapse
|
12
|
Mondarte EAQ, Zamarripa EMM, Chang R, Wang F, Song S, Tahara H, Hayashi T. Interphase Protein Layers Formed on Self-Assembled Monolayers in Crowded Biological Environments: Analysis by Surface Force and Quartz Crystal Microbalance Measurements. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:1324-1333. [PMID: 35029393 DOI: 10.1021/acs.langmuir.1c02312] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
We investigated a viscous protein layer formed on self-assembled monolayers (SAMs) in crowded biological environments. The results were obtained through force spectroscopic measurements using colloidal probes and substantiated by exhaustive analysis using a quartz crystal microbalance with an energy dissipation technique. A hydrophobic SAM of n-octanethiol (C8 SAM) in bovine serum albumin (BSA) solution is buried under an adlayer of denatured BSA molecules and an additional viscous interphase layer that is five times more viscous than the bulk solution. C8 SAMs in fetal bovine serum induced a formation of a thicker adsorbed protein layer but with no observable viscous interphase layer. These findings show that a fouling surface is essentially inaccessible to any approaching molecules and thus has a new biological and physical identity arising from its surrounding protein layers. In contrast, the SAMs composed of sulfobetaine-terminated alkanethiol proved to be sufficiently protein-resistant and bio-inert even under crowded conditions due to a protective barrier of its interfacial water, which has implications in the accurate targeting of artificial particles for drug delivery and similar applications by screening any non-specific interactions. Finally, our strategies provide a platform for the straightforward yet effectual in vitro characterization of diverse types of surfaces in the context of targeted interactions in crowded biological environments.
Collapse
Affiliation(s)
- Evan Angelo Quimada Mondarte
- Tokyo Institute of Technology, Department of Materials Science and Engineering, School of Materials and Chemical Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8502, Japan
| | - Elisa Margarita Mendoza Zamarripa
- Tokyo Institute of Technology, Department of Materials Science and Engineering, School of Materials and Chemical Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8502, Japan
| | - Ryongsok Chang
- Tokyo Institute of Technology, Department of Materials Science and Engineering, School of Materials and Chemical Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8502, Japan
| | - Fan Wang
- Tokyo Institute of Technology, Department of Materials Science and Engineering, School of Materials and Chemical Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8502, Japan
| | - Subin Song
- Tokyo Institute of Technology, Department of Materials Science and Engineering, School of Materials and Chemical Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8502, Japan
| | - Hiroyuki Tahara
- Tokyo Institute of Technology, Department of Materials Science and Engineering, School of Materials and Chemical Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8502, Japan
| | - Tomohiro Hayashi
- Tokyo Institute of Technology, Department of Materials Science and Engineering, School of Materials and Chemical Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8502, Japan
- The Institute for Solid State Physics, The University of Tokyo, 5-1-5, Kashiwanoha, Kashiwa, Chiba 277-0882, Japan
| |
Collapse
|
13
|
Meyer N, Henkel L, Linder B, Zielke S, Tascher G, Trautmann S, Geisslinger G, Münch C, Fulda S, Tegeder I, Kögel D. Autophagy activation, lipotoxicity and lysosomal membrane permeabilization synergize to promote pimozide- and loperamide-induced glioma cell death. Autophagy 2021; 17:3424-3443. [PMID: 33461384 PMCID: PMC8632287 DOI: 10.1080/15548627.2021.1874208] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/06/2021] [Indexed: 12/22/2022] Open
Abstract
Increasing evidence suggests that induction of lethal macroautophagy/autophagy carries potential significance for the treatment of glioblastoma (GBM). In continuation of previous work, we demonstrate that pimozide and loperamide trigger an ATG5- and ATG7 (autophagy related 5 and 7)-dependent type of cell death that is significantly reduced with cathepsin inhibitors and the lipid reactive oxygen species (ROS) scavenger α-tocopherol in MZ-54 GBM cells. Global proteomic analysis after treatment with both drugs also revealed an increase of proteins related to lipid and cholesterol metabolic processes. These changes were accompanied by a massive accumulation of cholesterol and other lipids in the lysosomal compartment, indicative of impaired lipid transport/degradation. In line with these observations, pimozide and loperamide treatment were associated with a pronounced increase of bioactive sphingolipids including ceramides, glucosylceramides and sphingoid bases measured by targeted lipidomic analysis. Furthermore, pimozide and loperamide inhibited the activity of SMPD1/ASM (sphingomyelin phosphodiesterase 1) and promoted induction of lysosomal membrane permeabilization (LMP), as well as release of CTSB (cathepsin B) into the cytosol in MZ-54 wild-type (WT) cells. Whereas LMP and cell death were significantly attenuated in ATG5 and ATG7 knockout (KO) cells, both events were enhanced by depletion of the lysophagy receptor VCP (valosin containing protein), supporting a pro-survival function of lysophagy under these conditions. Collectively, our data suggest that pimozide and loperamide-driven autophagy and lipotoxicity synergize to induce LMP and cell death. The results also support the notion that simultaneous overactivation of autophagy and induction of LMP represents a promising approach for the treatment of GBM.Abbreviations: ACD: autophagic cell death; AKT1: AKT serine/threonine kinase 1; ATG5: autophagy related 5; ATG7: autophagy related 7; ATG14: autophagy related 14; CERS1: ceramide synthase 1; CTSB: cathepsin B; CYBB/NOX2: cytochrome b-245 beta chain; ER: endoplasmatic reticulum; FBS: fetal bovine serum; GBM: glioblastoma; GO: gene ontology; HTR7/5-HT7: 5-hydroxytryptamine receptor 7; KD: knockdown; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LAP: LC3-associated phagocytosis; LMP: lysosomal membrane permeabilization; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; MTOR: mechanistic target of rapamycin kinase; RB1CC1: RB1 inducible coiled-coil 1; ROS: reactive oxygen species; RPS6: ribosomal protein S6; SMPD1/ASM: sphingomyelin phosphodiesterase 1; VCP/p97: valosin containing protein; WT: wild-type.
Collapse
Affiliation(s)
- Nina Meyer
- Experimental Neurosurgery, Goethe University Hospital Frankfurt/Main, Frankfurt, Germany
| | - Lisa Henkel
- Experimental Neurosurgery, Goethe University Hospital Frankfurt/Main, Frankfurt, Germany
| | - Benedikt Linder
- Experimental Neurosurgery, Goethe University Hospital Frankfurt/Main, Frankfurt, Germany
| | - Svenja Zielke
- Experimental Cancer Research in Pediatrics, Goethe University Hospital Frankfurt/Main, Frankfurt, Germany
| | - Georg Tascher
- Institute of Biochemistry II, Goethe University Hospital Frankfurt/Main, Frankfurt, Germany
| | - Sandra Trautmann
- Institute of Clinical Pharmacology, Goethe University Hospital Frankfurt/Main, Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Goethe University Hospital Frankfurt/Main, Frankfurt, Germany
| | - Christian Münch
- Institute of Biochemistry II, Goethe University Hospital Frankfurt/Main, Frankfurt, Germany
| | - Simone Fulda
- Experimental Cancer Research in Pediatrics, Goethe University Hospital Frankfurt/Main, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe University Hospital Frankfurt/Main, Frankfurt, Germany
| | - Donat Kögel
- Experimental Neurosurgery, Goethe University Hospital Frankfurt/Main, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
| |
Collapse
|
14
|
Cooper-Knock J, Harvey C, Zhang S, Moll T, Timpanaro IS, Kenna KP, Iacoangeli A, Veldink JH. Advances in the genetic classification of amyotrophic lateral sclerosis. Curr Opin Neurol 2021; 34:756-764. [PMID: 34343141 PMCID: PMC7612116 DOI: 10.1097/wco.0000000000000986] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Amyotrophic lateral sclerosis (ALS) is an archetypal complex disease wherein disease risk and severity are, for the majority of patients, the product of interaction between multiple genetic and environmental factors. We are in a period of unprecedented discovery with new large-scale genome-wide association study (GWAS) and accelerating discovery of risk genes. However, much of the observed heritability of ALS is undiscovered and we are not yet approaching elucidation of the total genetic architecture, which will be necessary for comprehensive disease subclassification. RECENT FINDINGS We summarize recent developments and discuss the future. New machine learning models will help to address nonlinear genetic interactions. Statistical power for genetic discovery may be boosted by reducing the search-space using cell-specific epigenetic profiles and expanding our scope to include genetically correlated phenotypes. Structural variation, somatic heterogeneity and consideration of environmental modifiers represent significant challenges which will require integration of multiple technologies and a multidisciplinary approach, including clinicians, geneticists and pathologists. SUMMARY The move away from fully penetrant Mendelian risk genes necessitates new experimental designs and new standards for validation. The challenges are significant, but the potential reward for successful disease subclassification is large-scale and effective personalized medicine.
Collapse
Affiliation(s)
- Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Calum Harvey
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Sai Zhang
- Department of Genetics
- Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Tobias Moll
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Ilia Sarah Timpanaro
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Kevin P Kenna
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Alfredo Iacoangeli
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London
- National Institute for Health Research Biomedical Research Centre and Dementia Unit, South London and Maudsley NHS Foundation Trust and King's College London, London, UK
| | - Jan H Veldink
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
15
|
Davuluri GVN, Chen CC, Chiu YC, Tsai HW, Chiu HC, Chen YL, Tsai PJ, Kuo WT, Tsao N, Lin YS, Chang CP. Autophagy Drives Galectin-1 Secretion From Tumor-Associated Macrophages Facilitating Hepatocellular Carcinoma Progression. Front Cell Dev Biol 2021; 9:741820. [PMID: 34552935 PMCID: PMC8450461 DOI: 10.3389/fcell.2021.741820] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
Galectin-1 (Gal-1) is a secretory lectin with pro-tumor activities and is associated strongly with hepatocellular carcinoma (HCC) development. Although Gal-1 is a well-known soluble pro-tumor factor in the tumor microenvironment (TME), the secretion mode of Gal-1 is not clearly defined. On the other hand, in addition to cancer cells, Gal-1 is widely expressed in tumor stromal cells, including tumor-associated macrophages (TAMs). TAMs are a significant component of stromal cells in TME; however, their contributions in producing Gal-1 to TME are still not explored. Here we reveal that TAMs can actively secrete Gal-1 in response to stimuli of HCC cells. Gal-1 produced by TAMs leads to an increase of the systemic level of Gal-1 and HCC tumor growth in mice. Mechanistically, TLR2-dependent secretory autophagy is found to be responsible for Gal-1 secretion from TAMs. Gal-1 acts as a cargo of autophagosomes to fuse with multivesicular bodies via Rab11 and VAMP7-mediated vesicle trafficking before being secreted. This autophagy-regulated Gal-1 secretion in TAMs correlates to poor overall survival and progression-free survival rates of HCC patients. Our findings uncover the secretion mode of Gal-1 via secretory autophagy and highlight the pathological role of TAM-produced Gal-1 in HCC progression.
