1
|
Lampkin BJ, Goldberg BJ, Kritzer JA. Multiplexed no-wash cellular imaging using BenzoTag, an evolved self-labeling protein. Chem Sci 2024; 15:d4sc05090h. [PMID: 39430930 PMCID: PMC11487927 DOI: 10.1039/d4sc05090h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024] Open
Abstract
Self-labeling proteins are powerful tools for exploring biology as they enable the precise cellular localization of a synthetic molecule, often a fluorescent dye. HaloTag7 is the most popular self-labeling protein due to its broad utility, its bio-orthogonality, and the simplicity of its chloroalkane ligand. However, reaction rates of HaloTag7 with different chloroalkane-containing substrates are highly variable and rates are only very fast for rhodamine-based dyes. This is a major limitation for the HaloTag system because fast labeling rates are critical for live-cell assays. Here, we use yeast surface display to produce a HaloTag variant, BenzoTag, with improved performance with a fluorogenic benzothiadiazole dye. Molecular evolution improved conjugation kinetics and increased the signal from the dye-protein complex, allowing for robust, no-wash fluorescence labeling in live cells. The new BenzoTag-benzothiadiazole system has improved performance compared to the best existing HaloTag7-silicon rhodamine system, including saturation of intracellular enzyme in under 100 seconds and robust labeling at dye concentrations as low as 7 nM. The BenzoTag system was also found to be sufficiently orthogonal to the HaloTag7-silicon rhodamine system to enable multiplexed no-wash labeling in live cells. The BenzoTag system will be immediately useful for a large variety of cell-based assays monitoring biological processes and drug action in real time.
Collapse
Affiliation(s)
- Bryan J Lampkin
- Department of Chemistry, Tufts University Medford MA 02155 USA
| | | | | |
Collapse
|
2
|
Reynolds CA, Pelka S, Gjergjova F, Tasset I, Khawaja RR, Lindenau K, Krause GJ, Gavathiotis E, Cuervo AM, Macian F. Restoration of LAMP2A expression in old mice leads to changes in the T cell compartment that support improved immune function. Proc Natl Acad Sci U S A 2024; 121:e2322929121. [PMID: 39259591 PMCID: PMC11420204 DOI: 10.1073/pnas.2322929121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 08/08/2024] [Indexed: 09/13/2024] Open
Abstract
Chaperone-mediated autophagy (CMA) is a selective form of autophagy that contributes to the maintenance of cellular homeostasis. CMA activity declines with age in most tissues and systems, including the immune system, due to a reduction in levels of lysosome-associated membrane protein type 2A (LAMP2A), an essential CMA component. In this study, we show that overexpressing a copy of hLAMP2A within T cells since middle-age can prevent some of their age-associated loss of function. Our data support the idea that preserving LAMP2A expression with age through genetic means leads to enhanced proliferative responses, decreased number of regulatory T cell populations, and down-regulated expression of inhibitory receptors by T cells. During aging, elevated numbers of these immunosuppressive T cell populations significantly contribute to the age-associated downregulation of T cell responses. Using comparative proteomics, we confirm that preservation of CMA activity in old mice prevents age-related changes in both the resting and the activated T cell proteome. We also explore the effect of using first-in-class small molecule activators of CMA and demonstrate improved T cell response upon their administration to old mice. We conclude that sustaining CMA activity constitutes a potentially viable therapeutic approach to improving T cell function with age.
Collapse
Affiliation(s)
- Cara A. Reynolds
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY10461
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY10461
| | - Sandra Pelka
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY10461
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY10461
| | - Floralba Gjergjova
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY10461
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY10461
| | - Inmaculada Tasset
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY10461
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY10461
- Department of Biochemistry and Molecular Biology, University of Cordoba, Spain14071
| | - Rabia R. Khawaja
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY10461
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY10461
| | - Kristen Lindenau
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY10461
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY10461
| | - Gregory J. Krause
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY10461
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY10461
| | - Evripidis Gavathiotis
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY10461
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY10461
| | - Ana Maria Cuervo
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY10461
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY10461
| | - Fernando Macian
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY10461
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY10461
| |
Collapse
|
3
|
Cheng Z, Gan W, Xiang Q, Zhao K, Gao H, Chen Y, Shi P, Zhang A, Li G, Song Y, Feng X, Yang C, Zhang Y. Impaired degradation of PLCG1 by chaperone-mediated autophagy promotes cellular senescence and intervertebral disc degeneration. Autophagy 2024:1-23. [PMID: 39212196 DOI: 10.1080/15548627.2024.2395797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Defects in chaperone-mediated autophagy (CMA) are associated with cellular senescence, but the mechanism remains poorly understood. Here, we found that CMA inhibition induced cellular senescence in a calcium-dependent manner and identified its role in TNF-induced senescence of nucleus pulposus cells (NPC) and intervertebral disc degeneration. Based on structural and functional proteomic screens, PLCG1 (phospholipase C gamma 1) was predicted as a potential substrate for CMA deficiency to affect calcium homeostasis. We further confirmed that PLCG1 was a key mediator of CMA in the regulation of intracellular calcium flux. Aberrant accumulation of PLCG1 caused by CMA blockage resulted in calcium overload, thereby inducing NPC senescence. Immunoassays on human specimens showed that reduced LAMP2A, the rate-limiting protein of CMA, or increased PLCG1 was associated with disc senescence, and the TNF-induced disc degeneration in rats was inhibited by overexpression of Lamp2a or knockdown of Plcg1. Because CMA dysregulation, calcium overload, and cellular senescence are common features of disc degeneration and other age-related degenerative diseases, the discovery of actionable molecular targets that can link these perturbations may have therapeutic value.Abbreviation: ATRA: all-trans-retinoic acid; BrdU: bromodeoxyuridine; CDKN1A/p21: cyclin dependent kinase inhibitor 1A; CDKN2A/p16-INK4A: cyclin dependent kinase inhibitor 2A; CMA: chaperone-mediated autophagy; DHI: disc height index; ER: endoplasmic reticulum; IP: immunoprecipitation; IP3: inositol 1,4,5-trisphosphate; ITPR/IP3R: inositol 1,4,5-trisphosphate receptor; IVD: intervertebral disc; IVDD: intervertebral disc degeneration; KD: knockdown; KO: knockout; Leu: leupeptin; MRI: magnetic resonance imaging; MS: mass spectrometry; N/L: NH4Cl and leupeptin; NP: nucleus pulposus; NPC: nucleus pulposus cells; PI: protease inhibitors; PLC: phospholipase C; PLCG1: phospholipase C gamma 1; ROS: reactive oxygen species; RT-qPCR: real-time quantitative reverse transcription PCR; SA-GLB1/β-gal: senescence-associated galactosidase beta 1; SASP: senescence-associated secretory phenotype; STV: starvation; TMT: tandem mass tag; TNF: tumor necrosis factor; TP53: tumor protein p53; UPS: ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Zhangrong Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Weikang Gan
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qian Xiang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China
| | - Kangcheng Zhao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Haiyang Gao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuhang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Pengzhi Shi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Anran Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Gaocai Li
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu Song
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaobo Feng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Cao Yang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yukun Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
4
|
Li W, Zhang M, Wang Y, Zhao S, Xu P, Cui Z, Chen J, Xia P, Zhang Y. PRRSV GP5 inhibits the antivirus effects of chaperone-mediated autophagy by targeting LAMP2A. mBio 2024; 15:e0053224. [PMID: 38940560 PMCID: PMC11323736 DOI: 10.1128/mbio.00532-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/17/2024] [Indexed: 06/29/2024] Open
Abstract
Autophagy is an important biological process in host defense against viral infection. However, many viruses have evolved various strategies to disrupt the host antiviral system. Porcine reproductive and respiratory syndrome virus (PRRSV) is a typical immunosuppressive virus with a large economic impact on the swine industry. At present, studies on the escape mechanism of PRRSV in the autophagy process, especially through chaperone-mediated autophagy (CMA), are limited. This study confirmed that PRRSV glycoprotein 5 (GP5) could disrupt the formation of the GFAP-LAMP2A complex by inhibiting the MTORC2/PHLPP1/GFAP pathway, promoting the dissociation of the pGFAP-EF1α complex, and blocking the K63-linked polyubiquitination of LAMP2A to inhibit the activity of CMA. Further research demonstrated that CMA plays an anti-PRRSV role by antagonizing nonstructural protein 11 (NSP11)-mediated inhibition of type I interferon (IFN-I) signaling. Taken together, these results indicate that PRRSV GP5 inhibits the antiviral effect of CMA by targeting LAMP2A. This research provides new insight into the escape mechanism of immunosuppressive viruses in CMA. IMPORTANCE Viruses have evolved sophisticated mechanisms to manipulate autophagy to evade degradation and immune responses. Porcine reproductive and respiratory syndrome virus (PRRSV) is a typical immunosuppressive virus that causes enormous economic losses in the swine industry. However, the mechanism by which PRRSV manipulates autophagy to defend against host antiviral effects remains unclear. In this study, we found that PRRSV GP5 interacts with LAMP2A and disrupts the formation of the GFAP-LAMP2A complex, thus inhibiting the activity of CMA and subsequently enhancing the inhibitory effect of the NSP11-mediated IFN-I signaling pathway, ultimately facilitating PRRSV replication. Our study revealed a novel mechanism by which PRRSV escapes host antiviral effects through CMA, providing a potential host target, LAMP2A, for developing antiviral drugs and contributing to understanding the escape mechanism of immunosuppressive viruses.
Collapse
Affiliation(s)
- Wen Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Mengting Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Yueshuai Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Shijie Zhao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Pengli Xu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Zhiying Cui
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
| | - Jing Chen
- College of Life Science, Henan Agricultural University, Zhengzhou, Henan, China
| | - Pingan Xia
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan, China
| | - Yina Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Jafari M, Macho-González A, Diaz A, Lindenau K, Santiago-Fernández O, Zeng M, Massey AC, de Cabo R, Kaushik S, Cuervo AM. Calorie restriction and calorie-restriction mimetics activate chaperone-mediated autophagy. Proc Natl Acad Sci U S A 2024; 121:e2317945121. [PMID: 38889154 PMCID: PMC11214046 DOI: 10.1073/pnas.2317945121] [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: 10/17/2023] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Chaperone-mediated autophagy (CMA) is part of the mammalian cellular proteostasis network that ensures protein quality control, maintenance of proteome homeostasis, and proteome changes required for the adaptation to stress. Loss of proteostasis is one of the hallmarks of aging. CMA decreases with age in multiple rodent tissues and human cell types. A decrease in lysosomal levels of the lysosome-associated membrane protein type 2A (LAMP2A), the CMA receptor, has been identified as a main reason for declined CMA in aging. Here, we report constitutive activation of CMA with calorie restriction (CR), an intervention that extends healthspan, in old rodent livers and in an in vitro model of CR with cultured fibroblasts. We found that CR-mediated upregulation of CMA is due to improved stability of LAMP2A at the lysosome membrane. We also explore the translational value of our observations using calorie-restriction mimetics (CRMs), pharmacologically active substances that reproduce the biochemical and functional effects of CR. We show that acute treatment of old mice with CRMs also robustly activates CMA in several tissues and that this activation is required for the higher resistance to lipid dietary challenges conferred by treatment with CRMs. We conclude that part of the beneficial effects associated with CR/CRMs could be a consequence of the constitutive activation of CMA mediated by these interventions.
Collapse
Affiliation(s)
- Maryam Jafari
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, BronxNY10461
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY10461
| | - Adrián Macho-González
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, BronxNY10461
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY10461
| | - Antonio Diaz
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, BronxNY10461
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY10461
| | - Kristen Lindenau
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, BronxNY10461
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY10461
| | - Olaya Santiago-Fernández
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, BronxNY10461
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY10461
| | - Mei Zeng
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, BronxNY10461
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY10461
| | - Ashish C. Massey
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, BronxNY10461
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY10461
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD21224
| | - Susmita Kaushik
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, BronxNY10461
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY10461
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, BronxNY10461
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY10461
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY10461
| |
Collapse
|
6
|
Shao J, Lin X, Wang H, Zhao C, Yao SQ, Ge J, Zeng S, Qian L. Targeted Degradation of Cell-Surface Proteins via Chaperone-Mediated Autophagy by Using Peptide-Conjugated Antibodies. Angew Chem Int Ed Engl 2024; 63:e202319232. [PMID: 38472118 DOI: 10.1002/anie.202319232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 03/14/2024]
Abstract
Cell-surface proteins are important drug targets but historically have posed big challenges for the complete elimination of their functions. Herein, we report antibody-peptide conjugates (Ab-CMAs) in which a peptide targeting chaperone-mediated autophagy (CMA) was conjugated with commercially available monoclonal antibodies for specific cell-surface protein degradation by taking advantage of lysosomal degradation pathways. Unique features of Ab-CMAs, including cell-surface receptor- and E3 ligase-independent degradation, feasibility towards different cell-surface proteins (e.g., epidermal growth factor receptor (EGFR), programmed cell death ligand 1 (PD-L1), human epidermal growth factor receptor 2 (HER2)) by a simple change of the antibody, and successful tumor inhibition in vivo, make them attractive protein degraders for biomedical research and therapeutic applications. As the first example employing CMA to degrade proteins from the outside in, our findings may also shed new light on CMA, a degradation pathway typically targeting cytosolic proteins.
Collapse
Affiliation(s)
- Jinning Shao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, China, 310058
| | - Xuefen Lin
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, China, 310058
| | - Haoting Wang
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, China, 310058
| | - Chuhan Zhao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, China, 310058
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore, 117544, Singapore
| | - Jingyan Ge
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology, Zhejiang University of Technology, Hangzhou, China, 310014
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, China, 310058
| | - Linghui Qian
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, China, 310058
| |
Collapse
|
7
|
Rafiq S, Mungure I, Banz Y, Niklaus NJ, Kaufmann T, Müller S, Jacquel A, Robert G, Auberger P, Torbett BE, Muller S, Tschan MP, Humbert M. HSPA8 Chaperone Complex Drives Chaperone-Mediated Autophagy Regulation in Acute Promyelocytic Leukemia Cell Differentiation. Pharmacology 2024; 109:216-230. [PMID: 38569476 DOI: 10.1159/000537864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/14/2024] [Indexed: 04/05/2024]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is a cancer of the hematopoietic system characterized by hyperproliferation of undifferentiated cells of the myeloid lineage. While most of AML therapies are focused toward tumor debulking, all-trans retinoic acid (ATRA) induces neutrophil differentiation in the AML subtype acute promyelocytic leukemia (APL). Macroautophagy has been extensively investigated in the context of various cancers and is often dysregulated in AML where it can have context-dependent pro- or anti-leukemogenic effects. On the contrary, the implications of chaperone-mediated autophagy (CMA) on the pathophysiology of diseases are still being explored and its role in AML remains elusive. METHODS We took advantage of human AML primary samples and databases to analyze CMA gene expression and activity. Furthermore, we used ATRA-sensitive (NB4) and -resistant (NB4-R1) APL cells to further dissect a potential function for CMA in ATRA-mediated neutrophil differentiation. NB4-R1 cells are unique in that they do respond to retinoic acid transcriptionally but do not mature in response to retinoid signaling alone unless maturation is triggered by adding cyclic adenosine monophosphate. RESULTS Here, we report that CMA-related mRNA transcripts are significantly higher expressed in immature hematopoietic cells as compared to neutrophils, contrasting the macroautophagy gene expression patterns. Accordingly, lysosomal degradation of an mCherry-KFERQ CMA reporter decreases during ATRA-induced differentiation of APL cells. On the other hand, using NB4-R1 cells we found that macroautophagy flux primed ATRA-resistant NB4-R1 cells to differentiate upon ATRA treatment but reduced the association of lysosome-associated membrane protein type 2A (LAMP-2A) and heat shock protein family A (Hsp70) member 8 (HSPA8), necessary for complete neutrophil maturation. Accordingly, depletion of HSPA8 attenuated CMA activity and facilitated APL cell differentiation. In contrast, maintaining high CMA activity by ectopic expression of LAMP-2A impeded APL differentiation. CONCLUSION Overall, our findings suggest that APL neutrophil differentiation requires CMA inactivation and that this pathway predominantly depends on HSPA8 and is possibly assisted by other co-chaperones.
Collapse
Affiliation(s)
- Sreoshee Rafiq
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Irene Mungure
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Yara Banz
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Nicolas J Niklaus
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Stefan Müller
- Flow Cytometry and Cell Sorting Core Facility, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | | | | | | | - Bruce E Torbett
- Department of Pediatrics, School of Medicine, Center for Immunity and Immunotherapies, University of Washington and Seattle Children's Research Institute, Seattle, Washington, USA
| | - Sylviane Muller
- TRANSAUTOPHAGY: European Network of Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, Brussels, Belgium
- Ecole Supérieure de Biotechnologie de Strasbourg, CNRS and Strasbourg University, Unit Biotechnology and Cell Signaling, Illkirch, France
- Strasbourg Drug Discovery and Development Institute (IMS), Strasbourg, France
- Chair Therapeutic Immunology, University of Strasbourg Institute for Advanced Study, Strasbourg, France
| | - Mario P Tschan
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- TRANSAUTOPHAGY: European Network of Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, Brussels, Belgium
| | - Magali Humbert
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- TRANSAUTOPHAGY: European Network of Multidisciplinary Research and Translation of Autophagy Knowledge, COST Action CA15138, Brussels, Belgium
| |
Collapse
|
8
|
Lampkin BJ, Goldberg BJ, Kritzer JA. BenzoHTag, a fluorogenic self-labeling protein developed using molecular evolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.29.564634. [PMID: 38617361 PMCID: PMC11014480 DOI: 10.1101/2023.10.29.564634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Self-labeling proteins are powerful tools in chemical biology as they enable the precise cellular localization of a synthetic molecule, often a fluorescent dye, with the genetic specificity of a protein fusion. HaloTag7 is the most popular self-labeling protein due to its fast labeling kinetics and the simplicity of its chloroalkane ligand. Reaction rates of HaloTag7 with different chloroalkane-containing substrates is highly variable and rates are only very fast for rhodamine-based dyes. This is a major limitation for the HaloTag system because fast labeling rates are critical for live-cell assays. Here, we report a molecular evolution system for HaloTag using yeast surface display that enables the screening of libraries up to 108 variants to improve reaction rates with any substrate of interest. We applied this method to produce a HaloTag variant, BenzoHTag, which has improved performance with a fluorogenic benzothiadiazole dye. The resulting system has improved brightness and conjugation kinetics, allowing for robust, no-wash fluorescent labeling in live cells. The new BenzoHTag-benzothiadiazole system has improved performance in live-cell assays compared to the existing HaloTag7-silicon rhodamine system, including saturation of intracellular enzyme in under 100 seconds and robust labeling at dye concentrations as low as 7 nM. It was also found to be orthogonal to the silicon HaloTag7-rhodamine system, enabling multiplexed no-wash labeling in live cells. The BenzoHTag system, and the ability to optimize HaloTag for a broader collection of substrates using molecular evolution, will be very useful for the development of cell-based assays for chemical biology and drug development.
Collapse
|
9
|
Li GL, Han YQ, Su BQ, Yu HS, Zhang S, Yang GY, Wang J, Liu F, Ming SL, Chu BB. Porcine reproductive and respiratory syndrome virus 2 hijacks CMA-mediated lipolysis through upregulation of small GTPase RAB18. PLoS Pathog 2024; 20:e1012123. [PMID: 38607975 PMCID: PMC11014436 DOI: 10.1371/journal.ppat.1012123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 03/14/2024] [Indexed: 04/14/2024] Open
Abstract
RAB GTPases (RABs) control intracellular membrane trafficking with high precision. In the present study, we carried out a short hairpin RNA (shRNA) screen focused on a library of 62 RABs during infection with porcine reproductive and respiratory syndrome virus 2 (PRRSV-2), a member of the family Arteriviridae. We found that 13 RABs negatively affect the yield of PRRSV-2 progeny virus, whereas 29 RABs have a positive impact on the yield of PRRSV-2 progeny virus. Further analysis revealed that PRRSV-2 infection transcriptionally regulated RAB18 through RIG-I/MAVS-mediated canonical NF-κB activation. Disrupting RAB18 expression led to the accumulation of lipid droplets (LDs), impaired LDs catabolism, and flawed viral replication and assembly. We also discovered that PRRSV-2 co-opts chaperone-mediated autophagy (CMA) for lipolysis via RAB18, as indicated by the enhanced associations between RAB18 and perlipin 2 (PLIN2), CMA-specific lysosomal associated membrane protein 2A (LAMP2A), and heat shock protein family A (Hsp70) member 8 (HSPA8/HSC70) during PRRSV-2 infection. Knockdown of HSPA8 and LAMP2A impacted on the yield of PRRSV-2 progeny virus, implying that the virus utilizes RAB18 to promote CMA-mediated lipolysis. Importantly, we determined that the C-terminal domain (CTD) of HSPA8 could bind to the switch II domain of RAB18, and the CTD of PLIN2 was capable of associating with HSPA8, suggesting that HSPA8 facilitates the interaction between RAB18 and PLIN2 in the CMA process. In summary, our findings elucidate how PRRSV-2 hijacks CMA-mediated lipid metabolism through innate immune activation to enhance the yield of progeny virus, offering novel insights for the development of anti-PRRSV-2 treatments.
Collapse
Affiliation(s)
- Guo-Li Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Biochemistry and Nutrition, Zhengzhou, Henan Province, Ministry of Agriculture and Rural Affairs of the People’s Republic of China
- Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, Henan Province, China
| | - Ying-Qian Han
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Biochemistry and Nutrition, Zhengzhou, Henan Province, Ministry of Agriculture and Rural Affairs of the People’s Republic of China
- Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, Henan Province, China
| | - Bing-Qian Su
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Biochemistry and Nutrition, Zhengzhou, Henan Province, Ministry of Agriculture and Rural Affairs of the People’s Republic of China
- Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, Henan Province, China
| | - Hai-Shen Yu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Biochemistry and Nutrition, Zhengzhou, Henan Province, Ministry of Agriculture and Rural Affairs of the People’s Republic of China
- Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, Henan Province, China
| | - Shuang Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Biochemistry and Nutrition, Zhengzhou, Henan Province, Ministry of Agriculture and Rural Affairs of the People’s Republic of China
- Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, Henan Province, China
| | - Guo-Yu Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Zhengzhou, Henan Province, Ministry of Agriculture and Rural Affairs of the People’s Republic of China
- Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, Henan Province, China
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Biochemistry and Nutrition, Zhengzhou, Henan Province, Ministry of Agriculture and Rural Affairs of the People’s Republic of China
- Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, Henan Province, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan Province, China
| | - Fang Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, China
| | - Sheng-Li Ming
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Biochemistry and Nutrition, Zhengzhou, Henan Province, Ministry of Agriculture and Rural Affairs of the People’s Republic of China
- Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, Henan Province, China
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, Henan Province, China
- Key Laboratory of Animal Biochemistry and Nutrition, Zhengzhou, Henan Province, Ministry of Agriculture and Rural Affairs of the People’s Republic of China
- Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou, Henan Province, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, Henan Province, China
- International Joint Research Center of National Animal Immunology, Henan Agricultural University, Zhengzhou, Henan Province, China
- Longhu Advanced Immunization Laboratory, Zhengzhou, Henan Province, China
| |
Collapse
|
10
|
Vélez EJ, Schnebert S, Goguet M, Balbuena-Pecino S, Dias K, Beauclair L, Fontagné-Dicharry S, Véron V, Depincé A, Beaumatin F, Herpin A, Seiliez I. Chaperone-mediated autophagy protects against hyperglycemic stress. Autophagy 2024; 20:752-768. [PMID: 37798944 PMCID: PMC11062381 DOI: 10.1080/15548627.2023.2267415] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/26/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023] Open
Abstract
Chaperone-mediated autophagy (CMA) is a major pathway of lysosomal proteolysis critical for cellular homeostasis and metabolism, and whose defects have been associated with several human pathologies. While CMA has been well described in mammals, functional evidence has only recently been documented in fish, opening up new perspectives to tackle this function under a novel angle. Now we propose to explore CMA functions in the rainbow trout (RT, Oncorhynchus mykiss), a fish species recognized as a model organism of glucose intolerance and characterized by the presence of two paralogs of the CMA-limiting factor Lamp2A (lysosomal associated membrane protein 2A). To this end, we validated a fluorescent reporter (KFERQ-PA-mCherry1) previously used to track functional CMA in mammalian cells, in an RT hepatoma-derived cell line (RTH-149). We found that incubation of cells with high-glucose levels (HG, 25 mM) induced translocation of the CMA reporter to lysosomes and/or late endosomes in a KFERQ- and Lamp2A-dependent manner, as well as reduced its half-life compared to the control (5 mM), thus demonstrating increased CMA flux. Furthermore, we observed that activation of CMA upon HG exposure was mediated by generation of mitochondrial reactive oxygen species, and involving the antioxidant transcription factor Nfe2l2/Nrf2 (nfe2 like bZIP transcription factor 2). Finally, we demonstrated that CMA plays an important protective role against HG-induced stress, primarily mediated by one of the two RT Lamp2As. Together, our results provide unequivocal evidence for CMA activity existence in RT and highlight both the role and regulation of CMA during glucose-related metabolic disorders.Abbreviations: AREs: antioxidant response elements; CHC: α-cyano -4-hydroxycinnamic acid; Chr: chromosome; CMA: chaperone-mediated autophagy; CT: control; DMF: dimethyl fumarate; Emi: endosomal microautophagy; HG: high-glucose; HMOX1: heme oxygenase 1; H2O2: hydrogen peroxide; KFERQ: lysine-phenylalanine-glutamate-arginine-glutamine; LAMP1: lysosomal associated membrane protein 1; LAMP2A: lysosomal associated membrane protein 2A; MCC: Manders' correlation coefficient; Manders' correlation coefficient Mo: morpholino oligonucleotide; NAC: N-acetyl cysteine; NFE2L2/NRF2: NFE2 like bZIP transcription factor 2; PA-mCherry: photoactivable mCherry; PCC: Pearson's correlation coefficient; ROS: reactive oxygen species; RT: rainbow trout; siRNAs: small interfering RNAs; SOD: superoxide dismutase; Tsg101: tumor susceptibility 101; TTFA: 2-thenoyltrifluoroacetone; WGD: whole-genome duplication.