Collapse
Affiliation(s)
| | - Chien-Chin Chen
- Department of Pathology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, Taiwan.,Department of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Yen-Cheng Chiu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hung-Wen Tsai
- Department of Pathology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hung-Chih Chiu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yuh-Ling Chen
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Jane Tsai
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Wan-Ting Kuo
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Nina Tsao
- Department of Medical Laboratory Science, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Yee-Shin Lin
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Peng Chang
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
16
|
Mukai H, Takanashi M, Ogawara KI, Maruyama M, Higaki K. Possible Regulation of P-glycoprotein Function by Adrenergic Agonists in a Vascular-luminal Perfused Preparation of Small Intestine. J Pharm Sci 2021; 110:3889-3895. [PMID: 34530005 DOI: 10.1016/j.xphs.2021.09.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 11/28/2022]
Abstract
Although the functions of small intestine are largely regulated by enteric nervous system (ENS), an independent intrinsic innervation, as well as central nervous system (CNS), the neural regulation of drug absorption from the small intestine still remains to be clarified. To obtain some information on it, the effect of adrenergic agonists on P-glycoprotein (P-gp) function was investigated by utilizing a vascular-luminal perfused rat small intestine. Adrenaline significantly decreased the secretion of rhodamine-123 (R-123) into the intestinal lumen, but dibutyryl cAMP (DBcAMP) significantly enhanced R-123 secretion. The inhibition study with quinidine clearly indicated that the decrease in secretory clearance of R-123 by adrenaline or the increase by DBcAMP would be attributed to the decrease or increase in P-gp activity, respectively. Expression levels of P-gp in whole mucosal homogenates were not changed at all by any chemicals examined, but those on brush border membrane (BBM) of intestinal epithelial cells were significantly decreased or increased by adrenaline or DBcAMP, respectively. Furthermore, changes in P-gp activity caused by adrenergic agonists and DBcAMP were significantly correlated with changes in expression level of P-gp in BBM, suggesting that the trafficking of P-gp from cytosolic pool to BBM would be regulated by adrenergic agonists and DBcAMP.
Collapse
Affiliation(s)
- Hironori Mukai
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan; Production Department, Odawara Central Factory, Nippon Shinyaku Co., Ltd., 676-1 Kuwahara, Odawara, Kanagawa 250-0861, Japan
| | - Masashi Takanashi
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan; Central Hyogo Area, Hanshin Dispensing Pharmacy, I & H Co., Ltd., 1-18 Ohmasu-cho, Ashiya, Hyogo 659-0066, Japan
| | - Ken-Ichi Ogawara
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan; Laboratory of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1, Motoyamakita, Higashinada-ku, Kobe, Hyogo 658-8558, Japan
| | - Masato Maruyama
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Kazutaka Higaki
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan.
| |
Collapse
|
17
|
Begum S, Moreau F, Dufour A, Chadee K. Entamoeba histolytica exploits the autophagy pathway in macrophages to trigger inflammation in disease pathogenesis. Mucosal Immunol 2021; 14:1038-1054. [PMID: 33963264 DOI: 10.1038/s41385-021-00408-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/25/2021] [Accepted: 04/16/2021] [Indexed: 02/04/2023]
Abstract
The mechanism whereby Entamoeba histolytica (Eh) binding with macrophages at the intercellular junction triggers aggressive pro-inflammatory responses in disease pathogenesis is not well understood. The host intracellular protein degradation process autophagy and its regulatory proteins are involved in maintenance of cellular homeostasis and excessive inflammatory responses. In this study we unraveled how Eh hijacks the autophagy process in macrophages to dysregulate pro-inflammatory responses. Direct contact of live Eh with macrophages activated caspase-6 that induced rapid proteolytic degradation of the autophagy ATG16L1 protein complex independent of NLRP3 inflammasome and caspase-3/8 activation. Crohn's disease susceptible ATG16L1 T300A variant was highly susceptible to Eh-mediated degradation that augmented pro-inflammatory cytokines in mice. Quantitative proteomics revealed downregulation of autophagy and vesicle-mediated transport and upregulation of cysteine-type endopeptidase pathways in response to Eh. We conclude during Eh-macrophage outside-in signaling, ATG16L1 protein complex plays an overlooked regulatory role in shaping the pro-inflammatory landscape in amebiasis.
Collapse
Affiliation(s)
- Sharmin Begum
- Departments of Microbiology, Immunology and Infectious Diseases, Calgary, AB, Canada
| | - France Moreau
- Departments of Microbiology, Immunology and Infectious Diseases, Calgary, AB, Canada
| | - Antoine Dufour
- Physiology and Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.,Biochemistry and Molecular Biology, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Kris Chadee
- Departments of Microbiology, Immunology and Infectious Diseases, Calgary, AB, Canada.
| |
Collapse
|
18
|
Cosio C, Degli-Esposti D, Almunia C, Gaillet V, Sartelet H, Armengaud J, Chaumot A, Geffard O, Geffard A. Subcellular Distribution of Dietary Methyl-Mercury in Gammarus fossarum and Its Impact on the Amphipod Proteome. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2021; 55:10514-10523. [PMID: 34283579 DOI: 10.1021/acs.est.1c02385] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The transfer of methyl-Hg (MeHg) from food is central for its effects in aquatic animals, but we still lack knowledge concerning its impact on invertebrate primary consumers. In aquatic environments, cell walls of plants are particularly recalcitrant to degradation and as such remain available as a food source for long periods. Here, the impact at the proteomic level of dietary MeHg in Gammarus fossarum was established and linked to subcellular distribution of Hg. Individuals of G. fossarum were fed with MeHg in cell wall or intracellular compartments of Elodea nuttallii. Hg concentrations in subcellular fractions were 2 to 6 times higher in animals fed with cell wall than intracellular compartments. At the higher concentrations tested, the proportion of Hg in metal-sensitive fraction increased from 30.0 ± 6.1 to 41.0 ± 5.7% for individuals fed with intracellular compartment, while biologically detoxified metal fraction increased from 30.0 ± 6.1 to 50.0 ± 2.8% when fed with cell wall compartment. Data suggested that several thresholds of proteomic response are triggered by increased bioaccumulation in each subcellular fraction in correlation with Hg exclusively bound to the metal-sensitive fraction, while the increase of biologically detoxified metal likely had a cost for fitness. Proteomics analysis supported that the different binding sites and speciation in shoots subsequently resulted in different fate and cellular toxicity pathways to consumers. Our data confirmed that Hg bound in cell walls of plants can be assimilated by G. fossarum, which is consistent with its feeding strategy, hence pointing cell walls as a significant source for Hg transfers and toxicity in primary consumers. The high accumulation of Hg in macrophytes makes them a risk for food web transfer in shallow ecosystems. The present results allowed gaining new insights into the effects and uptake mechanisms of MeHg in aquatic primary consumers.
Collapse
Affiliation(s)
- Claudia Cosio
- Université de Reims Champagne-Ardenne, UMR-I 02 INERIS-URCA-ULH SEBIO, Campus du Moulin de la Housse, BP 1039, Cedex, Reims 51687, France
| | | | - Christine Almunia
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, 30200 Bagnols-sur-Cèze, France
| | - Véronique Gaillet
- Université de Reims Champagne-Ardenne, UMR-I 02 INERIS-URCA-ULH SEBIO, Campus du Moulin de la Housse, BP 1039, Cedex, Reims 51687, France
| | - Hervé Sartelet
- Université de Reims Champagne-Ardenne, UMR CNRS/URCA 7369, Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Campus du Moulin de la Housse, BP 1039, Cedex, Reims 51687, France
| | - Jean Armengaud
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, 30200 Bagnols-sur-Cèze, France
| | - Arnaud Chaumot
- INRAE, UR RiverLy, Laboratoire d'écotoxicologie, F-69625 Villeurbanne, France
| | - Olivier Geffard
- INRAE, UR RiverLy, Laboratoire d'écotoxicologie, F-69625 Villeurbanne, France
| | - Alain Geffard
- Université de Reims Champagne-Ardenne, UMR-I 02 INERIS-URCA-ULH SEBIO, Campus du Moulin de la Housse, BP 1039, Cedex, Reims 51687, France
| |
Collapse
|
19
|
Wang X, Guo G, Zhang J, Aebez N, Liu Z, Liu CF, Ross CA, Smith WW. Mutant-TMEM230-induced neurodegeneration and impaired axonal mitochondrial transport. Hum Mol Genet 2021; 30:1535-1542. [PMID: 34002226 DOI: 10.1093/hmg/ddab128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease with movement disorders including resting tremor, rigidity, bradykinesia and postural instability. Recent studies have identified a new PD associated gene, TMEM230 (transmembrane protein 230). However, the pathological roles of TMEM230 and its variants are not fully understood. TMEM230 gene encodes two protein isoforms. Isoform2 is the major protein form (~95%) in human. In this study, we overexpress isoform2 TMEM230 variants (WT or PD-linked *184Wext*5 mutant) or knockdown endogenous protein in cultured SH-5Y5Y cells and mouse primary hippocampus neurons to study their pathological roles. We found that overexpression of WT and mutant TMEM230 or knockdown of endogenous TMEM230-induced neurodegeneration and impaired mitochondria transport at the retrograde direction in axons. Mutant TMEM230 caused more severe neurotoxicity and mitochondrial transport impairment than WT-TMEM230 did. Our results demonstrate that maintaining TMEM230 protein levels is critical for neuron survival and axon transport. These findings suggest that mutant-TMEM230-induced mitochondrial transport impairment could be the early event leading to neurite injury and neurodegeneration in PD development.