Collapse
Affiliation(s)
- Emilio J. Vélez
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Simon Schnebert
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Maxime Goguet
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Sara Balbuena-Pecino
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Karine Dias
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Linda Beauclair
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Stéphanie Fontagné-Dicharry
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Vincent Véron
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Alexandra Depincé
- INRAE, UR1037 Laboratory of Fish Physiology and Genomics, Campus de Beaulieu, Rennes, France
| | - Florian Beaumatin
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| | - Amaury Herpin
- INRAE, UR1037 Laboratory of Fish Physiology and Genomics, Campus de Beaulieu, Rennes, France
| | - Iban Seiliez
- Université de Pau et des Pays de l‘Adour, E2S UPPA, INRAE, UMR1419 Nutrition Métabolisme et Aquaculture, Saint-Pée-sur-Nivelle, France
| |
Collapse
|
11
|
Liu M, Li S, Yin M, Li Y, Chen J, Chen Y, Zhou Y, Li Q, Xu F, Dai C, Xia Y, Chen A, Lu D, Chen Z, Qian J, Ge J. Pinacidil ameliorates cardiac microvascular ischemia-reperfusion injury by inhibiting chaperone-mediated autophagy of calreticulin. Basic Res Cardiol 2024; 119:113-131. [PMID: 38168863 PMCID: PMC10837255 DOI: 10.1007/s00395-023-01028-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024]
Abstract
Calcium overload is the key trigger in cardiac microvascular ischemia-reperfusion (I/R) injury, and calreticulin (CRT) is a calcium buffering protein located in the endoplasmic reticulum (ER). Additionally, the role of pinacidil, an antihypertensive drug, in protecting cardiac microcirculation against I/R injury has not been investigated. Hence, this study aimed to explore the benefits of pinacidil on cardiac microvascular I/R injury with a focus on endothelial calcium homeostasis and CRT signaling. Cardiac vascular perfusion and no-reflow area were assessed using FITC-lectin perfusion assay and Thioflavin-S staining. Endothelial calcium homeostasis, CRT-IP3Rs-MCU signaling expression, and apoptosis were assessed by real-time calcium signal reporter GCaMP8, western blotting, and fluorescence staining. Drug affinity-responsive target stability (DARTS) assay was adopted to detect proteins that directly bind to pinacidil. The present study found pinacidil treatment improved capillary density and perfusion, reduced no-reflow and infraction areas, and improved cardiac function and hemodynamics after I/R injury. These benefits were attributed to the ability of pinacidil to alleviate calcium overload and mitochondria-dependent apoptosis in cardiac microvascular endothelial cells (CMECs). Moreover, the DARTS assay showed that pinacidil directly binds to HSP90, through which it inhibits chaperone-mediated autophagy (CMA) degradation of CRT. CRT overexpression inhibited IP3Rs and MCU expression, reduced mitochondrial calcium inflow and mitochondrial injury, and suppressed endothelial apoptosis. Importantly, endothelial-specific overexpression of CRT shared similar benefits with pinacidil on cardiovascular protection against I/R injury. In conclusion, our data indicate that pinacidil attenuated microvascular I/R injury potentially through improving CRT degradation and endothelial calcium overload.
Collapse
Affiliation(s)
- Muyin Liu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Su Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Ming Yin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Youran Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Jinxiang Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Yuqiong Chen
- Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Nanjing Medical University, Nanjing, China
| | - You Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Qiyu Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Fei Xu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chunfeng Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yan Xia
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Ao Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Danbo Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Zhangwei Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| |
Collapse
|
12
|
Krause GJ, Kirchner P, Stiller B, Morozova K, Diaz A, Chen KH, Krogan NJ, Agullo-Pascual E, Clement CC, Lindenau K, Swaney DL, Dilipkumar S, Bravo-Cordero JJ, Santambrogio L, Cuervo AM. Molecular determinants of the crosstalk between endosomal microautophagy and chaperone-mediated autophagy. Cell Rep 2023; 42:113529. [PMID: 38060380 PMCID: PMC10807933 DOI: 10.1016/j.celrep.2023.113529] [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: 01/11/2022] [Revised: 09/12/2023] [Accepted: 11/17/2023] [Indexed: 12/30/2023] Open
Abstract
Chaperone-mediated autophagy (CMA) and endosomal microautophagy (eMI) are pathways for selective degradation of cytosolic proteins in lysosomes and late endosomes, respectively. These autophagic processes share as a first step the recognition of the same five-amino-acid motif in substrate proteins by the Hsc70 chaperone, raising the possibility of coordinated activity of both pathways. In this work, we show the existence of a compensatory relationship between CMA and eMI and identify a role for the chaperone protein Bag6 in triage and internalization of eMI substrates into late endosomes. Association and dynamics of Bag6 at the late endosome membrane change during starvation, a stressor that, contrary to other autophagic pathways, causes a decline in eMI activity. Collectively, these results show a coordinated function of eMI with CMA, identify the interchangeable subproteome degraded by these pathways, and start to elucidate the molecular mechanisms that facilitate the switch between them.
Collapse
Affiliation(s)
- Gregory J Krause
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Philipp Kirchner
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Barbara Stiller
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kateryna Morozova
- Department of Radiation Oncology, Weill Cornell School of Medicine, New York, NY 10021, USA
| | - Antonio Diaz
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kuei-Ho Chen
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; The J. David Gladstone Institutes, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nevan J Krogan
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; The J. David Gladstone Institutes, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94158, USA
| | | | - Cristina C Clement
- Department of Radiation Oncology, Weill Cornell School of Medicine, New York, NY 10021, USA
| | - Kristen Lindenau
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Danielle L Swaney
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; The J. David Gladstone Institutes, San Francisco, CA 94158, USA; Quantitative Biosciences Institute (QBI), University of California, San Francisco, San Francisco, CA 94158, USA
| | - Shilpa Dilipkumar
- Microscopy CoRE, Dean's CoREs, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jose Javier Bravo-Cordero
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell School of Medicine, New York, NY 10021, USA.
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
13
|
Jonnavithula A, Tandar M, Umar M, Orton SN, Woodrum MC, Mookherjee S, Boudina S, Symons JD, Ghosh R. A Fluorogenic-Based Assay to Measure Chaperone-Mediated Autophagic Activity in Cells and Tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.14.571785. [PMID: 38168260 PMCID: PMC10760084 DOI: 10.1101/2023.12.14.571785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Objective Pathologies including cardiovascular diseases, cancer, and neurological disorders are caused by the accumulation of misfolded / damaged proteins. Intracellular protein degradation mechanisms play a critical role in the clearance of these disease-causing proteins. Chaperone mediated autophagy (CMA) is a protein degradation pathway that employs chaperones to bind proteins, bearing a unique KFERQ-like motif, for delivery to a CMA-specific Lysosome Associated Membrane Protein 2a (LAMP2a) receptor for lysosomal degradation. To date, steady-state CMA function has been assessed by measuring LAMP2A protein expression. However, this does not provide information regarding CMA degradation activity. To fill this dearth of tools / assays to measure CMA activity, we generated a CMA-specific fluorogenic substrate assay. Methods A KFERQ-AMC [Lys-Phe-Asp-Arg-Gln-AMC(7-amino-4-methylcou-marin)] fluorogenic CMA substrate was synthesized from Solid-Phase Peptide Synthesis. KFERQ-AMC, when cleaved via lysosomal hydrolysis, causes AMC to release and fluoresce (Excitation:355 nm, Emission:460 nm). Using an inhibitor of lysosomal proteases, i.e., E64D [L-trans-Epoxy-succinyl-leucylamido(4-guanidino)butane)], responsible for cleaving CMA substrates, the actual CMA activity was determined. Essentially, CMA activity = (substrate) fluorescence - (substrate+E64D) fluorescence . To confirm specificity of the KFERQ sequence for CMA, negative control peptides were used. Results Heart, liver, and kidney lysates containing intact lysosomes were obtained from 4-month-old adult male mice. First, lysates incubated with KFERQ-AMC displayed a time dependent (0-5 hour) increase in AMC fluorescence vs. lysates incubated with negative control peptides. These data validate the specificity of KFERQ for CMA. Of note, liver exhibited the highest CMA (6-fold; p<0.05) > kidney (2.4-fold) > heart (0.4-fold) at 5-hours. Second, E64D prevented KFERQ-AMC degradation, substantiating that KFERQ-AMC is degraded via lysosomes. Third, cleavage of KFERQ-AMC and resulting AMC fluorescence was inhibited in Human embryonic kidney (HEK) cells and H9c2 cardiac cells transfected with Lamp2a vs. control siRNA. Further, enhancing CMA using Lamp2a adenovirus upregulated KFERQ degradation. These data suggest that LAMP2A is required for KFERQ degradation. Conclusion. We have generated a novel assay for measuring CMA activity in cells and tissues in a variety of experimental contexts. Abstract Figure
Collapse
|
14
|
Asare PF, Hurtado PR, Tran HB, Perkins GB, Roscioli E, Hodge S. Reduction in Rubicon by cigarette smoke is associated with impaired phagocytosis and occurs through lysosomal degradation pathway. Clin Exp Med 2023; 23:4041-4055. [PMID: 37310658 DOI: 10.1007/s10238-023-01105-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 05/26/2023] [Indexed: 06/14/2023]
Abstract
BACKGROUND A common feature of COPD is a defective lung macrophage phagocytic capacity that can contribute to chronic lung inflammation and infection. The precise mechanisms remain incompletely understood, although cigarette smoke is a known contributor. We previously showed deficiency of the LC3-associated phagocytosis (LAP) regulator, Rubicon, in macrophages from COPD subjects and in response to cigarette smoke. The current study investigated the molecular basis by which cigarette smoke extract (CSE) reduces Rubicon in THP-1, alveolar and blood monocyte-derived macrophages, and the relationship between Rubicon deficiency and CSE-impaired phagocytosis. METHODOLOGY Phagocytic capacity of CSE-treated macrophages was measured by flow cytometry, Rubicon expression by Western blot and real time polymerase chain reaction, and autophagic-flux by LC3 and p62 levels. The effect of CSE on Rubicon degradation was determined using cycloheximide inhibition and Rubicon protein synthesis and half-life assessment. RESULTS Phagocytosis was significantly impaired in CSE-exposed macrophages and strongly correlated with Rubicon expression. CSE-impaired autophagy, accelerated Rubicon degradation, and reduced its half-life. Lysosomal protease inhibitors, but not proteasome inhibitors, attenuated this effect. Autophagy induction did not significantly affect Rubicon expression. CONCLUSIONS CSE decreases Rubicon through the lysosomal degradation pathway. Rubicon degradation and/or LAP impairment may contribute to dysregulated phagocytosis perpetuated by CSE.
Collapse
Affiliation(s)
- Patrick F Asare
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.
- Department of Thoracic Medicine, Faculty of Health and Medical Science, The University of Adelaide, Adelaide, Australia.
| | - Plinio R Hurtado
- School of Medicine, University of Adelaide, Adelaide, Australia
- Department of Renal Medicine, Royal Adelaide Hospital, Adelaide, Australia
| | - Hai B Tran
- School of Medicine, University of Adelaide, Adelaide, Australia
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia
| | - Griffith B Perkins
- Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Eugene Roscioli
- School of Medicine, University of Adelaide, Adelaide, Australia
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia
| | - Sandra Hodge
- School of Medicine, University of Adelaide, Adelaide, Australia
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia
| |
Collapse
|
15
|
Zhang KK, Burns CM, Skinner ME, Lombard DB, Miller RA, Endicott SJ. PTEN is both an activator and a substrate of chaperone-mediated autophagy. J Cell Biol 2023; 222:e202208150. [PMID: 37418003 PMCID: PMC10327811 DOI: 10.1083/jcb.202208150] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 04/17/2023] [Accepted: 05/26/2023] [Indexed: 07/08/2023] Open
Abstract
PTEN is a crucial negative regulator of the INS/PI3K/AKT pathway and is one of the most commonly mutated tumor suppressors in cancer. Global overexpression (OE) of PTEN in mice shifts metabolism to favor oxidative phosphorylation over glycolysis, reduces fat mass, and extends the lifespan of both sexes. We demonstrate that PTEN regulates chaperone-mediated autophagy (CMA). Using cultured cells and mouse models, we show that PTEN OE enhances CMA, dependent upon PTEN's lipid phosphatase activity and AKT inactivation. Reciprocally, PTEN knockdown reduces CMA, which can be rescued by inhibiting class I PI3K or AKT. Both PTEN and CMA are negative regulators of glycolysis and lipid droplet formation. We show that suppression of glycolysis and lipid droplet formation downstream of PTEN OE depends on CMA activity. Finally, we show that PTEN protein levels are sensitive to CMA and that PTEN accumulates in lysosomes with elevated CMA. Collectively, these data suggest that CMA is both an effector and a regulator of PTEN.