Collapse
Affiliation(s)
- Xiaobo Wang
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Institute of Neuroscience, Soochow University School of Medicine, Suzhou, Jiangsu 215123, China
| | - Gongbo Guo
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jinru Zhang
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Nicolas Aebez
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Zhaohui Liu
- Department of Human Anatomy and Cytoneurobiology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chun-Feng Liu
- Institute of Neuroscience, Soochow University School of Medicine, Suzhou, Jiangsu 215123, China.,Department of Neurology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Christopher A Ross
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Wanli W Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
20
|
Kim YS, Nam HJ, Han CY, Joo MS, Jang K, Jun DW, Kim SG. Liver X Receptor Alpha Activation Inhibits Autophagy and Lipophagy in Hepatocytes by Dysregulating Autophagy-Related 4B Cysteine Peptidase and Rab-8B, Reducing Mitochondrial Fuel Oxidation. Hepatology 2021; 73:1307-1326. [PMID: 32557804 DOI: 10.1002/hep.31423] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Fat accumulation results from increased fat absorption and/or defective fat metabolism. Currently, the lipid-sensing nuclear receptor that controls fat utilization in hepatocytes is elusive. Liver X receptor alpha (LXRα) promotes accumulation of lipids through the induction of several lipogenic genes. However, its effect on lipid degradation is open for study. Here, we investigated the inhibitory role of LXRα in autophagy/lipophagy in hepatocytes and the underlying basis. APPROACH AND RESULTS In LXRα knockout mice fed a high-fat diet, or cell models, LXRα activation suppressed the function of mitochondria by inhibiting autophagy/lipophagy and induced hepatic steatosis. Gene sets associated with "autophagy" were enriched in hepatic transcriptome data. Autophagy flux was markedly augmented in the LXRα knockout mouse liver and primary hepatocytes. Mechanistically, LXRα suppressed autophagy-related 4B cysteine peptidase (ATG4B) and Rab-8B, responsible for autophagosome and -lysosome formation, by inducing let-7a and microRNA (miR)-34a. Chromatin immunoprecipitation assay enabled us to find LXRα as a transcription factor of let-7a and miR-34a. Moreover, 3' untranslated region luciferase assay substantiated the direct inhibitory effects of let-7a and miR-34a on ATG4B and Rab-8B. Consistently, either LXRα activation or the let-7a/miR-34a transfection lowered mitochondrial oxygen consumption rate and mitochondrial transmembrane potential and increased fat levels. In obese animals or nonalcoholic fatty liver disease (NAFLD) patients, let-7a and miR-34a levels were elevated with simultaneous decreases in ATG4B and Rab-8B levels. CONCLUSIONS LXRα inhibits autophagy in hepatocytes through down-regulating ATG4B and Rab-8B by transcriptionally activating microRNA let-7a-2 and microRNA 34a genes and suppresses mitochondrial biogenesis and fuel consumption. This highlights a function of LXRα that culminates in the progression of liver steatosis and steatohepatitis, and the identified targets may be applied for a therapeutic strategy in the treatment of NAFLD.
Collapse
Affiliation(s)
- Yun Seok Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Hyeon Joo Nam
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Chang Yeob Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea.,School of Pharmacy, Jeonbuk National University, Jeonju-si, Jeonbuk, Korea
| | - Min Sung Joo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Kiseok Jang
- Department of Pathology, Hanyang University School of Medicine, Seoul, Korea
| | - Dae Won Jun
- Internal Medicine, Hanyang University School of Medicine, Seoul, Korea
| | - Sang Geon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
21
|
Straka T, Schröder C, Roos A, Kollipara L, Sickmann A, Williams MPI, Hafner M, Khan MM, Rudolf R. Regulatory Function of Sympathetic Innervation on the Endo/Lysosomal Trafficking of Acetylcholine Receptor. Front Physiol 2021; 12:626707. [PMID: 33776791 PMCID: PMC7991846 DOI: 10.3389/fphys.2021.626707] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/08/2021] [Indexed: 01/02/2023] Open
Abstract
Recent studies have demonstrated that neuromuscular junctions are co-innervated by sympathetic neurons. This co-innervation has been shown to be crucial for neuromuscular junction morphology and functional maintenance. To improve our understanding of how sympathetic innervation affects nerve–muscle synapse homeostasis, we here used in vivo imaging, proteomic, biochemical, and microscopic approaches to compare normal and sympathectomized mouse hindlimb muscles. Live confocal microscopy revealed reduced fiber diameters, enhanced acetylcholine receptor turnover, and increased amounts of endo/lysosomal acetylcholine-receptor-bearing vesicles. Proteomics analysis of sympathectomized skeletal muscles showed that besides massive changes in mitochondrial, sarcomeric, and ribosomal proteins, the relative abundance of vesicular trafficking markers was affected by sympathectomy. Immunofluorescence and Western blot approaches corroborated these findings and, in addition, suggested local upregulation and enrichment of endo/lysosomal progression and autophagy markers, Rab 7 and p62, at the sarcomeric regions of muscle fibers and neuromuscular junctions. In summary, these data give novel insights into the relevance of sympathetic innervation for the homeostasis of muscle and neuromuscular junctions. They are consistent with an upregulation of endocytic and autophagic trafficking at the whole muscle level and at the neuromuscular junction.
Collapse
Affiliation(s)
- Tatjana Straka
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, Mannheim, Germany.,Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Charlotte Schröder
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
| | - Andreas Roos
- Department of Neuropediatrics, University Hospital Essen, Essen, Germany.,Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | | | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany.,Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, United Kingdom.,Medizinische Fakultät, Medizinische Proteom-Center (MPC), Ruhr-Universität Bochum, Bochum, Germany
| | | | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, Mannheim, Germany
| | - Muzamil Majid Khan
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, Mannheim, Germany.,Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, Mannheim, Germany.,Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany.,Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
22
|
Robert G, Yagyu M, Koizumi T, Naya L, Masclaux-Daubresse C, Yoshimoto K. Ammonium stress increases microautophagic activity while impairing macroautophagic flux in Arabidopsis roots. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2021; 105:1083-1097. [PMID: 33222335 DOI: 10.1111/tpj.15091] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 06/11/2023]
Abstract
Plant responses to NH4+ stress are complex, and multiple mechanisms underlying NH4+ sensitivity and tolerance in plants may be involved. Here, we demonstrate that macro- and microautophagic activities are oppositely affected in plants grown under NH4+ toxicity conditions. When grown under NH4+ stress conditions, macroautophagic activity was impaired in roots. Root cells accumulated autophagosomes in the cytoplasm, but showed less autophagic flux, indicating that late steps of the macroautophagy process are affected under NH4+ stress conditions. Under this scenario, we also found that the CCZ1-MON1 complex, a critical factor for vacuole delivery pathways, functions in the late step of the macroautophagic pathway in Arabidopsis. In contrast, an accumulation of tonoplast-derived vesicles was observed in vacuolar lumens of root cells of NH4+ -stressed plants, suggesting the induction of a microautophagy-like process. In this sense, some SYP22-, but mainly VAMP711-positive vesicles were observed inside vacuole in roots of NH4+ -stressed plants. Consistent with the increased tonoplast degradation and the reduced membrane flow to the vacuole due to the impaired macroautophagic flux, the vacuoles of root cells of NH4+ -stressed plants showed a simplified structure and lower tonoplast content. Taken together, this study presents evidence that postulates late steps of the macroautophagic process as a relevant physiological mechanism underlying the NH4+ sensitivity response in Arabidopsis, and additionally provides insights into the molecular tools for studying microautophagy in plants.
Collapse
Affiliation(s)
- Germán Robert
- Institut Jean-Pierre Bourgin, INRAE, AgroParisTech, Université Paris-Saclay, Versailles, 78000, France
- Instituto Nacional de Tecnología Agropecuaria (INTA) - Instituto de Fisiología y Recursos Genéticos Vegetales (IFRGV), Av. 11 de Septiembre, Córdoba, 4755-X5020ICA, Argentina
- Unidad de doble dependencia INTA-CONICET (UDEA), Av. 11 de Septiembre, Córdoba, 4755-X5020ICA, Argentina
| | - Mako Yagyu
- Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1, Higashimita, Tama-ku, Kawasaki-shi, Kanagawa, 214-8571, Japan
| | - Takaya Koizumi
- Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1, Higashimita, Tama-ku, Kawasaki-shi, Kanagawa, 214-8571, Japan
| | - Loreto Naya
- Institut Jean-Pierre Bourgin, INRAE, AgroParisTech, Université Paris-Saclay, Versailles, 78000, France
| | - Céline Masclaux-Daubresse
- Institut Jean-Pierre Bourgin, INRAE, AgroParisTech, Université Paris-Saclay, Versailles, 78000, France
| | - Kohki Yoshimoto
- Institut Jean-Pierre Bourgin, INRAE, AgroParisTech, Université Paris-Saclay, Versailles, 78000, France
- Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1, Higashimita, Tama-ku, Kawasaki-shi, Kanagawa, 214-8571, Japan
| |
Collapse
|
23
|
Yamazaki T, Bravo-San Pedro JM, Galluzzi L, Kroemer G, Pietrocola F. Autophagy in the cancer-immunity dialogue. Adv Drug Deliv Rev 2021; 169:40-50. [PMID: 33301821 DOI: 10.1016/j.addr.2020.12.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/24/2020] [Accepted: 12/04/2020] [Indexed: 02/08/2023]
Abstract
Autophagy is quintessential for the maintenance of cellular homeostasis in all eukaryotic cells, explaining why both normal and malignant cells benefit from proficient autophagic responses. Moreover, autophagy is intimately involved in the immunological control of malignant transformation, tumor progression and response to therapy. However, the net effect of autophagy activation or inhibition on the natural growth or therapeutic response of tumors evolving in immunocompetent hosts exhibits a considerable degree of context dependency. Here, we discuss the complex cross-talk between autophagy and immuno-oncology as delineated by genetic and pharmacological approaches in mouse models of cancer.