Collapse
Affiliation(s)
- Katherine K. Zhang
- College of Literature, Arts, and the Sciences, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
| | - Calvin M. Burns
- Department of Pathology, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
| | - Mary E. Skinner
- Department of Neurology, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
| | - David B. Lombard
- Department of Pathology and Laboratory Medicine, and Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Richard A. Miller
- Department of Pathology, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
- Geriatrics Center, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
| | - S. Joseph Endicott
- Department of Pathology, University of Michigan, Ann Arbor, Ann Arbor, MI, USA
| |
Collapse
|
16
|
Grochowska KM, Sperveslage M, Raman R, Failla AV, Głów D, Schulze C, Laprell L, Fehse B, Kreutz MR. Chaperone-mediated autophagy in neuronal dendrites utilizes activity-dependent lysosomal exocytosis for protein disposal. Cell Rep 2023; 42:112998. [PMID: 37590146 DOI: 10.1016/j.celrep.2023.112998] [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: 02/18/2023] [Revised: 06/16/2023] [Accepted: 08/01/2023] [Indexed: 08/19/2023] Open
Abstract
The complex morphology of neurons poses a challenge for proteostasis because the majority of lysosomal degradation machinery is present in the cell soma. In recent years, however, mature lysosomes were identified in dendrites, and a fraction of those appear to fuse with the plasma membrane and release their content to the extracellular space. Here, we report that dendritic lysosomes are heterogeneous in their composition and that only those containing lysosome-associated membrane protein (LAMP) 2A and 2B fuse with the membrane and exhibit activity-dependent motility. Exocytotic lysosomes dock in close proximity to GluN2B-containing N-methyl-D-aspartate-receptors (NMDAR) via an association of LAMP2B to the membrane-associated guanylate kinase family member SAP102/Dlg3. NMDAR-activation decreases lysosome motility and promotes membrane fusion. We find that chaperone-mediated autophagy is a supplier of content that is released to the extracellular space via lysosome exocytosis. This mechanism enables local disposal of aggregation-prone proteins like TDP-43 and huntingtin.
Collapse
Affiliation(s)
- Katarzyna M Grochowska
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany.
| | - Marit Sperveslage
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Rajeev Raman
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Antonio V Failla
- UKE Microscopic Imaging Facility (umif), University Medical Center Eppendorf, 20251 Hamburg, Germany
| | - Dawid Głów
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany
| | - Christian Schulze
- Institute of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Laura Laprell
- Institute of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany
| | - Michael R Kreutz
- Leibniz Group "Dendritic Organelles and Synaptic Function," Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany; Center for Behavioral Brain Sciences, Otto von Guericke University, 39120 Magdeburg, Germany; German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany.
| |
Collapse
|
17
|
Wang Y, Chen YY, Gao GB, Zheng YH, Yu NN, Ouyang L, Gao X, Li N, Wen SY, Huang S, Zhao Q, Liu L, Cao M, Zhang S, Zhang J, He QY. Polyphyllin D punctures hypertrophic lysosomes to reverse drug resistance of hepatocellular carcinoma by targeting acid sphingomyelinase. Mol Ther 2023; 31:2169-2187. [PMID: 37211762 PMCID: PMC10362416 DOI: 10.1016/j.ymthe.2023.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/13/2023] [Accepted: 05/18/2023] [Indexed: 05/23/2023] Open
Abstract
Hypertrophic lysosomes are critical for tumor progression and drug resistance; however, effective and specific lysosome-targeting compounds for cancer therapy are lacking. Here we conducted a lysosomotropic pharmacophore-based in silico screen in a natural product library (2,212 compounds), and identified polyphyllin D (PD) as a novel lysosome-targeted compound. PD treatment was found to cause lysosomal damage, as evidenced by the blockade of autophagic flux, loss of lysophagy, and the release of lysosomal contents, thus exhibiting anticancer effects on hepatocellular carcinoma (HCC) cell both in vitro and in vivo. Closer mechanistic examination revealed that PD suppressed the activity of acid sphingomyelinase (SMPD1), a lysosomal phosphodieserase that catalyzes the hydrolysis of sphingomyelin to produce ceramide and phosphocholine, by directly occupying its surface groove, with Trp148 in SMPD1 acting as a major binding residue; this suppression of SMPD1 activity irreversibly triggers lysosomal injury and initiates lysosome-dependent cell death. Furthermore, PD-enhanced lysosomal membrane permeabilization to release sorafenib, augmenting the anticancer effect of sorafenib both in vivo and in vitro. Overall, our study suggests that PD can potentially be further developed as a novel autophagy inhibitor, and a combination of PD with classical chemotherapeutic anticancer drugs could represent a novel therapeutic strategy for HCC intervention.
Collapse
Affiliation(s)
- Yang Wang
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| | - Yan-Yan Chen
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Gui-Bin Gao
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yang-Han Zheng
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Nan-Nan Yu
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Lan Ouyang
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xuejuan Gao
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Nan Li
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Shi-Yuan Wen
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Shangjia Huang
- MOE Key Laboratory of Tumor Molecular Biology, The First Affiliated Hospital of Jinan University, Guangzhou 510613, China
| | - Qian Zhao
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Langxia Liu
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Mingrong Cao
- Department of General Surgery, The First Affiliated Hospital, Jinan University, Guangzhou 510613, China
| | - Shuixing Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou 510613, China; MOE Key Laboratory of Tumor Molecular Biology, The First Affiliated Hospital of Jinan University, Guangzhou 510613, China.
| | - Jing Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou 510613, China; MOE Key Laboratory of Tumor Molecular Biology, The First Affiliated Hospital of Jinan University, Guangzhou 510613, China.
| | - Qing-Yu He
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; MOE Key Laboratory of Tumor Molecular Biology, The First Affiliated Hospital of Jinan University, Guangzhou 510613, China.
| |
Collapse
|
18
|
Binder MJ, Pedley AM. The roles of molecular chaperones in regulating cell metabolism. FEBS Lett 2023; 597:1681-1701. [PMID: 37287189 PMCID: PMC10984649 DOI: 10.1002/1873-3468.14682] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/09/2023]
Abstract
Fluctuations in nutrient and biomass availability, often as a result of disease, impart metabolic challenges that must be overcome in order to sustain cell survival and promote proliferation. Cells adapt to these environmental changes and stresses by adjusting their metabolic networks through a series of regulatory mechanisms. Our understanding of these rewiring events has largely been focused on those genetic transformations that alter protein expression and the biochemical mechanisms that change protein behavior, such as post-translational modifications and metabolite-based allosteric modulators. Mounting evidence suggests that a class of proteome surveillance proteins called molecular chaperones also can influence metabolic processes. Here, we summarize several ways the Hsp90 and Hsp70 chaperone families act on human metabolic enzymes and their supramolecular assemblies to change enzymatic activities and metabolite flux. We further highlight how these chaperones can assist in the translocation and degradation of metabolic enzymes. Collectively, these studies provide a new view for how metabolic processes are regulated to meet cellular demand and inspire new avenues for therapeutic intervention.
Collapse
|
19
|
Byun JK, Lee SH, Moon EJ, Park MH, Jang H, Weitzel DH, Kim HH, Basnet N, Kwon DY, Lee CT, Stephenson TN, Jeong JH, Patel BA, Park SJ, Chi JT, Dewhirst MW, Hong J, Lee YM. Manassantin A inhibits tumour growth under hypoxia through the activation of chaperone-mediated autophagy by modulating Hsp90 activity. Br J Cancer 2023; 128:1491-1502. [PMID: 36759727 PMCID: PMC10070431 DOI: 10.1038/s41416-023-02148-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Chaperon-mediated autophagy (CMA) has taken on a new emphasis in cancer biology. However, the roles of CMA in hypoxic tumours are poorly understood. We investigated the anti-tumour effects of the natural product ManA through the activation of CMA in tumour progression under hypoxia. METHODS The effect of ManA on CMA activation was assessed in mouse xenograft models and cells. The gene expressions of HIF-1α, HSP90AA1, and transcription factor EB (TFEB) were analysed using The Cancer Genome Atlas (TCGA) datasets to assess the clinical relevance of CMA. RESULTS ManA activates photoswitchable CMA reporter activity and inhibits Hsp90 chaperone function by disrupting the Hsp90/F1F0-ATP synthase complex. Hsp90 inhibition enhances the interaction between CMA substrates and LAMP-2A and TFEB nuclear localisation, suggesting CMA activation by ManA. ManA-activated CMA retards tumour growth and displays cooperative anti-tumour activity with anti-PD-1 antibody. TCGA datasets show that a combined expression of HSP90AA1High/HIF1AHigh or TFEBLow/HIF1AHigh is strongly correlated with poor prognosis in patients with lung cancer. CONCLUSIONS ManA-induced CMA activation by modulating Hsp90 under hypoxia induces HIF-1α degradation and reduces tumour growth. Thus, inducing CMA activity by targeting Hsp90 may be a promising therapeutic strategy against hypoxic tumours.
Collapse
Affiliation(s)
- Jun-Kyu Byun
- College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Buk-gu, 41566, Daegu, Republic of Korea
| | - Sun Hee Lee
- College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Buk-gu, 41566, Daegu, Republic of Korea
- Vessel-Organ Interaction Research Center, VOICE (MRC), Kyungpook National University, 80 Daehak-ro, Buk-gu, 41566, Daegu, Republic of Korea
| | - Eui Jung Moon
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Headington, OX3 7DQ, UK
| | - Myo-Hyeon Park
- College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Buk-gu, 41566, Daegu, Republic of Korea
- Vessel-Organ Interaction Research Center, VOICE (MRC), Kyungpook National University, 80 Daehak-ro, Buk-gu, 41566, Daegu, Republic of Korea
| | - Hyeonha Jang
- College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Buk-gu, 41566, Daegu, Republic of Korea
- Vessel-Organ Interaction Research Center, VOICE (MRC), Kyungpook National University, 80 Daehak-ro, Buk-gu, 41566, Daegu, Republic of Korea
| | - Douglas H Weitzel
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Hyun-Hwi Kim
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Nikita Basnet
- College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Buk-gu, 41566, Daegu, Republic of Korea
| | - Do-Yeon Kwon
- Department of Chemistry, Duke University, Durham, NC, 27708, USA
| | - Chen-Ting Lee
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | | | - Ji-Hak Jeong
- College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Buk-gu, 41566, Daegu, Republic of Korea
- Vessel-Organ Interaction Research Center, VOICE (MRC), Kyungpook National University, 80 Daehak-ro, Buk-gu, 41566, Daegu, Republic of Korea
| | - Bhargav A Patel
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, IN, 46556, USA
| | - Sung Jean Park
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, 21936, Republic of Korea
| | - Jen-Tsan Chi
- Department of Molecular Genetics & Microbiology, Duke University Medical Center, Durham, NC, 27710, USA
- Center for Genomic and Computation Biology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Mark W Dewhirst
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Jiyong Hong
- Department of Chemistry, Duke University, Durham, NC, 27708, USA.
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, USA.
| | - You Mie Lee
- College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Buk-gu, 41566, Daegu, Republic of Korea.
- Vessel-Organ Interaction Research Center, VOICE (MRC), Kyungpook National University, 80 Daehak-ro, Buk-gu, 41566, Daegu, Republic of Korea.
| |
Collapse
|
20
|
Xiong Q, Sun H, Xing W, Li X, Chen G, Zhao Z, Wu C, Li P. WDR45 mutation dysregulates iron homeostasis by promoting the chaperone-mediated autophagic degradation of ferritin heavy chain in an ER stress/p38 dependent mechanism. Free Radic Biol Med 2023; 201:89-97. [PMID: 36940732 DOI: 10.1016/j.freeradbiomed.2023.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/23/2023]
Abstract
Ferritin is the main iron storage protein that plays a pivotal role in the regulation of iron homeostasis. Mutations in the autophagy protein WD repeat domain 45 (WDR45) that lead to iron overload is associated with the human β-propeller protein-associated neurodegeneration (BPAN). Previous studies have demonstrated that ferritin was decreased in WDR45 deficient cells, but the mechanism remains unclear. In this study, we have demonstrated that the ferritin heavy chain (FTH) could be degraded via chaperone-mediated autophagy (CMA) in ER stress/p38-dependent pathway. In HeLa cells, inducing the ER stress activated CMA, therefore facilitated the degradation of FTH, and increased the content of Fe2+. However, the increased CMA activity and Fe2+ as well as the decreased FTH by ER stress inducer were restored by pre-treatment with p38 inhibitor. Overexpression of a mutant WDR45 activated CMA thus promoted the degradation of FTH. Furthermore, inhibition of ER stress/p38 pathway resulted in reduced activity of CMA, which consequently elevated the protein level of FTH but reduced the Fe2+ level. Our results revealed that WDR45 mutation dysregulates iron homeostasis by activating CMA, and promotes FTH degradation through ER stress/p38 signaling pathway.
Collapse
Affiliation(s)
- Qiuhong Xiong
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, 030006, China.
| | - Huimin Sun
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, 030006, China
| | - Wenxiu Xing
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, 030006, China
| | - Xin Li
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, 030006, China
| | - Guangxin Chen
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, 030006, China
| | - Zhonghua Zhao
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, 030006, China
| | - Changxin Wu
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, 030006, China.
| | - Ping Li
- Institutes of Biomedical Sciences, Shanxi Provincial Key Laboratory for Medical Molecular Cell Biology, Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, Taiyuan, 030006, China.
| |
Collapse
|
21
|
Li Q, Yue T, Du X, Tang Z, Cui J, Wang W, Xia W, Ren B, Kan S, Li C, Wu C, Niu X, Li B, Lin K, Luo J, Chen G, Wang Z. HSC70 mediated autophagic degradation of oxidized PRL2 is responsible for osteoclastogenesis and inflammatory bone destruction. Cell Death Differ 2023; 30:647-659. [PMID: 36182990 PMCID: PMC9984420 DOI: 10.1038/s41418-022-01068-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 02/25/2023] Open
Abstract
Inflammation leads to systemic osteoporosis or local bone destruction, however, the underlying molecular mechanisms are still poorly understood. In this study, we report that PRL2 is a negative regulator of osteoclastogenesis and bone absorption. Mice with PRL2 deficiency exhibit a decrease in bone volume and an increase in osteoclast numbers. PRL2 negatively regulates RANKL-induced reactive oxygen species production through the activation of RAC1, thus PRL2 deficient osteoclast precursors have both increased osteoclast differentiation ability and bone resorptive capacity. During inflammation, oxidized PRL2 is a selected substrate of HSC70 and conditions of oxidative stress trigger rapid degradation of PRL2 by HSC70 mediated endosomal microautophagy and chaperone-mediated autophagy. Ablation of PRL2 in mouse models of inflammatory bone disease leads to an increase in the number of osteoclasts and exacerbation of bone damage. Moreover, reduced PRL2 protein levels in peripheral myeloid cells are highly correlated with bone destruction in a mouse arthritis model and in human rheumatoid arthritis, while the autophagy inhibitor hydroxychloroquine blocked inflammation-induced PRL2 degradation and bone destruction in vivo. Therefore, our findings identify PRL2 as a new regulator in osteoimmunity, providing a link between inflammation and osteoporosis. As such, PRL2 is a potential therapeutic target for inflammatory bone disease and inhibition of HSC70 mediated autophagic degradation of PRL2 may offer new therapeutic tools for the treatment of inflammatory bone disease.