Collapse
|
24
|
Biogenesis of Fungal Extracellular Vesicles: What Do We Know? Curr Top Microbiol Immunol 2021; 432:1-11. [DOI: 10.1007/978-3-030-83391-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
25
|
Broggi G, Ieni A, Russo D, Varricchio S, Puzzo L, Russo A, Reibaldi M, Longo A, Tuccari G, Staibano S, Caltabiano R. The Macro-Autophagy-Related Protein Beclin-1 Immunohistochemical Expression Correlates With Tumor Cell Type and Clinical Behavior of Uveal Melanoma. Front Oncol 2020; 10:589849. [PMID: 33330070 PMCID: PMC7714947 DOI: 10.3389/fonc.2020.589849] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/20/2020] [Indexed: 01/01/2023] Open
Abstract
Uveal melanoma, in spite of its rarity, represents the most common primitive intraocular malignant neoplasm of the adults; it affects choroid, ciliary bodied and iris and remains clinically silent for a long time, being accidentally discovered by routine ophthalmic exams. Prognosis of uveal melanoma is poor and frequently characterized by liver metastases, within 10-15 years from diagnosis. Autophagy is a multi-step catabolic process by which cells remove damaged organelles and proteins and recycle nutrients. It has been hypothesized that in early stages of tumorigenesis autophagy has a tumor suppressor role while, in more advanced stages, it may represent a survival mechanism of neoplastic cells in response to stress. Several proteins related to autophagy cascade have been investigated in numerous subtypes of human cancer, with overall controversal results. In this paper we studied the immunohistochemical expression of 3 autophagy related proteins (Beclin-1, p62 and ATG7) in a cohort of 85 primary uveal melanoma treated by primary enucleation (39 with metastasis and 46 non metastatic) and correlated their expression with clinico-pathological parameters and blood vascular microvessel density, in order to investigate the potential prognostic role of autophagy in this rare neoplasm. We found that high immunohistochemical levels of Beclin-1 correlated with a lower risk of metastasis and higher disease-free survival times, indicating a positive prognostic role for Beclin-1 in uveal melanoma. No statistically significative differences regarding the expression of ATG7 and p62 between metastatic and non metastatic patients was detected.
Collapse
Affiliation(s)
- Giuseppe Broggi
- Section of Anatomic Pathology, Department Gian Filippo Ingrassia, University of Catania, Catania, Italy
| | - Antonio Ieni
- Section of Pathology, Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - Daniela Russo
- Pathology Unit, Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Silvia Varricchio
- Pathology Unit, Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Lidia Puzzo
- Section of Anatomic Pathology, Department Gian Filippo Ingrassia, University of Catania, Catania, Italy
| | - Andrea Russo
- Department of Ophthalmology, University of Catania, Catania, Italy
| | - Michele Reibaldi
- Department of Ophthalmology, University of Catania, Catania, Italy.,Department of Surgical Science, Eye Clinic, University of Torino, Torino, Italy
| | - Antonio Longo
- Department of Ophthalmology, University of Catania, Catania, Italy
| | - Giovanni Tuccari
- Section of Pathology, Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - Stefania Staibano
- Pathology Unit, Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Rosario Caltabiano
- Section of Anatomic Pathology, Department Gian Filippo Ingrassia, University of Catania, Catania, Italy
| |
Collapse
|
26
|
Atwood DJ, Brown CN, Holditch SJ, Pokhrel D, Thorburn A, Hopp K, Edelstein CL. The effect of trehalose on autophagy-related proteins and cyst growth in a hypomorphic Pkd1 mouse model of autosomal dominant polycystic kidney disease. Cell Signal 2020; 75:109760. [DOI: 10.1016/j.cellsig.2020.109760] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/27/2022]
|
27
|
Roy M, Roux S. Rab GTPases in Osteoclastic Bone Resorption and Autophagy. Int J Mol Sci 2020; 21:ijms21207655. [PMID: 33081155 PMCID: PMC7589333 DOI: 10.3390/ijms21207655] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/11/2020] [Accepted: 10/13/2020] [Indexed: 12/17/2022] Open
Abstract
Small guanosine triphosphate hydrolases (GTPases) of the Rab family are involved in plasma membrane delivery, fusion events, and lysosomal and autophagic degradation pathways, thereby regulating signaling pathways and cell differentiation and function. Osteoclasts are bone-resorbing cells that maintain bone homeostasis. Polarized vesicular trafficking pathways result in the formation of the ruffled border, the osteoclast’s resorptive organelle, which also assists in transcytosis. Here, we reviewed the different roles of Rab GTPases in the endomembrane machinery of osteoclasts and in bone diseases caused by the dysfunction of these proteins, with a particular focus on autophagy and bone resorption. Understanding the molecular mechanisms underlying osteoclast-related bone disease development is critical for developing and improving therapies.
Collapse
|
28
|
Wang J, Yeckel G, Kandoth PK, Wasala L, Hussey RS, Davis EL, Baum TJ, Mitchum MG. Targeted suppression of soybean BAG6-induced cell death in yeast by soybean cyst nematode effectors. MOLECULAR PLANT PATHOLOGY 2020; 21:1227-1239. [PMID: 32686295 PMCID: PMC7411569 DOI: 10.1111/mpp.12970] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 05/29/2023]
Abstract
While numerous effectors that suppress plant immunity have been identified from bacteria, fungi, and oomycete pathogens, relatively little is known for nematode effectors. Several dozen effectors have been reported from the soybean cyst nematode (SCN). Previous studies suggest that a hypersensitive response-like programmed cell death is triggered at nematode feeding sites in soybean during an incompatible interaction. However, virulent SCN populations overcome this incompatibility using unknown mechanisms. A soybean BAG6 (Bcl-2 associated anthanogene 6) gene previously reported by us to be highly up-regulated in degenerating feeding sites induced by SCN in a resistant soybean line was attenuated in response to a virulent SCN population. We show that GmBAG6-1 induces cell death in yeast like its Arabidopsis homolog AtBAG6 and also in soybean. This led us to hypothesize that virulent SCN may target GmBAG6-1 as part of their strategy to overcome soybean defence responses during infection. Thus, we used a yeast viability assay to screen SCN effector candidates for their ability to specifically suppress GmBAG6-1-induced cell death. We identified several effectors that strongly suppressed cell death mediated by GmBAG6-1. Two effectors identified as suppressors showed direct interaction with GmBAG6-1 in yeast, suggesting that one mechanism of cell death suppression may occur through an interaction with this host protein.
Collapse
Affiliation(s)
- Jianying Wang
- Division of Plant Sciences and Bond Life Sciences CenterUniversity of MissouriColumbiaMOUSA
| | - Greg Yeckel
- Division of Plant Sciences and Bond Life Sciences CenterUniversity of MissouriColumbiaMOUSA
- Present address:
Corteva AgriscienceJohnstonIAUSA
| | - Pramod K. Kandoth
- Division of Plant Sciences and Bond Life Sciences CenterUniversity of MissouriColumbiaMOUSA
- Present address:
National Agri‐food Biotechnology InstituteMohaliIndia
| | - Lakmini Wasala
- Division of Plant Sciences and Bond Life Sciences CenterUniversity of MissouriColumbiaMOUSA
- Present address:
Department of Veterinary PathobiologyUniversity of MissouriColumbiaMOUSA
| | | | - Eric L. Davis
- Department of Entomology and Plant PathologyNorth Carolina State UniversityRaleighNCUSA
| | - Thomas J. Baum
- Department of Plant Pathology and MicrobiologyIowa State UniversityAmesIAUSA
| | - Melissa G. Mitchum
- Division of Plant Sciences and Bond Life Sciences CenterUniversity of MissouriColumbiaMOUSA
- Department of Plant Pathology and Institute of Plant Breeding, Genetics, and GenomicsUniversity of GeorgiaAthensGAUSA
| |
Collapse
|
29
|
Kan LLY, Liu D, Chan BCL, Tsang MSM, Hou T, Leung PC, Lam CWK, Wong CK. The flavonoids of Sophora flavescens exerts anti-inflammatory activity via promoting autophagy of Bacillus Calmette-Guérin-stimulated macrophages. J Leukoc Biol 2020; 108:1615-1629. [PMID: 32794339 DOI: 10.1002/jlb.3ma0720-682rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 07/24/2020] [Accepted: 08/04/2020] [Indexed: 11/06/2022] Open
Abstract
Tuberculosis (TB), a highly infectious air-borne disease, has remained a global health problem. Conventional treatment and preventions such as antibiotics and Bacilli Calmette-Guerin (BCG) vaccine can be unreliable. In view of the increasing prevalence of anti-TB drug resistance, adjunctive therapy may be necessary to shorten the recovery time. We have previously shown that flavonoids in the medicinal herb Sophora flavescens exhibit anti-inflammatory and bactericidal activities. The aim of this study was to investigate the molecular and cellular characteristics of flavonoids of S. flavescens (FSF) in BCG-stimulated macrophages for assessing their roles in anti-inflammation and autophagy. Mouse alveolar macrophage (MH-S) cell line and primary mouse peritoneal macrophages were stimulated in vitro with heat-inactivated BCG and treated with FSF, with or without autophagy inhibitor Bafilomycin A1 (BafA1). Gene expression was analyzed using quantitative PCR, and cytokine/chemokine release was analyzed by Milliplex assay and ELISA. Autophagy-related proteins were quantified by Western blot and flow cytometry, and autophagolysosomes were detected using fluorescence microscopy. In both MH-S cell line and mouse peritoneal macrophages stimulated by heat-inactivated BCG, FSF was found to up-regulate autophagy-related proteins microtubule-associated protein 1A/1B-light chain 3 (LC3) and protein 62 (p62), and suppress the induced proinflammatory cytokine TNF-α, CCL5, and IL-6. FSF actively modulates immune processes through suppressing BCG-mediated inflammation by promoting autophagy in MH-S cells and mouse peritoneal macrophages. We suggest that FSF may be useful as an adjunctive therapeutic agent for TB infection by modulating cell survival through autophagy and reducing inflammation.
Collapse
Affiliation(s)
- Lea Ling-Yu Kan
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Dehua Liu
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Ben Chung-Lap Chan
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Miranda Sin-Man Tsang
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China.,Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Tianheng Hou
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Ping Chung Leung
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Christopher Wai-Kei Lam
- Faculty of Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Chun Kwok Wong
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China.,Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China.,Li Dak Sum Yip Yio Chin R & D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
30
|
Vesicular transport mediates the uptake of cytoplasmic proteins into mitochondria in Drosophila melanogaster. Nat Commun 2020; 11:2592. [PMID: 32444642 PMCID: PMC7244744 DOI: 10.1038/s41467-020-16335-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/27/2020] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial aging, which results in mitochondrial dysfunction, is strongly linked to many age-related diseases. Aging is associated with mitochondrial enlargement and transport of cytosolic proteins into mitochondria. The underlying homeostatic mechanisms that regulate mitochondrial morphology and function, and their breakdown during aging, remain unclear. Here, we identify a mitochondrial protein trafficking pathway in Drosophila melanogaster involving the mitochondria-associated protein Dosmit. Dosmit induces mitochondrial enlargement and the formation of double-membraned vesicles containing cytosolic protein within mitochondria. The rate of vesicle formation increases with age. Vesicles originate from the outer mitochondrial membrane as observed by tracking Tom20 localization, and the process is mediated by the mitochondria-associated Rab32 protein. Dosmit expression level is closely linked to the rate of ubiquitinated protein aggregation, which are themselves associated with age-related diseases. The mitochondrial protein trafficking route mediated by Dosmit offers a promising target for future age-related mitochondrial disease therapies. Mitochondrial dynamics change during ageing, with larger mitochondria and altered protein import in older animals. Here the authors show that Dosmit protein mediates mitochondrial morphology with Rab32 by inducing double-membraned vesicles that regulate protein trafficking into mitochondria.
Collapse
|
31
|
Li Z, Huang W, Wang W. Multifaceted roles of COPII subunits in autophagy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118627. [DOI: 10.1016/j.bbamcr.2019.118627] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 12/09/2019] [Accepted: 12/14/2019] [Indexed: 01/06/2023]
|
32
|
Zhang H, Wang G, Zhou R, Li X, Sun Y, Li Y, Du W, Yan X, Yang J, Chang X, Liu Z, Ma Z. SPIB promotes anoikis resistance via elevated autolysosomal process in lung cancer cells. FEBS J 2020; 287:4696-4709. [PMID: 32129936 DOI: 10.1111/febs.15272] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 01/23/2020] [Accepted: 02/29/2020] [Indexed: 12/27/2022]
Abstract
Anoikis (detachment-induced cell death) is a specific type of programmed cell death which occurs in response to the loss of the correct extracellular matrix connections. Anoikis resistance is an important mechanism in cancer invasiveness and metastatic behavior. Autophagy, on the other hand, involves the degradation of damaged organelles and the recycling of misfolded proteins and intracellular components. However, the intersection of these two cellular responses in lung cancer cells has not been extensively studied. Here, we identified that upon matrix deprivation, the lymphocyte lineage-specific Ets transcription factor SPIB was activated and directly enhanced SNAP47 transcription in certain lung cancer cells. Loss of attachment-induced autophagy significantly increased anoikis resistance by SPIB activation. Consistent with this function, SPIB depletion by short hairpin RNA abrogated SNAP47 transcriptional activation upon matrix deprivation. Therefore, these data delineate an important role of SPIB in autophagy-mediated anoikis resistance in lung cancer cells. Accordingly, these findings suggest that manipulating SPIB-regulated pathways in vivo and evaluating the impact of anoikis resistance warrant further investigation. DATABASE: RNA sequencing and ChIP sequencing data are available in Gene Expression Omnibus database under the accession numbers GSE106592 and GSE125561, respectively.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Immunology, Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, China.,Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, China
| | - Guobin Wang
- Department of Immunology, Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, China.,Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, China
| | - Ruimin Zhou
- Department of Immunology, Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, China.,Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, China
| | - Xiaobo Li
- Department of Immunology, Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, China.,Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, China
| | - Yanan Sun
- Department of Immunology, Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, China.,Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, China
| | - Yanzhe Li
- Department of Immunology, Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, China.,Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, China
| | - Wei Du
- Department of Immunology, Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, China.,Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, China
| | - Xiaojie Yan
- Department of Immunology, Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, China.,Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, China
| | - Jie Yang
- Department of Immunology, Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, China.,Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, China
| | - Xinzhong Chang
- Department of Breast Cancer, Breast Cancer Center, Tianjin Medical University Cancer Institute and Hospital, China
| | - Zhe Liu
- Department of Immunology, Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, China.,Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, China
| | - Zhenyi Ma
- Department of Immunology, Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, China.,Tianjin Key Laboratory of Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, China
| |
Collapse
|
33
|
Humbert M, Morán M, de la Cruz-Ojeda P, Muntané J, Wiedmer T, Apostolova N, McKenna SL, Velasco G, Balduini W, Eckhart L, Janji B, Sampaio-Marques B, Ludovico P, Žerovnik E, Langer R, Perren A, Engedal N, Tschan MP. Assessing Autophagy in Archived Tissue or How to Capture Autophagic Flux from a Tissue Snapshot. BIOLOGY 2020; 9:E59. [PMID: 32245178 PMCID: PMC7150830 DOI: 10.3390/biology9030059] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 12/14/2022]
Abstract
Autophagy is a highly conserved degradation mechanism that is essential for maintaining cellular homeostasis. In human disease, autophagy pathways are frequently deregulated and there is immense interest in targeting autophagy for therapeutic approaches. Accordingly, there is a need to determine autophagic activity in human tissues, an endeavor that is hampered by the fact that autophagy is characterized by the flux of substrates whereas histology informs only about amounts and localization of substrates and regulators at a single timepoint. Despite this challenging task, considerable progress in establishing markers of autophagy has been made in recent years. The importance of establishing clear-cut autophagy markers that can be used for tissue analysis cannot be underestimated. In this review, we attempt to summarize known techniques to quantify autophagy in human tissue and their drawbacks. Furthermore, we provide some recommendations that should be taken into consideration to improve the reliability and the interpretation of autophagy biomarkers in human tissue samples.
Collapse
Grants
- none Bernese Cancer League
- none Stiftung für klinisch-experimentelle Tumorforschung
- none Werner and Hedy Berger-Janser Foundation for Cancer Research
- PI14/01085 and PI17/00093 FIS and FEDER funds from the EU
- CPII16/00023 ISCIII and FSE funds
- RTI2018-096748-B-100 the Spanish Minsitry of Science, Innovation and Universities
- none University Professor Training Fellowship, Ministry of Science, Innovation and University, Government of Spain
- PI18/00442 the State Plan for R & D + I2013-2016 and funded by the Instituto de Salud Carlos III
- none European Regional Development Fund
- C18/BM/12670304/COMBATIC Luxembourg National Research Fund
- NORTE-01-0145-FEDER-000013 Northern Portugal Regional Operational Programme (NORTE 2020), under the Portugal 2020 Partnership Agreement, by the European Regional Development Fund (FEDER), through the Competitiveness Factors Operational Programme (COMPETE)
- POCI-01-0145-FEDER-028159 and POCI-01-0145-FEDER-030782 FEDER, through the COMPETE
- none National funds, through the Foundation for Science and Technology (FCT
- none ARRS - the Slovenian research agency, programme P1-0140: Proteolysis and its regulation
- KFS-3360-02-2014 the Swiss Cancer Research
- KFS-3409-02-2014 the Swiss Cancer Research
- 31003A_173219 Swiss National Science Foundation
Collapse
Affiliation(s)
- Magali Humbert
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH-3008 Bern, Switzerland; (T.W.); (R.L.); (A.P.)
| | - María Morán
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital ‘12 de Octubre’ (‘imas12’), 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Patricia de la Cruz-Ojeda
- Institute of Biomedicine of Seville (IBiS), Hospital University “Virgen del Rocío”/CSIC/University of Seville, 41013 Seville, Spain;
- Department of Surgery, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Jordi Muntané
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Institute of Biomedicine of Seville (IBiS), Hospital University “Virgen del Rocío”/CSIC/University of Seville, 41013 Seville, Spain;
- Department of Surgery, School of Medicine, University of Seville, 41009 Seville, Spain
- Spanish Network for Biomedical Research in Hepatic and Digestive Diseases (CIBERehd), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Tabea Wiedmer
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH-3008 Bern, Switzerland; (T.W.); (R.L.); (A.P.)
| | - Nadezda Apostolova
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Spanish Network for Biomedical Research in Hepatic and Digestive Diseases (CIBERehd), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- Department of Pharmacology, University of Valencia, 46010 Valencia, Spain
| | - Sharon L. McKenna
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Cancer Research at UCC, Western Gateway Building, University College Cork, T12 XF62 Cork, Ireland
| | - Guillermo Velasco
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University, and Instituto de Investigaciones Sanitarias San Carlos (IdISSC), 28040 Madrid, Spain
| | - Walter Balduini
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Leopold Eckhart
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Department of Dermatology, Medical University of Vienna, Vienna 1090, Austria
| | - Bassam Janji
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Oncology—Luxembourg Institute of Health, 1526 Luxembourg City, Luxembourg
| | - Belém Sampaio-Marques
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Paula Ludovico
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Eva Žerovnik
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
| | - Rupert Langer
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH-3008 Bern, Switzerland; (T.W.); (R.L.); (A.P.)
| | - Aurel Perren
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH-3008 Bern, Switzerland; (T.W.); (R.L.); (A.P.)
| | - Nikolai Engedal
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 0424 Oslo, Norway
| | - Mario P. Tschan
- TRANSAUTOPHAGY: European Network for Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, 08193 Barcelona, Spain; (M.M.); (J.M.); (N.A.); (S.L.M.); (G.V.); (W.B.); (L.E.); (B.J.); (B.S.-M.); (P.L.); (E.Ž.); (N.E.)
- Institute of Pathology, University of Bern, Murtenstrasse 31, CH-3008 Bern, Switzerland; (T.W.); (R.L.); (A.P.)
| |
Collapse
|
34
|
Garza-Lombó C, Pappa A, Panayiotidis MI, Franco R. Redox homeostasis, oxidative stress and mitophagy. Mitochondrion 2020; 51:105-117. [PMID: 31972372 DOI: 10.1016/j.mito.2020.01.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 12/21/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023]
Abstract
Autophagy is a ubiquitous homeostatic mechanism for the degradation or turnover of cellular components. Degradation of mitochondria via autophagy (mitophagy) is involved in a number of physiological processes including cellular homeostasis, differentiation and aging. Upon stress or injury, mitophagy prevents the accumulation of damaged mitochondria and the increased steady state levels of reactive oxygen species leading to oxidative stress and cell death. A number of human diseases, particularly neurodegenerative disorders, have been linked to the dysregulation of mitophagy. In this mini-review, we aimed to review the molecular mechanisms involved in the regulation of mitophagy and their relationship with redox signaling and oxidative stress.