Collapse
Affiliation(s)
- Qi Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tao Yue
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200052, China
| | - Xinyue Du
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zaiming Tang
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jinjie Cui
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Weifeng Wang
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200052, China
| | - Wenjie Xia
- Department of Rheumatology, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200052, China
| | - Baiyang Ren
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shuo Kan
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chang Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chenyun Wu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyin Niu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Bin Li
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Kaili Lin
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Jian Luo
- Yangzhi Rehabilitation Hospital (Sunshine Rehabilitation Center), Tongji University School of Medicine, Shanghai, 201619, China
| | - Guangjie Chen
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Zhaojun Wang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Key Laboratory of Parasite and Vector Biology, Ministry of Health, China; School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
22
|
Nikesitch N, Beraldi E, Zhang F, Adomat H, Bell R, Suzuki K, Fazli L, Hy Kung S, Wells C, Pinette N, Saxena N, Wang Y, Gleave M. Chaperone-mediated autophagy promotes PCa survival during ARPI through selective proteome remodeling. Oncogene 2023; 42:748-758. [PMID: 36611121 DOI: 10.1038/s41388-022-02573-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/05/2022] [Accepted: 12/07/2022] [Indexed: 01/09/2023]
Abstract
The androgen receptor (AR) plays an important role in PCa metabolism, with androgen receptor pathway inhibition (ARPI) subjecting PCa cells to acute metabolic stress caused by reduced biosynthesis and energy production. Defining acute stress response mechanisms that alleviate ARPI stress and therefore mediate prostate cancer (PCa) treatment resistance will help improve therapeutic outcomes of patients treated with ARPI. We identified the up-regulation of chaperone-mediated autophagy (CMA) in response to acute ARPI stress, which persisted in castration-resistant PCa (CRPC); previously undefined in PCa. CMA is a selective protein degradation pathway and a key stress response mechanism up-regulated under several stress stimuli, including metabolic stress. Through selective protein degradation, CMA orchestrates the cellular stress response by regulating cellular pathways through selective proteome remodeling. Through broad-spectrum proteomic analysis, CMA coordinates metabolic reprogramming of PCa cells to sustain PCa growth and survival during ARPI; through the upregulation of mTORC1 signaling and pathways associated with PCa biosynthesis and energetics. This not only promoted PCa growth during ARPI, but also promoted the emergence of CRPC in-vivo. During CMA inhibition, PCa metabolism is compromised, leading to ATP depletion, resulting in a profound anti-proliferative effect on PCa cells, and is enhanced when combined with ARPI. Furthermore, CMA inhibition prevented in-vivo tumour formation, and also re-sensitized enzalutamide-resistant cell lines in-vitro. The profound anti-proliferative effect of CMA inhibition was attributed to cell cycle arrest mediated through p53 transcriptional repression of E2F target genes. In summary, CMA is an acute ARPI stress response mechanism, essential in alleviating ARPI induced metabolic stress, essential for ensuring PCa growth and survival. CMA plays a critical role in the development of ARPI resistance in PCa.
Collapse
Affiliation(s)
- Nicholas Nikesitch
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Eliana Beraldi
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Fan Zhang
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Hans Adomat
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Robert Bell
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Kotaro Suzuki
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ladan Fazli
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Sonia Hy Kung
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Christopher Wells
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Nicholas Pinette
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Neetu Saxena
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Martin Gleave
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada.
- Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
23
|
Su CM, Hsu TW, Chen HA, Wang WY, Huang CY, Hung CC, Yeh MH, Su YH, Huang MT, Liao PH. Chaperone-mediated autophagy degrade Dicer to promote breast cancer metastasis. J Cell Physiol 2023; 238:829-841. [PMID: 36815383 DOI: 10.1002/jcp.30979] [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: 06/01/2022] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 02/24/2023]
Abstract
Metastasis in breast cancer usually lead to the majority of deaths on clinical patients. Accordingly, diagnosis of metastasis at the early stage in breast cancer is important to improve the prognosis. We observed that Dicer protein levels are significant decrease in highly invasive breast cancer cells and usually correlated with poor clinical outcomes. Following, we aim to clarify the molecular regulatory mechanism of this phenomenon in breast cancer to provide a new therapeutic target. In this study, we obtained that Dicer expression correlated with metastasis and invasion without affect cell stability in breast cancer cells. Importantly, we identified the regulatory mechanism of Dicer protein degradation, the chaperone-mediated autophagy (CMA)-mediated degradation that is major mechanism to decrease Dicer protein expression and lead to cancer metastasis. We discovered that heat shock cognate 71-kDa protein (Hsc70) which as a CMA-related factor interacts with the CMA-targeting motif I333A/K334A on Dicer to promote degradation through CMA. Taken together, our findings hint that Dicer highly correlated with cancer metastasis, we reveal the tumor-promoting effect of CMA-mediated Dicer degradation in breast cancer.
Collapse
Affiliation(s)
- Chih-Ming Su
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan
| | - Tung-Wei Hsu
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsin-An Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wan-Yu Wang
- Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Hualien, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung City, Taichung, Taiwan
| | - Chih-Chiang Hung
- Division of Breast Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Applied Cosmetology, College of Human Science and Social Innovation, Hungkuang University, Taichung, Taiwan
| | - Ming-Hsin Yeh
- Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan.,Institute of Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yen-Hao Su
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ming-Te Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan.,Division of General Surgery, Department of Surgery, Xin Tai General Hospital, New Taipei, Taiwan
| | - Po-Hsiang Liao
- Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan
| |
Collapse
|
24
|
Rahman MA, Engelsen AST, Sarowar S, Bindesbøll C, Birkeland E, Goplen D, Lotsberg ML, Knappskog S, Simonsen A, Chekenya M. Bortezomib abrogates temozolomide-induced autophagic flux through an ATG5 dependent pathway. Front Cell Dev Biol 2022; 10:1022191. [PMID: 36619857 PMCID: PMC9814514 DOI: 10.3389/fcell.2022.1022191] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction: Glioblastoma (GBM) is invariably resistant to temozolomide (TMZ) chemotherapy. Inhibiting the proteasomal pathway is an emerging strategy to accumulate damaged proteins and inhibit their lysosomal degradation. We hypothesized that pre-treatment of glioblastoma with bortezomib (BTZ) might sensitize glioblastoma to temozolomide by abolishing autophagy survival signals to augment DNA damage and apoptosis. Methods: P3 patient-derived glioblastoma cells, as well as the tumour cell lines U87, HF66, A172, and T98G were investigated for clonogenic survival after single or combined treatment with temozolomide and bortezomib in vitro. We investigated the requirement of functional autophagy machinery by utilizing pharmacological inhibitors or CRISPR-Cas9 knockout (KO) of autophagy-related genes -5 and -7 (ATG5 and ATG7) in glioblastoma cells and monitored changes in autophagic flux after temozolomide and/or bortezomib treatments. P3 wild-type and P3 ATG5-/- (ATG5 KO) cells were implanted orthotopically into NOD-SCID mice to assess the efficacy of bortezomib and temozolomide combination therapy with and without functional autophagy machinery. Results: The chemo-resistant glioblastoma cells increased autophagic flux during temozolomide treatment as indicated by increased degradation of long-lived proteins, diminished expression of autophagy markers LC3A/B-II and p62 (SQSTM1), increased co-localisation of LC3A/B-II with STX17, augmented and no induction of apoptosis. In contrast, bortezomib treatment abrogated autophagic flux indicated by the accumulation of LC3A/B-II and p62 (SQSTM1) positive autophagosomes that did not fuse with lysosomes and thus reduced the degradation of long-lived proteins. Bortezomib synergistically enhanced temozolomide efficacy by attenuating cell proliferation, increased DNA double-strand breaks, and apoptosis in an autophagy-dependent manner. Abolishing autophagy in ATG5 KOs reversed the bortezomib-induced toxicity, rescued glioblastoma cell death and reduced animal survival. Discussion: We conclude that bortezomib abrogates temozolomide induced autophagy flux through an ATG5 dependent pathway.
Collapse
Affiliation(s)
- Mohummad Aminur Rahman
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway,Department of Oncology, Haukeland University Hospital, Bergen, Norway,*Correspondence: Mohummad Aminur Rahman,
| | - Agnete S. T. Engelsen
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway,Department of Clinical Medicine and Centre for Cancer Biomarkers, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Shahin Sarowar
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Christian Bindesbøll
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Even Birkeland
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Dorota Goplen
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Maria L. Lotsberg
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway,Department of Clinical Medicine and Centre for Cancer Biomarkers, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Stian Knappskog
- Department of Oncology, Haukeland University Hospital, Bergen, Norway,Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway,Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Martha Chekenya
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
25
|
Kaushik S, Juste YR, Lindenau K, Dong S, Macho-González A, Santiago-Fernández O, McCabe M, Singh R, Gavathiotis E, Cuervo AM. Chaperone-mediated autophagy regulates adipocyte differentiation. SCIENCE ADVANCES 2022; 8:eabq2733. [PMID: 36383673 PMCID: PMC9668314 DOI: 10.1126/sciadv.abq2733] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 09/28/2022] [Indexed: 06/16/2023]
Abstract
Adipogenesis is a tightly orchestrated multistep process wherein preadipocytes differentiate into adipocytes. The most studied aspect of adipogenesis is its transcriptional regulation through timely expression and silencing of a vast number of genes. However, whether turnover of key regulatory proteins per se controls adipogenesis remains largely understudied. Chaperone-mediated autophagy (CMA) is a selective form of lysosomal protein degradation that, in response to diverse cues, remodels the proteome for regulatory purposes. We report here the activation of CMA during adipocyte differentiation and show that CMA regulates adipogenesis at different steps through timely degradation of key regulatory signaling proteins and transcription factors that dictate proliferation, energetic adaptation, and signaling changes required for adipogenesis.
Collapse
Affiliation(s)
- Susmita Kaushik
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yves R. Juste
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kristen Lindenau
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Shuxian Dong
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Adrián Macho-González
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Olaya Santiago-Fernández
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mericka McCabe
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rajat Singh
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Evripidis Gavathiotis
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
26
|
Rovira M, Sereda R, Pladevall‐Morera D, Ramponi V, Marin I, Maus M, Madrigal‐Matute J, Díaz A, García F, Muñoz J, Cuervo AM, Serrano M. The lysosomal proteome of senescent cells contributes to the senescence secretome. Aging Cell 2022; 21:e13707. [PMID: 36087066 PMCID: PMC9577959 DOI: 10.1111/acel.13707] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/01/2022] [Accepted: 08/13/2022] [Indexed: 01/31/2023] Open
Abstract
Senescent cells accumulate in tissues over time, favoring the onset and progression of multiple age-related diseases. Senescent cells present a remarkable increase in lysosomal mass and elevated autophagic activity. Here, we report that two main autophagic pathways macroautophagy (MA) and chaperone-mediated autophagy (CMA) are constitutively upregulated in senescent cells. Proteomic analyses of the subpopulations of lysosomes preferentially engaged in each of these types of autophagy revealed profound quantitative and qualitative changes in senescent cells, affecting both lysosomal resident proteins and cargo proteins delivered to lysosomes for degradation. These studies have led us to identify resident lysosomal proteins that are highly augmented in senescent cells and can be used as novel markers of senescence, such as arylsulfatase ARSA. The abundant secretome of senescent cells, known as SASP, is considered their main pathological mediator; however, little is known about the mechanisms of SASP secretion. Some secretory cells, including melanocytes, use the small GTPase RAB27A to perform lysosomal secretion. We found that this process is exacerbated in the case of senescent melanoma cells, as revealed by the exposure of lysosomal membrane integral proteins LAMP1 and LAMP2 in their plasma membrane. Interestingly, a subset of SASP components, including cytokines CCL2, CCL3, CXCL12, cathepsin CTSD, or the protease inhibitor SERPINE1, are secreted in a RAB27A-dependent manner in senescent melanoma cells. Finally, proteins previously identified as plasma biomarkers of aging are highly enriched in the lysosomes of senescent cells, including CTSD. We conclude that the lysosomal proteome of senescent cells is profoundly reconfigured, and that some senescent cells can be highly active in lysosomal exocytosis.