Collapse
Affiliation(s)
- Carla Garza-Lombó
- Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States.
| | - Aglaia Pappa
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | | | - Rodrigo Franco
- Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States.
| |
Collapse
|
35
|
Aivazidis S, Jain A, Rauniyar AK, Anderson CC, Marentette JO, Orlicky DJ, Fritz KS, Harris PS, Siegel D, Maclean KN, Roede JR. SNARE proteins rescue impaired autophagic flux in Down syndrome. PLoS One 2019; 14:e0223254. [PMID: 31714914 PMCID: PMC6850524 DOI: 10.1371/journal.pone.0223254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/17/2019] [Indexed: 01/20/2023] Open
Abstract
Down syndrome (DS) is a chromosomal disorder caused by trisomy of chromosome 21 (Ts21). Unbalanced karyotypes can lead to dysfunction of the proteostasis network (PN) and disrupted proteostasis is mechanistically associated with multiple DS comorbidities. Autophagy is a critical component of the PN that has not previously been investigated in DS. Based on our previous observations of PN disruption in DS, we investigated possible dysfunction of the autophagic machinery in human DS fibroblasts and other DS cell models. Following induction of autophagy by serum starvation, DS fibroblasts displayed impaired autophagic flux indicated by autophagolysosome accumulation and elevated p62, NBR1, and LC3-II abundance, compared to age- and sex-matched, euploid (CTL) fibroblasts. While lysosomal physiology was unaffected in both groups after serum starvation, we observed decreased basal abundance of the Soluble N-ethylmaleimide-sensitive-factor Attachment protein Receptor (SNARE) family members syntaxin 17 (STX17) and Vesicle Associated Membrane Protein 8 (VAMP8) indicating that decreased autophagic flux in DS is due at least in part to a possible impairment of autophagosome-lysosome fusion. This conclusion was further supported by the observation that over-expression of either STX17 or VAMP8 in DS fibroblasts restored autophagic degradation and reversed p62 accumulation. Collectively, our results indicate that impaired autophagic clearance is a characteristic of DS cells that can be reversed by enhancement of SNARE protein expression and provides further evidence that PN disruption represents a candidate mechanism for multiple aspects of pathogenesis in DS and a possible future target for therapeutic intervention.
Collapse
Affiliation(s)
- Stefanos Aivazidis
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - Abhilasha Jain
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - Abhishek K. Rauniyar
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - Colin C. Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - John O. Marentette
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - David J. Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Kristofer S. Fritz
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - Peter S. Harris
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - David Siegel
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
| | - Kenneth N. Maclean
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, United States of America
- The Linda Crnic Institute for Down Syndrome, University of Colorado, Aurora, CO, United States of America
| | - James R. Roede
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO, United States of America
- The Linda Crnic Institute for Down Syndrome, University of Colorado, Aurora, CO, United States of America
- * E-mail:
| |
Collapse
|
36
|
Denu RA, Kaur G, Sass MM, Lakkaraju A, Burkard ME. Centrosome Amplification in Cancer Disrupts Autophagy and Sensitizes to Autophagy Inhibition. Mol Cancer Res 2019; 18:33-45. [PMID: 31604847 DOI: 10.1158/1541-7786.mcr-19-0509] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/30/2019] [Accepted: 10/08/2019] [Indexed: 12/24/2022]
Abstract
Centrosome amplification (CA), or a numerical increase in centrosomes, is common in human cancers, particularly those with high-risk features. We have discovered that cells with CA have an increased burden of autophagy, a catabolic process whereby autophagosomes engulf damaged organelles and proteins and deliver these contents to the lysosome for degradation and subsequent recycling. Cells with CA demonstrate an accumulation of autophagosomes. We evaluated the alternative hypotheses that CA alters autophagy by modulating microtubule networks and impairing trafficking versus altering lysosome clustering and organization versus chromosome missegregation-induced proteotoxic stress. Using LC3 reporter assays and autophagosome tracking experiments, we demonstrate that CA causes an accumulation of autophagosomes by interfering with autophagosome trafficking. To establish whether this was a druggable weakness, we tested autophagy inhibitors in our cell models of CA. Cells with CA are sensitized to chemical and genetic autophagy inhibition. Taken together, our results suggest that autophagy is disrupted by CA and sensitizes cells to inhibition of autophagy. These findings suggest a novel precision medicine strategy, whereby CA increases reliance on autophagy and serves as a biomarker for autophagy inhibitors in high-risk cancers. IMPLICATIONS: Our study suggests that CA could be used as a predictive biomarker for treatment with autophagy inhibitors.
Collapse
Affiliation(s)
- Ryan A Denu
- Medical Scientist Training Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.,Division of Hematology/Oncology, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin.,Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Gulpreet Kaur
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Madilyn M Sass
- Division of Hematology/Oncology, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Aparna Lakkaraju
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California
| | - Mark E Burkard
- Division of Hematology/Oncology, Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin. .,Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
37
|
Valencia Lopez MJ, Schimmeck H, Gropengießer J, Middendorf L, Quitmann M, Schneider C, Holstermann B, Wacker R, Heussler V, Reimer R, Aepfelbacher M, Ruckdeschel K. Activation of the macroautophagy pathway by Yersinia enterocolitica promotes intracellular multiplication and egress of yersiniae from epithelial cells. Cell Microbiol 2019; 21:e13046. [PMID: 31099152 DOI: 10.1111/cmi.13046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 04/30/2019] [Accepted: 05/13/2019] [Indexed: 12/13/2022]
Abstract
The virulence strategy of pathogenic Yersinia spp. involves cell-invasive as well as phagocytosis-preventing tactics to enable efficient colonisation of the host organism. Enteropathogenic yersiniae display an invasive phenotype in early infection stages, which facilitates penetration of the intestinal mucosa. Here we show that invasion of epithelial cells by Yersinia enterocolitica is followed by intracellular survival and multiplication of a subset of ingested bacteria. The replicating bacteria were enclosed in vacuoles with autophagy-related characteristics, showing phagophore formation, xenophagy, and recruitment of cytoplasmic autophagosomes to the bacteria-containing compartments. The subsequent fusion of these vacuoles with lysosomes and concomitant vesicle acidification were actively blocked by Yersinia. This resulted in increased intracellular proliferation and detectable egress of yersiniae from infected cells. Notably, deficiency of the core autophagy machinery component FIP200 impaired the development of autophagic features at Yersinia-containing vacuoles as well as intracellular replication and release of bacteria to the extracellular environment. These results suggest that Y. enterocolitica may take advantage of the macroautophagy pathway in epithelial cells to create an autophagosomal niche that supports intracellular bacterial survival, replication, and, eventually, spread of the bacteria from infected cells.
Collapse
Affiliation(s)
- Maria Jose Valencia Lopez
- Institute for Medical Microbiology, Virology and Hygiene, University, Medical Center Eppendorf, Hamburg, Germany
| | - Hanna Schimmeck
- Institute for Medical Microbiology, Virology and Hygiene, University, Medical Center Eppendorf, Hamburg, Germany
| | - Julia Gropengießer
- Institute for Medical Microbiology, Virology and Hygiene, University, Medical Center Eppendorf, Hamburg, Germany
| | - Lukas Middendorf
- Institute for Medical Microbiology, Virology and Hygiene, University, Medical Center Eppendorf, Hamburg, Germany
| | - Melanie Quitmann
- Institute for Medical Microbiology, Virology and Hygiene, University, Medical Center Eppendorf, Hamburg, Germany
| | - Carola Schneider
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Barbara Holstermann
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Rahel Wacker
- Institute for Cell Biology, University of Bern, Bern, Switzerland
| | - Volker Heussler
- Institute for Cell Biology, University of Bern, Bern, Switzerland
| | - Rudolph Reimer
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Martin Aepfelbacher
- Institute for Medical Microbiology, Virology and Hygiene, University, Medical Center Eppendorf, Hamburg, Germany
| | - Klaus Ruckdeschel
- Institute for Medical Microbiology, Virology and Hygiene, University, Medical Center Eppendorf, Hamburg, Germany
| |
Collapse
|
38
|
Junín Virus Promotes Autophagy To Facilitate the Virus Life Cycle. J Virol 2019; 93:JVI.02307-18. [PMID: 31118257 DOI: 10.1128/jvi.02307-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 05/05/2019] [Indexed: 01/19/2023] Open
Abstract
Junín virus (JUNV), a member of the family Arenaviridae, is the etiological agent of Argentine hemorrhagic fever (AHF), a potentially deadly endemic-epidemic disease affecting the population of the most fertile farming land of Argentina. Autophagy is a degradative process with a crucial antiviral role; however, several viruses subvert the pathway to their benefit. We determined the role of autophagy in JUNV-infected cells by analyzing LC3, a cytoplasmic protein (LC3-I) that becomes vesicle membrane associated (LC3-II) upon induction of autophagy. Cells overexpressing enhanced green fluorescent protein (EGFP)-LC3 and infected with JUNV showed an increased number of LC3 punctate structures, similar to those obtained after starvation or bafilomycin A1 treatment, which leads to autophagosome induction or accumulation, respectively. We also monitored the conversion of LC3-I to LC3-II, observing LC3-II levels in JUNV-infected cells similar to those observed in starved cells. Additionally, we kinetically studied the number of LC3 dots after JUNV infection and found that the virus activated the pathway as early as 2 h postinfection (p.i.), whereas the UV-inactivated virus did not induce the pathway. Cells subjected to starvation or pretreated with rapamycin, a pharmacological autophagy inductor, enhanced virus yield. Also, we assayed the replication capacity of JUNV in Atg5 knockout or Beclin 1 knockdown cells (both critical components of the autophagic pathway) and found a significant decrease in JUNV replication. Taken together, our results constitute the first study indicating that JUNV infection induces an autophagic response, which is functionally required by the virus for efficient propagation.IMPORTANCE Mammalian arenaviruses are zoonotic viruses that cause asymptomatic and persistent infections in their rodent hosts but may produce severe and lethal hemorrhagic fevers in humans. Currently, there are neither effective therapeutic options nor effective vaccines for viral hemorrhagic fevers caused by human-pathogenic arenaviruses, except the vaccine Candid no. 1 against Argentine hemorrhagic fever (AHF), licensed for human use in areas of endemicity in Argentina. Since arenaviruses remain a severe threat to global public health, more in-depth knowledge of their replication mechanisms would improve our ability to fight these viruses. Autophagy is a lysosomal degradative pathway involved in maintaining cellular homeostasis, representing powerful anti-infective machinery. We show, for the first time for a member of the family Arenaviridae, a proviral role of autophagy in JUNV infection, providing new knowledge in the field of host-virus interaction. Therefore, modulation of virus-induced autophagy could be used as a strategy to block arenavirus infections.