Collapse
Affiliation(s)
- Miguel Rovira
- Cellular Plasticity and Disease GroupInstitute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Rebecca Sereda
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineNew YorkNew YorkUSA
- Institute for Aging StudiesAlbert Einstein College of MedicineNew YorkNew YorkUSA
| | - David Pladevall‐Morera
- Department of Cellular and Molecular Medicine, Center for Chromosome Stability and Center for Healthy AgingUniversity of CopenhagenCopenhagenDenmark
| | - Valentina Ramponi
- Cellular Plasticity and Disease GroupInstitute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Ines Marin
- Cellular Plasticity and Disease GroupInstitute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Mate Maus
- Cellular Plasticity and Disease GroupInstitute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
| | - Julio Madrigal‐Matute
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineNew YorkNew YorkUSA
- Institute for Aging StudiesAlbert Einstein College of MedicineNew YorkNew YorkUSA
- Instituto Biomédico de Nutrición y SaludEldaSpain
| | - Antonio Díaz
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineNew YorkNew YorkUSA
- Institute for Aging StudiesAlbert Einstein College of MedicineNew YorkNew YorkUSA
| | - Fernando García
- Proteomics UnitSpanish National Cancer Research Center (CNIO)MadridSpain
| | - Javier Muñoz
- Proteomics UnitSpanish National Cancer Research Center (CNIO)MadridSpain
- Biocruces Bizkaia Health Research InstituteBarakaldoSpain
- Ikerbasque, Basque Foundation for ScienceBilbaoSpain
| | - Ana María Cuervo
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineNew YorkNew YorkUSA
- Institute for Aging StudiesAlbert Einstein College of MedicineNew YorkNew YorkUSA
| | - Manuel Serrano
- Cellular Plasticity and Disease GroupInstitute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST)BarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
| |
Collapse
|
27
|
Chaudhry N, Sica M, Surabhi S, Hernandez DS, Mesquita A, Selimovic A, Riaz A, Lescat L, Bai H, MacIntosh GC, Jenny A. Lamp1 mediates lipid transport, but is dispensable for autophagy in Drosophila. Autophagy 2022; 18:2443-2458. [PMID: 35266854 PMCID: PMC9542896 DOI: 10.1080/15548627.2022.2038999] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 01/31/2022] [Accepted: 02/03/2022] [Indexed: 01/03/2023] Open
Abstract
The endolysosomal system not only is an integral part of the cellular catabolic machinery that processes and recycles nutrients for synthesis of biomaterials, but also acts as signaling hub to sense and coordinate the energy state of cells with growth and differentiation. Lysosomal dysfunction adversely influences vesicular transport-dependent macromolecular degradation and thus causes serious problems for human health. In mammalian cells, loss of the lysosome associated membrane proteins LAMP1 and LAMP2 strongly affects autophagy and cholesterol trafficking. Here we show that the previously uncharacterized Drosophila Lamp1 is a bona fide ortholog of vertebrate LAMP1 and LAMP2. Surprisingly and in contrast to lamp1 lamp2 double-mutant mice, Drosophila Lamp1 is not required for viability or autophagy, suggesting that fly and vertebrate LAMP proteins acquired distinct functions, or that autophagy defects in lamp1 lamp2 mutants may have indirect causes. However, Lamp1 deficiency results in an increase in the number of acidic organelles in flies. Furthermore, we find that Lamp1 mutant larvae have defects in lipid metabolism as they show elevated levels of sterols and diacylglycerols (DAGs). Because DAGs are the main lipid species used for transport through the hemolymph (blood) in insects, our results indicate broader functions of Lamp1 in lipid transport. Our findings make Drosophila an ideal model to study the role of LAMP proteins in lipid assimilation without the confounding effects of their storage and without interfering with autophagic processes.Abbreviations: aa: amino acid; AL: autolysosome; AP: autophagosome; APGL: autophagolysosome; AV: autophagic vacuole (i.e. AP and APGL/AL); AVi: early/initial autophagic vacuoles; AVd: late/degradative autophagic vacuoles; Atg: autophagy-related; CMA: chaperone-mediated autophagy; Cnx99A: Calnexin 99A; DAG: diacylglycerol; eMI: endosomal microautophagy; ESCRT: endosomal sorting complexes required for transport; FB: fat body; HDL: high-density lipoprotein; Hrs: Hepatocyte growth factor regulated tyrosine kinase substrate; LAMP: lysosomal associated membrane protein; LD: lipid droplet; LDL: low-density lipoprotein; Lpp: lipophorin; LTP: Lipid transfer particle; LTR: LysoTracker Red; MA: macroautophagy; MCC: Manders colocalization coefficient; MEF: mouse embryonic fibroblast MTORC: mechanistic target of rapamycin kinase complex; PV: parasitophorous vacuole; SNARE: soluble N-ethylmaleimide sensitive factor attachment protein receptor; Snap: Synaptosomal-associated protein; st: starved; TAG: triacylglycerol; TEM: transmission electron microscopy; TFEB/Mitf: transcription factor EB; TM: transmembrane domain; tub: tubulin; UTR: untranslated region.
Collapse
Affiliation(s)
- Norin Chaudhry
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, USA
| | - Margaux Sica
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, USA
| | - Satya Surabhi
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, USA
| | | | - Ana Mesquita
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, USA
| | - Adem Selimovic
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, USA
| | - Ayesha Riaz
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, USA
| | - Laury Lescat
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, USA
| | - Hua Bai
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, USA
| | - Gustavo C. MacIntosh
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, USA
| | - Andreas Jenny
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, USA
- Department of Genetics, Albert Einstein College of MedicineNew York, NY, USA
| |
Collapse
|
28
|
Modulating Chaperone-Mediated Autophagy and Its Clinical Applications in Cancer. Cells 2022; 11:cells11162562. [PMID: 36010638 PMCID: PMC9406970 DOI: 10.3390/cells11162562] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/14/2022] [Accepted: 08/15/2022] [Indexed: 11/25/2022] Open
Abstract
Autophagy is a central mechanism for maintaining cellular homeostasis in health and disease as it provides the critical energy through the breakdown and recycling of cellular components and molecules within lysosomes. One of the three types of autophagy is chaperone-mediated autophagy (CMA), a degradation pathway selective for soluble cytosolic proteins that contain a targeting motif related to KFERQ in their amino acid sequence. This motif marks them as CMA substrate and is, in the initial step of CMA, recognised by the heat shock protein 70 (Hsc70). The protein complex is then targeted to the lysosomal membrane where the interaction with the splice variant A of the lysosomal-associated membrane protein-2 (LAMP-2A) results in its unfolding and translocation into the lysosome for degradation. Altered levels of CMA have been reported in a wide range of pathologies including many cancer types that upregulate CMA as part of the pro-tumorigenic phenotype, while in aging a decline is observed and associated with a decrease of LAMP-2 expression. The potential of altering CMA to modify a physiological or pathological process has been firmly established through genetic manipulation in animals and chemical interference with this pathway. However, its use for therapeutic purposes has remained limited. Compounds used to target and modify CMA have been applied successfully to gain a better understanding of its cellular mechanisms, but they are mostly not specific, also influence other autophagic pathways and are associated with high levels of toxicity. Here, we will focus on the molecular mechanisms involved in CMA regulation as well as on potential ways to intersect them, describe modulators successfully used, their mechanism of action and therapeutic potential. Furthermore, we will discuss the potential benefits and drawbacks of CMA modulation in diseases such as cancer.
Collapse
|
29
|
Gómez-Virgilio L, Silva-Lucero MDC, Flores-Morelos DS, Gallardo-Nieto J, Lopez-Toledo G, Abarca-Fernandez AM, Zacapala-Gómez AE, Luna-Muñoz J, Montiel-Sosa F, Soto-Rojas LO, Pacheco-Herrero M, Cardenas-Aguayo MDC. Autophagy: A Key Regulator of Homeostasis and Disease: An Overview of Molecular Mechanisms and Modulators. Cells 2022; 11:cells11152262. [PMID: 35892559 PMCID: PMC9329718 DOI: 10.3390/cells11152262] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 01/18/2023] Open
Abstract
Autophagy is a highly conserved lysosomal degradation pathway active at basal levels in all cells. However, under stress conditions, such as a lack of nutrients or trophic factors, it works as a survival mechanism that allows the generation of metabolic precursors for the proper functioning of the cells until the nutrients are available. Neurons, as post-mitotic cells, depend largely on autophagy to maintain cell homeostasis to get rid of damaged and/or old organelles and misfolded or aggregated proteins. Therefore, the dysfunction of this process contributes to the pathologies of many human diseases. Furthermore, autophagy is highly active during differentiation and development. In this review, we describe the current knowledge of the different pathways, molecular mechanisms, factors that induce it, and the regulation of mammalian autophagy. We also discuss its relevant role in development and disease. Finally, here we summarize several investigations demonstrating that autophagic abnormalities have been considered the underlying reasons for many human diseases, including liver disease, cardiovascular, cerebrovascular diseases, neurodegenerative diseases, neoplastic diseases, cancers, and, more recently, infectious diseases, such as SARS-CoV-2 caused COVID-19 disease.
Collapse
Affiliation(s)
- Laura Gómez-Virgilio
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
| | - Maria-del-Carmen Silva-Lucero
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
| | - Diego-Salvador Flores-Morelos
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Guerrero, Mexico;
| | - Jazmin Gallardo-Nieto
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
- Biotechnology Engeniering, Universidad Politécnica de Quintana Roo, Cancún 77500, Quintana Roo, Mexico
| | - Gustavo Lopez-Toledo
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
| | - Arminda-Mercedes Abarca-Fernandez
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
- Biotechnology Engeniering, Universidad Politécnica de Quintana Roo, Cancún 77500, Quintana Roo, Mexico
| | - Ana-Elvira Zacapala-Gómez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39070, Guerrero, Mexico;
| | - José Luna-Muñoz
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlan Izcalli 53150, Estado de México, Mexico; (J.L.-M.); (F.M.-S.)
- Banco Nacional de Cerebros-UNPHU, Universidad Nacional Pedro Henríquez Ureña, Santo Domingo 11805, Dominican Republic
| | - Francisco Montiel-Sosa
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlan Izcalli 53150, Estado de México, Mexico; (J.L.-M.); (F.M.-S.)
| | - Luis O. Soto-Rojas
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de México, Mexico;
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de México, Mexico
| | - Mar Pacheco-Herrero
- Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Católica Madre y Maestra, Santiago de los Caballeros 51000, Dominican Republic;
| | - Maria-del-Carmen Cardenas-Aguayo
- Laboratory of Cellular Reprogramming, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico; (L.G.-V.); (M.-d.-C.S.-L.); (D.-S.F.-M.); (J.G.-N.); (G.L.-T.); (A.-M.A.-F.)
- Correspondence: ; Tel.: +52-55-2907-0937
| |
Collapse
|
30
|
Gomez-Sintes R, Xin Q, Jimenez-Loygorri JI, McCabe M, Diaz A, Garner TP, Cotto-Rios XM, Wu Y, Dong S, Reynolds CA, Patel B, de la Villa P, Macian F, Boya P, Gavathiotis E, Cuervo AM. Targeting retinoic acid receptor alpha-corepressor interaction activates chaperone-mediated autophagy and protects against retinal degeneration. Nat Commun 2022; 13:4220. [PMID: 35864098 PMCID: PMC9304322 DOI: 10.1038/s41467-022-31869-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/30/2022] [Indexed: 11/09/2022] Open
Abstract
Chaperone-mediated autophagy activity, essential in the cellular defense against proteotoxicity, declines with age, and preventing this decline in experimental genetic models has proven beneficial. Here, we have identified the mechanism of action of selective chaperone-mediated autophagy activators previously developed by our group and have leveraged that information to generate orally bioavailable chaperone-mediated autophagy activators with favorable brain exposure. Chaperone-mediated autophagy activating molecules stabilize the interaction between retinoic acid receptor alpha - a known endogenous inhibitor of chaperone-mediated autophagy - and its co-repressor, nuclear receptor corepressor 1, resulting in changes of a discrete subset of the retinoic acid receptor alpha transcriptional program that leads to selective chaperone-mediated autophagy activation. Chaperone-mediated autophagy activators molecules activate this pathway in vivo and ameliorate retinal degeneration in a retinitis pigmentosa mouse model. Our findings reveal a mechanism for pharmacological targeting of chaperone-mediated autophagy activation and suggest a therapeutic strategy for retinal degeneration.
Collapse
Affiliation(s)
- Raquel Gomez-Sintes
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, 28040, Spain
| | - Qisheng Xin
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Juan Ignacio Jimenez-Loygorri
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, 28040, Spain
| | - Mericka McCabe
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Antonio Diaz
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Thomas P Garner
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Xiomaris M Cotto-Rios
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Yang Wu
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Shuxian Dong
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Cara A Reynolds
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Bindi Patel
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Pedro de la Villa
- Department; of System Biology, Universidad de Alcalá, Madrid, Spain and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, 28801, Spain
| | - Fernando Macian
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Patricia Boya
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, 28040, Spain.
| | - Evripidis Gavathiotis
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
31
|
Endicott SJ, Monovich AC, Huang EL, Henry EI, Boynton DN, Beckmann LJ, MacCoss MJ, Miller RA. Lysosomal targetomics of ghr KO mice shows chaperone-mediated autophagy degrades nucleocytosolic acetyl-coA enzymes. Autophagy 2022; 18:1551-1571. [PMID: 34704522 PMCID: PMC9298451 DOI: 10.1080/15548627.2021.1990670] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mice deficient in GHR (growth hormone receptor; ghr KO) have a dramatic lifespan extension and elevated levels of hepatic chaperone-mediated autophagy (CMA). Using quantitative proteomics to identify protein changes in purified liver lysosomes and whole liver lysates, we provide evidence that elevated CMA in ghr KO mice downregulates proteins involved in ribosomal structure, translation initiation and elongation, and nucleocytosolic acetyl-coA production. Following up on these initial proteomics findings, we used a cell culture approach to show that CMA is necessary and sufficient to regulate the abundance of ACLY and ACSS2, the two enzymes that produce nucleocytosolic (but not mitochondrial) acetyl-coA. Inhibition of CMA in NIH3T3 cells has been shown to lead to aberrant accumulation of lipid droplets. We show that this lipid droplet phenotype is rescued by knocking down ACLY or ACSS2, suggesting that CMA regulates lipid droplet formation by controlling ACLY and ACSS2. This evidence leads to a model of how constitutive activation of CMA can shape specific metabolic pathways in long-lived endocrine mutant mice.Abbreviations: CMA: chaperone-mediated autophagy; DIA: data-independent acquisition; ghr KO: growth hormone receptor knockout; GO: gene ontology; I-WAT: inguinal white adipose tissue; KFERQ: a consensus sequence resembling Lys-Phe-Glu-Arg-Gln; LAMP2A: lysosomal-associated membrane protein 2A; LC3-I: non-lipidated MAP1LC3; LC3-II: lipidated MAP1LC3; PBS: phosphate-buffered saline; PI3K: phosphoinositide 3-kinase.
Collapse
Affiliation(s)
| | | | - Eric L. Huang
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Evelynn I. Henry
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA,Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Dennis N. Boynton
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, USA
| | - Logan J. Beckmann
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI, USA
| | - Michael J. MacCoss
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Richard A. Miller
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA,Geriatrics Center, University of Michigan, Ann Arbor, MI, USA,CONTACT Richard A. Miller Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
32
|
Microglial NF-κB drives tau spreading and toxicity in a mouse model of tauopathy. Nat Commun 2022; 13:1969. [PMID: 35413950 PMCID: PMC9005658 DOI: 10.1038/s41467-022-29552-6] [Citation(s) in RCA: 122] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/21/2022] [Indexed: 01/31/2023] Open
Abstract
Activation of microglia is a prominent pathological feature in tauopathies, including Alzheimer's disease. How microglia activation contributes to tau toxicity remains largely unknown. Here we show that nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, activated by tau, drives microglial-mediated tau propagation and toxicity. Constitutive activation of microglial NF-κB exacerbated, while inactivation diminished, tau seeding and spreading in young PS19 mice. Inhibition of NF-κB activation enhanced the retention while reduced the release of internalized pathogenic tau fibrils from primary microglia and rescued microglial autophagy deficits. Inhibition of microglial NF-κB in aged PS19 mice rescued tau-mediated learning and memory deficits, restored overall transcriptomic changes while increasing neuronal tau inclusions. Single cell RNA-seq revealed that tau-associated disease states in microglia were diminished by NF-κB inactivation and further transformed by constitutive NF-κB activation. Our study establishes a role for microglial NF-κB signaling in mediating tau spreading and toxicity in tauopathy.
Collapse
|
33
|
Madrigal-Matute J, de Bruijn J, van Kuijk K, Riascos-Bernal DF, Diaz A, Tasset I, Martín-Segura A, Gijbels MJJ, Sander B, Kaushik S, Biessen EAL, Tiano S, Bourdenx M, Krause GJ, McCracken I, Baker AH, Jin H, Sibinga NES, Bravo-Cordero JJ, Macian F, Singh R, Rensen PCN, Berbée JFP, Pasterkamp G, Sluimer JC, Cuervo AM. Protective role of chaperone-mediated autophagy against atherosclerosis. Proc Natl Acad Sci U S A 2022; 119:e2121133119. [PMID: 35363568 PMCID: PMC9168839 DOI: 10.1073/pnas.2121133119] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/14/2022] [Indexed: 12/11/2022] Open
Abstract
Chaperone-mediated autophagy (CMA) contributes to regulation of energy homeostasis by timely degradation of enzymes involved in glucose and lipid metabolism. Here, we report reduced CMA activity in vascular smooth muscle cells and macrophages in murine and human arteries in response to atherosclerotic challenges. We show that in vivo genetic blockage of CMA worsens atherosclerotic pathology through both systemic and cell-autonomous changes in vascular smooth muscle cells and macrophages, the two main cell types involved in atherogenesis. CMA deficiency promotes dedifferentiation of vascular smooth muscle cells and a proinflammatory state in macrophages. Conversely, a genetic mouse model with up-regulated CMA shows lower vulnerability to proatherosclerotic challenges. We propose that CMA could be an attractive therapeutic target against cardiovascular diseases.
Collapse
Affiliation(s)
- Julio Madrigal-Matute
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Jenny de Bruijn
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
| | - Kim van Kuijk
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- Institute of Experimental Medicine and Systems Biology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany
| | | | - Antonio Diaz
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Inmaculada Tasset
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Adrián Martín-Segura
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Marion J. J. Gijbels
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam University Medical Centers, University of Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Bianca Sander
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
| | - Susmita Kaushik
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Erik A. L. Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, 52074 Aachen, Germany
| | - Simoni Tiano
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Mathieu Bourdenx
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Gregory J. Krause
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Ian McCracken
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Andrew H. Baker
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Han Jin
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
| | - Nicholas E. S. Sibinga
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Jose Javier Bravo-Cordero
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Fernando Macian
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Rajat Singh
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Patrick C. N. Rensen
- Section of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jimmy F. P. Berbée
- Section of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Gerard Pasterkamp
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Judith C. Sluimer
- Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Ana Maria Cuervo
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461
| |
Collapse
|
34
|
Chaperone-Mediated Autophagy in Neurodegenerative Diseases and Acute Neurological Insults in the Central Nervous System. Cells 2022; 11:cells11071205. [PMID: 35406769 PMCID: PMC8997510 DOI: 10.3390/cells11071205] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/27/2022] [Accepted: 03/30/2022] [Indexed: 12/12/2022] Open
Abstract
Autophagy is an important function that mediates the degradation of intracellular proteins and organelles. Chaperone-mediated autophagy (CMA) degrades selected proteins and has a crucial role in cellular proteostasis under various physiological and pathological conditions. CMA dysfunction leads to the accumulation of toxic protein aggregates in the central nervous system (CNS) and is involved in the pathogenic process of neurodegenerative diseases, including Parkinson’s disease and Alzheimer’s disease. Previous studies have suggested that the activation of CMA to degrade aberrant proteins can provide a neuroprotective effect in the CNS. Recent studies have shown that CMA activity is upregulated in damaged neural tissue following acute neurological insults, such as cerebral infarction, traumatic brain injury, and spinal cord injury. It has been also suggested that various protein degradation mechanisms are important for removing toxic aberrant proteins associated with secondary damage after acute neurological insults in the CNS. Therefore, enhancing the CMA pathway may induce neuroprotective effects not only in neurogenerative diseases but also in acute neurological insults. We herein review current knowledge concerning the biological mechanisms involved in CMA and highlight the role of CMA in neurodegenerative diseases and acute neurological insults. We also discuss the possibility of developing CMA-targeted therapeutic strategies for effective treatments.
Collapse
|
35
|
Auzmendi-Iriarte J, Otaegi-Ugartemendia M, Carrasco-Garcia E, Azkargorta M, Diaz A, Saenz-Antoñanzas A, Andermatten JA, Garcia-Puga M, Garcia I, Elua-Pinin A, Ruiz I, Sampron N, Elortza F, Cuervo AM, Matheu A. Chaperone-Mediated Autophagy Controls Proteomic and Transcriptomic Pathways to Maintain Glioma Stem Cell Activity. Cancer Res 2022; 82:1283-1297. [PMID: 35131870 PMCID: PMC9359743 DOI: 10.1158/0008-5472.can-21-2161] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 12/15/2021] [Accepted: 01/27/2022] [Indexed: 02/07/2023]
Abstract
Chaperone-mediated autophagy (CMA) is a homeostatic process essential for the lysosomal degradation of a selected subset of the proteome. CMA activity directly depends on the levels of LAMP2A, a critical receptor for CMA substrate proteins at the lysosomal membrane. In glioblastoma (GBM), the most common and aggressive brain cancer in adulthood, high levels of LAMP2A in the tumor and tumor-associated pericytes have been linked to temozolomide resistance and tumor progression. However, the role of LAMP2A, and hence CMA, in any cancer stem cell type or in glioblastoma stem cells (GSC) remains unknown. In this work, we show that LAMP2A expression is enriched in patient-derived GSCs, and its depletion diminishes GSC-mediated tumorigenic activities. Conversely, overexpression of LAMP2A facilitates the acquisition of GSC properties. Proteomic and transcriptomic analysis of LAMP2A-depleted GSCs revealed reduced extracellular matrix interaction effectors in both analyses. Moreover, pathways related to mitochondrial metabolism and the immune system were differentially deregulated at the proteome level. Furthermore, clinical samples of GBM tissue presented overexpression of LAMP2, which correlated with advanced glioma grade and poor overall survival. In conclusion, we identified a novel role of CMA in directly regulating GSCs activity via multiple pathways at the proteome and transcriptome levels. SIGNIFICANCE A receptor of chaperone-mediated autophagy regulates glioblastoma stem cells and may serve as a potential biomarker for advanced tumor grade and poor survival in this disease.
Collapse
Affiliation(s)
| | | | | | - Mikel Azkargorta
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, ProteoRed-ISCIII, Spain
| | - Antonio Diaz
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, New York
| | | | | | - Mikel Garcia-Puga
- Cellular Oncology Group, Biodonostia Health Research Institute, San Sebastian, Spain
| | - Idoia Garcia
- Cellular Oncology Group, Biodonostia Health Research Institute, San Sebastian, Spain
| | | | - Irune Ruiz
- Cellular Oncology Group, Biodonostia Health Research Institute, San Sebastian, Spain.,Donostia University Hospital, Osakidetza, San Sebastian, Spain
| | - Nicolas Sampron
- Cellular Oncology Group, Biodonostia Health Research Institute, San Sebastian, Spain.,Donostia University Hospital, Osakidetza, San Sebastian, Spain
| | - Felix Elortza
- Proteomics Platform, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, ProteoRed-ISCIII, Spain
| | - Ana Maria Cuervo
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, New York
| | - Ander Matheu
- Cellular Oncology Group, Biodonostia Health Research Institute, San Sebastian, Spain.,CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Madrid, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.,Corresponding Author: Ander Matheu, Cellular Oncology, Biodonostia Health Research Institute, Paseo Dr. Beguiristain s/n, San Sebastian 20014, Spain. E-mail:
| |
Collapse
|
36
|
Jin C, Ou Q, Chen J, Wang T, Zhang J, Wang Z, Wang Y, Tian H, Xu JY, Gao F, Wang J, Li J, Lu L, Xu GT. Chaperone-mediated autophagy plays an important role in regulating retinal progenitor cell homeostasis. Stem Cell Res Ther 2022; 13:136. [PMID: 35365237 PMCID: PMC8973999 DOI: 10.1186/s13287-022-02809-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/12/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE To explore the function and regulatory mechanism of IFITM3 in mouse neural retinal progenitor cells (mNRPCs), which was found to be very important not only in the development of the retina in embryos but also in NRPCs after birth. METHODS Published single-cell sequencing data were used to analyze IFITM3 expression in mNRPCs. RNA interference was used to knock down the expression of IFITM3. CCK-8 assays were used to analyze cell viability. RNA-seq was used to assess mRNA expression, as confirmed by real-time quantitative PCR, and immunofluorescence assays and western blots were used to validate the levels of relative proteins, and autophagy flux assay. Lysosomal trackers were used to track the organelle changes. RESULTS The results of single-cell sequencing data showed that IFITM3 is highly expressed in the embryo, and after birth, RNA-seq showed high IFITM3 expression in mNRPCs. Proliferation and cell viability were greatly reduced after IFITM3 was knocked down. The cell membrane system and lysosomes were dramatically changed, and lysosomes were activated and evidently agglomerated in RAMP-treated cells. The expression of LAMP1 was significantly increased with lysosome agglomeration after treatment with rapamycin (RAMP). Further detection showed that SQSTM1/P62, HSC70 and LAMP-2A were upregulated, while no significant difference in LC3A/B expression was observed; no autophagic flux was generated. CONCLUSION IFITM3 regulates mNRPC viability and proliferation mainly through chaperone-mediated autophagy (CMA) but not macroautophagy (MA). IFITM3 plays a significant role in maintaining the homeostasis of progenitor cell self-renewal by sustaining low-level activation of CMA to eliminate deleterious factors in cells.