Collapse
|
39
|
Santana RAG, Oliveira MC, Cabral I, Junior RCAS, de Sousa DRT, Ferreira L, Lacerda MVG, Monteiro WM, Abrantes P, Guerra MDGVB, Silveira H. Anopheles aquasalis transcriptome reveals autophagic responses to Plasmodium vivax midgut invasion. Parasit Vectors 2019; 12:261. [PMID: 31126324 PMCID: PMC6534896 DOI: 10.1186/s13071-019-3506-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 05/14/2019] [Indexed: 01/23/2023] Open
Abstract
Background Elimination of malaria depends on mastering transmission and understanding the biological basis of Plasmodium infection in the vector. The first mosquito organ to interact with the parasite is the midgut and its transcriptomic characterization during infection can reveal effective antiplasmodial responses able to limit the survival of the parasite. The vector response to Plasmodium vivax is not fully characterized, and its specificities when compared with other malaria parasites can be of fundamental interest for specific control measures. Methods Experimental infections were performed using a membrane-feeding device. Three groups were used: P. vivax-blood-fed, blood-fed on inactivated gametocytes, and unfed mosquitoes. Twenty-four hours after feeding, the mosquitoes were dissected and the midgut collected for transcriptomic analysis using RNAseq. Nine cDNA libraries were generated and sequenced on an Illumina HiSeq2500. Readings were checked for quality control and analysed using the Trinity platform for de novo transcriptome assembly. Transcript quantification was performed and the transcriptome was functionally annotated. Differential expression gene analysis was carried out. The role of the identified mechanisms was further explored using functional approaches. Results Forty-nine genes were identified as being differentially expressed with P. vivax infection: 34 were upregulated and 15 were downregulated. Half of the P. vivax-related differentially expressed genes could be related to autophagy; therefore, the effect of the known inhibitor (wortmannin) and activator (spermidine) was tested on the infection outcome. Autophagic activation significantly reduced the intensity and prevalence of infection. This was associated with transcription alterations of the autophagy regulating genes Beclin, DRAM and Apg8. Conclusions Our data indicate that P. vivax invasion of An. aquasalis midgut epithelium triggers an autophagic response and its activation reduces infection. This suggests a novel mechanism that mosquitoes can use to fight Plasmodium infection. Electronic supplementary material The online version of this article (10.1186/s13071-019-3506-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rosa Amélia Gonçalves Santana
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Maurício Costa Oliveira
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Iria Cabral
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Rubens Celso Andrade Silva Junior
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Débora Raysa Teixeira de Sousa
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Lucas Ferreira
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Marcus Vinícius Guimarães Lacerda
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil.,Instituto Leônidas & Maria Deane, Fundação Oswaldo Cruz, Manaus, Brazil
| | - Wuelton Marcelo Monteiro
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Patrícia Abrantes
- Instituto de Higiene e Medicina Tropical, Global Health and Tropical Medicine, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Maria das Graças Vale Barbosa Guerra
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Henrique Silveira
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas/Fundação de Medicina Tropical Dr. Heitor Vieira Dourado, Manaus, Brazil. .,Instituto de Higiene e Medicina Tropical, Global Health and Tropical Medicine, Universidade Nova de Lisboa, Lisboa, Portugal.
| |
Collapse
|
40
|
Tettamanti G, Carata E, Montali A, Dini L, Fimia GM. Autophagy in development and regeneration: role in tissue remodelling and cell survival. EUROPEAN ZOOLOGICAL JOURNAL 2019. [DOI: 10.1080/24750263.2019.1601271] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- G. Tettamanti
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - E. Carata
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
| | - A. Montali
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - L. Dini
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - G. M. Fimia
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Lecce, Italy
- Department of Epidemiology and Preclinical Research, National Institute for Infectious Diseases IRCCS “Lazzaro Spallanzani”, Rome, Italy
| |
Collapse
|
41
|
Mahapatra KK, Panigrahi DP, Praharaj PP, Bhol CS, Patra S, Mishra SR, Behera BP, Bhutia SK. Molecular interplay of autophagy and endocytosis in human health and diseases. Biol Rev Camb Philos Soc 2019; 94:1576-1590. [PMID: 30989802 DOI: 10.1111/brv.12515] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 03/27/2019] [Accepted: 03/29/2019] [Indexed: 12/14/2022]
Abstract
Autophagy, an evolutionarily conserved process for maintaining the physio-metabolic equilibrium of cells, shares many common effector proteins with endocytosis. For example, tethering proteins involved in fusion like Ras-like GTPases (Rabs), soluble N-ethylmaleimide sensitive factor attachment protein receptors (SNAREs), lysosomal-associated membrane protein (LAMP), and endosomal sorting complex required for transport (ESCRT) have a dual role in endocytosis and autophagy, and the trafficking routes of these processes converge at lysosomes. These common effectors indicate an association between budding and fusion of membrane-bound vesicles that may have a substantial role in autophagic lysosome reformation, by sensing cellular stress levels. Therefore, autophagy-endocytosis crosstalk may be significant and implicates a novel endocytic regulatory pathway of autophagy. Moreover, endocytosis has a pivotal role in the intake of signalling molecules, which in turn activates cascades that can result in pathophysiological conditions. This review discusses the basic mechanisms of this crosstalk and its implications in order to identify potential novel therapeutic targets for various human diseases.
Collapse
Affiliation(s)
- Kewal K Mahapatra
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Debasna P Panigrahi
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Prakash P Praharaj
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Chandra S Bhol
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Srimanta Patra
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Soumya R Mishra
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Bishnu P Behera
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Sujit K Bhutia
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| |
Collapse
|
42
|
Gratton R, Agrelli A, Tricarico PM, Brandão L, Crovella S. Autophagy in Zika Virus Infection: A Possible Therapeutic Target to Counteract Viral Replication. Int J Mol Sci 2019; 20:ijms20051048. [PMID: 30823365 PMCID: PMC6429311 DOI: 10.3390/ijms20051048] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/21/2019] [Accepted: 02/23/2019] [Indexed: 12/12/2022] Open
Abstract
Zika virus (ZIKV) still constitutes a public health concern, however, no vaccines or therapies are currently approved for treatment. A fundamental process involved in ZIKV infection is autophagy, a cellular catabolic pathway delivering cytoplasmic cargo to the lysosome for degradation—considered as a primordial form of innate immunity against invading microorganisms. ZIKV is thought to inhibit the Akt-mTOR signaling pathway, which causes aberrant activation of autophagy promoting viral replication and propagation. It is therefore appealing to study the role of autophagic molecular effectors during viral infection to identify potential targets for anti-ZIKV therapeutic intervention.
Collapse
Affiliation(s)
- Rossella Gratton
- Department of Advanced Diagnostics, IRCCS Burlo Garofolo, Via dell'Istria 65/1, 34137 Trieste, Italy.
| | - Almerinda Agrelli
- Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235-Cidade Universitária, 50670-901 Recife, Brazil.
| | - Paola Maura Tricarico
- Department of Advanced Diagnostics, IRCCS Burlo Garofolo, Via dell'Istria 65/1, 34137 Trieste, Italy.
| | - Lucas Brandão
- Department of Pathology, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235-Cidade Universitária, 50670-901 Recife, Brazil.
| | - Sergio Crovella
- Department of Advanced Diagnostics, IRCCS Burlo Garofolo, Via dell'Istria 65/1, 34137 Trieste, Italy.
- Department of Medical Surgical and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy.
| |
Collapse
|
43
|
Pirmoradi L, Seyfizadeh N, Ghavami S, Zeki AA, Shojaei S. Targeting cholesterol metabolism in glioblastoma: a new therapeutic approach in cancer therapy. J Investig Med 2019; 67:715-719. [PMID: 30765502 DOI: 10.1136/jim-2018-000962] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2018] [Indexed: 12/11/2022]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive malignant brain tumor known with a poor survival rate despite current advances in the field of cancer. Additional research into the pathophysiology of GBM is urgently needed given the devastating nature of this disease. Recent studies have revealed the unique cellular physiology of GBM cells as compared with healthy astrocytes. Intriguingly, GBM cells are incapable of de novo cholesterol synthesis via the mevalonate pathway. Thus, the survival of GBM cells depends on cholesterol uptake via low-density lipoprotein receptors (LDLRs) in the form of apolipoprotein-E-containing lipoproteins and ATP-binding cassette transporter A1 (ABCA1) that efflux surplus cholesterol out of cells. Liver X receptors regulate intracellular cholesterol levels in neurons and healthy astrocytes through changes in the expression of LDLR and ABCA1 in response to cholesterol and its derivatives. In GBM cells, due to the dysregulation of this surveillance pathway, there is an accumulation of intracellular cholesterol. Furthermore, intracellular cholesterol regulates temozolomide-induced cell death in glioblastoma cells via accumulation and activation of death receptor 5 in plasma membrane lipid rafts. The mevalonate pathway and autophagy flux are also fundamentally related with implications for cell health and death. Thus, via cholesterol metabolism, the mevalonate pathway may be a crucial player in the pathogenesis and treatment of GBM where our current understanding is still lacking. Targeting cholesterol metabolism in GBM may hold promise as a novel adjunctive clinical therapy for this devastating cancer.
Collapse
Affiliation(s)
- Leila Pirmoradi
- Department of Physiology and Pharmacology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Kurdistan, Iran
| | - Nayer Seyfizadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Biology of Breathing, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada.,Research Institute in Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, Manitoba, Canada.,Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Amir A Zeki
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of California Davis, Sacramento, California, USA.,Center for Comparative Respiratory Biology and Medicine, University of California, Davis, School of Medicine, Davis, California, USA
| | - Shahla Shojaei
- Department of Human Anatomy and Cell Sciences, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.,Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Isfahan, Iran
| |
Collapse
|
44
|
Elhassan SAM, Candasamy M, Chan EWL, Bhattamisra SK. Autophagy and GLUT4: The missing pieces. Diabetes Metab Syndr 2018; 12:1109-1116. [PMID: 29843994 DOI: 10.1016/j.dsx.2018.05.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 05/21/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND Autophagy is a process devoted to degrade and recycle cellular components inside mammalian cells through lysosomal system. It plays a main function in the pathophysiology of several diseases. In type 2 diabetes, works demonstrated the dual functions of autophagy in diabetes biology. Studies had approved the role of autophagy in promoting different routes for movement of integral membrane proteins to the plasma membrane. But its role in regulation of GLUT4 trafficking has not been widely observed. In normal conditions, insulin promotes GLUT4 translocation from intracellular membrane compartments to the plasma membrane, while in type 2 diabetes defects occur in this translocation. METHOD Intriguing evidences discussed the contribution of different intracellular compartments in autophagy membrane formation. Furthermore, autophagy serves to mobilise membranes within cells, thereby promoting cytoplasmic components reorganisation. The intent of this review is to focus on the possibility of autophagy to act as a carrier for GLUT4 through regulating GLUT4 endocytosis, intracellular trafficking in different compartments, and translocation to cell membrane. RESULTS The common themes of autophagy and GLUT4 have been highlighted. The review discussed the overlapping of endocytosis mechanism and intracellular compartments, and has shown that autophagy and GLUT4 utilise similar proteins (SNAREs) which are used for exocytosis. On top of that, PI3K and AMPK also control both autophagy and GLUT4. CONCLUSION The control of GLUT4 trafficking through autophagy could be a promising field for treating type 2 diabetes.
Collapse
Affiliation(s)
- Safa Abdelgadir Mohamed Elhassan
- School of Postgraduate Studies, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia.
| | - Mayuren Candasamy
- Department of Life Sciences, School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia.
| | - Elaine Wan Ling Chan
- Institute of Research, Development and Innovation, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia.
| | - Subrat Kumar Bhattamisra
- Department of Life Sciences, School of Pharmacy, International Medical University, No 126, Jalan Jalil Perkasa 19, Bukit Jalil 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
45
|
Rab GTPases in Osteoclastic Endomembrane Systems. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4541538. [PMID: 30186859 PMCID: PMC6114073 DOI: 10.1155/2018/4541538] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 07/18/2018] [Indexed: 12/13/2022]
Abstract
Osteoclasts (OCs) are bone-resorbing cells that maintain bone homeostasis. OC differentiation, survival, and activity are regulated by numerous small GTPases, including those of the Rab family, which are involved in plasma membrane delivery and lysosomal and autophagic degradation pathways. In resorbing OCs, polarized vesicular trafficking pathways also result in formation of the ruffled membrane, the resorbing organelle, and in transcytosis.
Collapse
|
46
|
Orsini M, Morceau F, Dicato M, Diederich M. Autophagy as a pharmacological target in hematopoiesis and hematological disorders. Biochem Pharmacol 2018; 152:347-361. [PMID: 29656115 DOI: 10.1016/j.bcp.2018.04.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/10/2018] [Indexed: 12/14/2022]
Abstract
Autophagy is involved in many cellular processes, including cell homeostasis, cell death/survival balance and differentiation. Autophagy is essential for hematopoietic stem cell survival, quiescence, activation and differentiation. The deregulation of this process is associated with numerous hematological disorders and pathologies, including cancers. Thus, the use of autophagy modulators to induce or inhibit autophagy emerges as a potential therapeutic approach for treating these diseases and could be particularly interesting for differentiation therapy of leukemia cells. This review presents therapeutic strategies and pharmacological agents in the context of hematological disorders. The pros and cons of autophagy modulators in therapy will also be discussed.
Collapse
Affiliation(s)
- Marion Orsini
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Franck Morceau
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Mario Dicato
- Laboratoire de Biologie Moléculaire et Cellulaire du Cancer, Hôpital Kirchberg, 9, rue Edward Steichen, L-2540 Luxembourg, Luxembourg
| | - Marc Diederich
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| |
Collapse
|
47
|
Castets P, Frank S, Sinnreich M, Rüegg MA. "Get the Balance Right": Pathological Significance of Autophagy Perturbation in Neuromuscular Disorders. J Neuromuscul Dis 2018; 3:127-155. [PMID: 27854220 PMCID: PMC5271579 DOI: 10.3233/jnd-160153] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent research has revealed that autophagy, a major catabolic process in cells, is dysregulated in several neuromuscular diseases and contributes to the muscle wasting caused by non-muscle disorders (e.g. cancer cachexia) or during aging (i.e. sarcopenia). From there, the idea arose to interfere with autophagy or manipulate its regulatory signalling to help restore muscle homeostasis and attenuate disease progression. The major difficulty for the development of therapeutic strategies is to restore a balanced autophagic flux, due to the dynamic nature of autophagy. Thus, it is essential to better understand the mechanisms and identify the signalling pathways at play in the control of autophagy in skeletal muscle. A comprehensive analysis of the autophagic flux and of the causes of its dysregulation is required to assess the pathogenic role of autophagy in diseased muscle. Furthermore, it is essential that experiments distinguish between primary dysregulation of autophagy (prior to disease onset) and impairments as a consequence of the pathology. Of note, in most muscle disorders, autophagy perturbation is not caused by genetic modification of an autophagy-related protein, but rather through indirect alteration of regulatory signalling or lysosomal function. In this review, we will present the mechanisms involved in autophagy, and those ensuring its tight regulation in skeletal muscle. We will then discuss as to how autophagy dysregulation contributes to the pathogenesis of neuromuscular disorders and possible ways to interfere with this process to limit disease progression.
Collapse
Affiliation(s)
| | - Stephan Frank
- Institute of Pathology, Division of Neuropathology Basel University Hospital, Basel, Switzerland
| | - Michael Sinnreich
- Neuromuscular Research Center, Departments of Neurology and Biomedicine, Pharmazentrum, Basel, Switzerland
| | | |
Collapse
|
48
|
Abstract
Autophagy is an evolutionarily conserved degradation pathway for cells to maintain homeostasis, produce energy, degrade misfolded proteins and damaged organelles, and fight against intracellular pathogens. The process of autophagy entails the isolation of cytoplasmic cargo into double membrane bound autophagosomes that undergo maturation by fusion with endosomes and lysosomes to obtain degradation capacity. RAB proteins regulate intracellular vesicle trafficking events including autophagy. RAB24 is an atypical RAB protein that is required for the clearance of late autophagic vacuoles under basal conditions. RAB24 has also been connected to several diseases including ataxia, cancer and tuberculosis. This review gives a short summary on autophagy and RAB proteins, and an overview on the current knowledge on the roles of RAB24 in autophagy and disease.
Collapse
Affiliation(s)
- Päivi Ylä-Anttila
- a Department of Biosciences , University of Helsinki , Helsinki , Finland
| | | |
Collapse
|
49
|
Zhao S, Yang XF, Shen DF, Gao Y, Shi S, Wu JC, Liu HX, Sun HZ, Su RJ, Zheng HC. The down-regulated ING5 expression in lung cancer: a potential target of gene therapy. Oncotarget 2018; 7:54596-54615. [PMID: 27409347 PMCID: PMC5342367 DOI: 10.18632/oncotarget.10519] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 05/28/2016] [Indexed: 12/11/2022] Open
Abstract
ING5 can interact with p53, thereby inhibiting cell growth and inducing apoptosis. We found that ING5 overexpression not only inhibited proliferation, migration, and invasion, but also induced G2 arrest, differentiation, autophagy, apoptosis, glycolysis and mitochondrial respiration in lung cancer cells. ING5 transfection up-regulated the expression of Cdc2, ATG13, ATG14, Beclin-1, LC-3B, AIF, cytochrome c, Akt1/2/3, ADFP, PFK-1 and PDPc, while down-regulated the expression of Bcl-2, XIAP, survivin,β-catenin and HXK1. ING5 transfection desensitized cells to the chemotherapy of MG132, paclitaxel, and SAHA, which paralleled with apoptotic alteration. ING5 overexpression suppressed the xenograft tumor growth by inhibiting proliferation and inducing apoptosis. ING5 expression level was significantly higher in normal tissue than that in lung cancer at both protein and mRNA levels. Nuclear ING5 expression was positively correlated with ki-67 expression and cytoplasmic ING5 expression. Cytoplasmic ING5 expression was positively associated with lymph node metastasis, and negatively with age, lymphatic invasion or CPP32 expression. ING5 expression was different in histological classification: squamous cell carcinoma > adenocarcinoma > large cell carcinoma > small cell carcinoma. Taken together, our data suggested that ING5 downregulation might involved in carcinogenesis, growth, and invasion of lung cancer and could be considered as a promising marker to gauge the aggressiveness of lung cancer. It might be employed as a potential target for gene therapy of lung cancer.
Collapse
Affiliation(s)
- Shuang Zhao
- Cancer Center, Key Laboratory of Brain and Spinal Cord Injury of Liaoning Province, and Animal Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Xue-Feng Yang
- Cancer Center, Key Laboratory of Brain and Spinal Cord Injury of Liaoning Province, and Animal Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Dao-Fu Shen
- Cancer Center, Key Laboratory of Brain and Spinal Cord Injury of Liaoning Province, and Animal Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Yang Gao
- Cancer Center, Key Laboratory of Brain and Spinal Cord Injury of Liaoning Province, and Animal Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Shuai Shi
- Cancer Center, Key Laboratory of Brain and Spinal Cord Injury of Liaoning Province, and Animal Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Ji-Cheng Wu
- Cancer Center, Key Laboratory of Brain and Spinal Cord Injury of Liaoning Province, and Animal Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Hong-Xu Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Hong-Zhi Sun
- Cancer Center, Key Laboratory of Brain and Spinal Cord Injury of Liaoning Province, and Animal Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China
| | - Rong-Jian Su
- Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, China
| | - Hua-Chuan Zheng
- Cancer Center, Key Laboratory of Brain and Spinal Cord Injury of Liaoning Province, and Animal Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, China.,Life Science Institute of Jinzhou Medical University, Jinzhou, 121001, China
| |
Collapse
|
50
|
Gregory DJ, Kramnik I, Kobzik L. Protection of macrophages from intracellular pathogens by miR-182-5p mimic-a gene expression meta-analysis approach. FEBS J 2017; 285:244-260. [PMID: 29197182 DOI: 10.1111/febs.14348] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 09/29/2017] [Accepted: 11/28/2017] [Indexed: 12/25/2022]
Abstract
The goals of this study were to (a) define which host genes are of particular importance during the interactions between macrophages and intracellular pathogens, and (b) use this knowledge to gain fresh, experimental understanding of how macrophage activities may be manipulated during host defense. We designed an in silico method for meta-analysis of microarray gene expression data, and used this to combine data from 16 different studies of cells in the monocyte-macrophage lineage infected with seven different pathogens. Three thousand four hundred ninety-eight genes were identified, which we call the macrophage intracellular pathogen response (macIPR) gene set. As expected, the macIPR gene set showed a strong bias toward genes previously associated with the immune response. Predicted target sites for miR-182-5p (miR-182) were strongly over-represented among macIPR genes, indicating an unexpected role for miR-182-regulatable genes during intracellular pathogenesis. We therefore transfected primary human alveolar macrophage-like monocyte-derived macrophages from multiple different donors with synthetic miR-182, and found that miR-182 overexpression (a) increases proinflammatory gene induction during infection with Francisella tularensis live vaccine strain (LVS), (b) primes macrophages for increased autophagy, and (c) enhances macrophage control of both gram negative F. tularensisLVS and gram positive Bacillus anthracisANR-1 spores. These data therefore suggest a new application for miR-182 in promoting resistance to intracellular pathogens.
Collapse
Affiliation(s)
- David J Gregory
- Molecular and Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Igor Kramnik
- Pulmonary Center, Department of Medicine, National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, MA, USA
| | - Lester Kobzik
- Molecular and Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|