Collapse
Affiliation(s)
- Caixia Jin
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China
| | - Qingjian Ou
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jie Chen
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tao Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jieping Zhang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.,Teaching Laboratory Center of Medicine and Life Science, School of Medicine, Tongji University, Shanghai, China
| | - Zhe Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanyuan Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haibin Tian
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jing-Ying Xu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Furong Gao
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Juan Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiao Li
- Teaching Laboratory Center of Medicine and Life Science, School of Medicine, Tongji University, Shanghai, China
| | - Lixia Lu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China. .,Department of Biochemistry and Molecular Biology, School of Medicine, Tongji University, Shanghai, China.
| | - Guo-Tong Xu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China. .,Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, China.
| |
Collapse
|
37
|
Arotcarena M, Soria FN, Cunha A, Doudnikoff E, Prévot G, Daniel J, Blanchard‐Desce M, Barthélémy P, Bezard E, Crauste‐Manciet S, Dehay B. Acidic nanoparticles protect against α-synuclein-induced neurodegeneration through the restoration of lysosomal function. Aging Cell 2022; 21:e13584. [PMID: 35318803 PMCID: PMC9009122 DOI: 10.1111/acel.13584] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 01/11/2022] [Accepted: 02/01/2022] [Indexed: 12/17/2022] Open
Abstract
Parkinson's disease (PD) is an age‐related neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra, associated with the accumulation of misfolded α‐synuclein and lysosomal impairment, two events deemed interconnected. Protein aggregation is linked to defects in degradation systems such as the autophagy‐lysosomal pathway, while lysosomal dysfunction is partly related to compromised acidification. We have recently proven that acidic nanoparticles (aNPs) can re‐acidify lysosomes and ameliorate neurotoxin‐mediated dopaminergic neurodegeneration in mice. However, no lysosome‐targeted approach has yet been tested in synucleinopathy models in vivo. Here, we show that aNPs increase α‐synuclein degradation through enhancing lysosomal activity in vitro. We further demonstrate in vivo that aNPs protect nigral dopaminergic neurons from cell death, ameliorate α‐synuclein pathology, and restore lysosomal function in mice injected with PD patient‐derived Lewy body extracts carrying toxic α‐synuclein aggregates. Our results support lysosomal re‐acidification as a disease‐modifying strategy for the treatment of PD and other age‐related proteinopathies.
Collapse
Affiliation(s)
| | - Federico N. Soria
- Univ. Bordeaux CNRS IMN UMR 5293 Bordeaux France
- Achucarro Basque Center for Neuroscience Dpto. Neurociencias Universidad del País Vasco (UPV/EHU) Leioa Spain
| | - Anthony Cunha
- Univ. Bordeaux CNRS IMN UMR 5293 Bordeaux France
- Université de Bordeaux INSERM U1212 CNRS UMR 5320 ARNA ARN: Régulations Naturelle et Artificielle ChemBioPharm Bordeaux France
| | | | - Geoffrey Prévot
- Univ. Bordeaux CNRS IMN UMR 5293 Bordeaux France
- Université de Bordeaux INSERM U1212 CNRS UMR 5320 ARNA ARN: Régulations Naturelle et Artificielle ChemBioPharm Bordeaux France
- Biomedical Engineering and Imaging Institute Icahn School of Medicine at Mount Sinai New York New York USA
| | - Jonathan Daniel
- Université de Bordeaux Institut des Sciences Moléculaires CNRS UMR 5255 Talence France
| | | | - Philippe Barthélémy
- Université de Bordeaux INSERM U1212 CNRS UMR 5320 ARNA ARN: Régulations Naturelle et Artificielle ChemBioPharm Bordeaux France
| | - Erwan Bezard
- Univ. Bordeaux CNRS IMN UMR 5293 Bordeaux France
| | - Sylvie Crauste‐Manciet
- Université de Bordeaux INSERM U1212 CNRS UMR 5320 ARNA ARN: Régulations Naturelle et Artificielle ChemBioPharm Bordeaux France
| | | |
Collapse
|
38
|
Ferreira JV, da Rosa Soares A, Ramalho J, Máximo Carvalho C, Cardoso MH, Pintado P, Carvalho AS, Beck HC, Matthiesen R, Zuzarte M, Girão H, van Niel G, Pereira P. LAMP2A regulates the loading of proteins into exosomes. SCIENCE ADVANCES 2022; 8:eabm1140. [PMID: 35333565 PMCID: PMC8956266 DOI: 10.1126/sciadv.abm1140] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 02/04/2022] [Indexed: 05/21/2023]
Abstract
Exosomes are extracellular vesicles of endosomal origin that are released by practically all cell types across metazoans. Exosomes are active vehicles of intercellular communication and can transfer lipids, RNAs, and proteins between different cells, tissues, or organs. Here, we describe a mechanism whereby proteins containing a KFERQ motif pentapeptide are loaded into a subpopulation of exosomes in a process that is dependent on the membrane protein LAMP2A. Moreover, we demonstrate that this mechanism is independent of the ESCRT machinery but dependent on HSC70, CD63, Alix, Syntenin-1, Rab31, and ceramides. We show that the master regulator of hypoxia HIF1A is loaded into exosomes by this mechanism to transport hypoxia signaling to normoxic cells. In addition, by tagging fluorescent proteins with KFERQ-like sequences, we were able to follow the interorgan transfer of exosomes. Our findings open new avenues for exosome engineering by allowing the loading of bioactive proteins by tagging them with KFERQ-like motifs.
Collapse
Affiliation(s)
- João Vasco Ferreira
- Proteostasis and Proteolytic Signalling Lab, Chronic Diseases Research Centre (CEDOC), NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Ana da Rosa Soares
- Proteostasis and Proteolytic Signalling Lab, Chronic Diseases Research Centre (CEDOC), NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - José Ramalho
- Proteostasis and Proteolytic Signalling Lab, Chronic Diseases Research Centre (CEDOC), NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Catarina Máximo Carvalho
- Proteostasis and Proteolytic Signalling Lab, Chronic Diseases Research Centre (CEDOC), NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Maria Helena Cardoso
- Proteostasis and Proteolytic Signalling Lab, Chronic Diseases Research Centre (CEDOC), NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Petra Pintado
- Fish Facility, CEDOC, NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Ana Sofia Carvalho
- Computational and Experimental Biology Group, CEDOC, NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Hans Christian Beck
- Centre for Clinical Proteomics, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Rune Matthiesen
- Computational and Experimental Biology Group, CEDOC, NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Mónica Zuzarte
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Center for Innovative Biomedicine and Biotechnology (CIBB), Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - Henrique Girão
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Center for Innovative Biomedicine and Biotechnology (CIBB), Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - Guillaume van Niel
- Université de Paris, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, F-75014 Paris, France
- GHU Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, F-75014 Paris, France
| | - Paulo Pereira
- Proteostasis and Proteolytic Signalling Lab, Chronic Diseases Research Centre (CEDOC), NOVA Medical School, Faculdade de Ciencias Medicas, Universidade NOVA de Lisboa, Lisbon, Portugal
- Corresponding author.
| |
Collapse
|
39
|
Espinosa R, Gutiérrez K, Rios J, Ormeño F, Yantén L, Galaz-Davison P, Ramírez-Sarmiento CA, Parra V, Albornoz A, Alfaro IE, Burgos PV, Morselli E, Criollo A, Budini M. Palmitic and Stearic Acids Inhibit Chaperone-Mediated Autophagy (CMA) in POMC-like Neurons In Vitro. Cells 2022; 11:cells11060920. [PMID: 35326371 PMCID: PMC8945987 DOI: 10.3390/cells11060920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/09/2022] [Accepted: 02/22/2022] [Indexed: 12/28/2022] Open
Abstract
The intake of food with high levels of saturated fatty acids (SatFAs) is associated with the development of obesity and insulin resistance. SatFAs, such as palmitic (PA) and stearic (SA) acids, have been shown to accumulate in the hypothalamus, causing several pathological consequences. Autophagy is a lysosomal-degrading pathway that can be divided into macroautophagy, microautophagy, and chaperone-mediated autophagy (CMA). Previous studies showed that PA impairs macroautophagy function and insulin response in hypothalamic proopiomelanocortin (POMC) neurons. Here, we show in vitro that the exposure of POMC neurons to PA or SA also inhibits CMA, possibly by decreasing the total and lysosomal LAMP2A protein levels. Proteomics of lysosomes from PA- and SA-treated cells showed that the inhibition of CMA could impact vesicle formation and trafficking, mitochondrial components, and insulin response, among others. Finally, we show that CMA activity is important for regulating the insulin response in POMC hypothalamic neurons. These in vitro results demonstrate that CMA is inhibited by PA and SA in POMC-like neurons, giving an overview of the CMA-dependent cellular pathways that could be affected by such inhibition and opening a door for in vivo studies of CMA in the context of the hypothalamus and obesity.
Collapse
Affiliation(s)
- Rodrigo Espinosa
- Molecular and Cellular Pathology Laboratory, Institute in Dentistry Sciences, Dentistry Faculty, University of Chile, Santiago 8380544, Chile; (R.E.); (K.G.); (J.R.); (F.O.)
| | - Karla Gutiérrez
- Molecular and Cellular Pathology Laboratory, Institute in Dentistry Sciences, Dentistry Faculty, University of Chile, Santiago 8380544, Chile; (R.E.); (K.G.); (J.R.); (F.O.)
| | - Javiera Rios
- Molecular and Cellular Pathology Laboratory, Institute in Dentistry Sciences, Dentistry Faculty, University of Chile, Santiago 8380544, Chile; (R.E.); (K.G.); (J.R.); (F.O.)
| | - Fernando Ormeño
- Molecular and Cellular Pathology Laboratory, Institute in Dentistry Sciences, Dentistry Faculty, University of Chile, Santiago 8380544, Chile; (R.E.); (K.G.); (J.R.); (F.O.)
| | - Liliana Yantén
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Avda. Zañartu 1482, Ñuñoa, Santiago 7780272, Chile; (L.Y.); (A.A.); (I.E.A.); (P.V.B.)
| | - Pablo Galaz-Davison
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile; (P.G.-D.); (C.A.R.-S.)
| | - César A. Ramírez-Sarmiento
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile; (P.G.-D.); (C.A.R.-S.)
| | - Valentina Parra
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago 8380544, Chile; (V.P.); (A.C.)
- Autophagy Research Center (ARC), Santiago 8380544, Chile;
| | - Amelina Albornoz
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Avda. Zañartu 1482, Ñuñoa, Santiago 7780272, Chile; (L.Y.); (A.A.); (I.E.A.); (P.V.B.)
- Departamento de Ciencias Básicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile
| | - Iván E. Alfaro
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Avda. Zañartu 1482, Ñuñoa, Santiago 7780272, Chile; (L.Y.); (A.A.); (I.E.A.); (P.V.B.)
- Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7610658, Chile
| | - Patricia V. Burgos
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Avda. Zañartu 1482, Ñuñoa, Santiago 7780272, Chile; (L.Y.); (A.A.); (I.E.A.); (P.V.B.)
- Autophagy Research Center (ARC), Santiago 8380544, Chile;
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile
- Centro de Envejecimiento y Regeneración (CARE-UC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica, Santiago 8331150, Chile
| | - Eugenia Morselli
- Autophagy Research Center (ARC), Santiago 8380544, Chile;
- Departamento de Ciencias Básicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago 8331150, Chile
| | - Alfredo Criollo
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago 8380544, Chile; (V.P.); (A.C.)
- Autophagy Research Center (ARC), Santiago 8380544, Chile;
- Cellular and Molecular Biology Laboratory, Institute in Dentistry Sciences, Dentistry Faculty, University of Chile, Santiago 8380544, Chile
| | - Mauricio Budini
- Molecular and Cellular Pathology Laboratory, Institute in Dentistry Sciences, Dentistry Faculty, University of Chile, Santiago 8380544, Chile; (R.E.); (K.G.); (J.R.); (F.O.)
- Autophagy Research Center (ARC), Santiago 8380544, Chile;
- Correspondence:
| |
Collapse
|
40
|
Kuo SH, Tasset I, Cheng MM, Diaz A, Pan MK, Lieberman OJ, Hutten SJ, Alcalay RN, Kim S, Ximénez-Embún P, Fan L, Kim D, Ko HS, Yacoubian T, Kanter E, Liu L, Tang G, Muñoz J, Sardi SP, Li A, Gan L, Cuervo AM, Sulzer D. Mutant glucocerebrosidase impairs α-synuclein degradation by blockade of chaperone-mediated autophagy. SCIENCE ADVANCES 2022; 8:eabm6393. [PMID: 35138901 DOI: 10.1126/sciadv.abm6393] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The most common genetic risk factors for Parkinson's disease (PD) are a set of heterozygous mutant (MT) alleles of the GBA1 gene that encodes β-glucocerebrosidase (GCase), an enzyme normally trafficked through the ER/Golgi apparatus to the lysosomal lumen. We found that half of the GCase in lysosomes from postmortem human GBA-PD brains was present on the lysosomal surface and that this mislocalization depends on a pentapeptide motif in GCase used to target cytosolic protein for degradation by chaperone-mediated autophagy (CMA). MT GCase at the lysosomal surface inhibits CMA, causing accumulation of CMA substrates including α-synuclein. Single-cell transcriptional analysis and proteomics of brains from GBA-PD patients confirmed reduced CMA activity and proteome changes comparable to those in CMA-deficient mouse brain. Loss of the MT GCase CMA motif rescued primary substantia nigra dopaminergic neurons from MT GCase-induced neuronal death. We conclude that MT GBA1 alleles block CMA function and produce α-synuclein accumulation.
Collapse
Affiliation(s)
- Sheng-Han Kuo
- Department of Neurology, Columbia University , New York, NY 10032, USA
| | - Inmaculada Tasset
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Biochemistry and Molecular Biology, Universidad de Cordoba, Cordoba, Spain
| | - Melody M Cheng
- Department of Neurology, Columbia University , New York, NY 10032, USA
| | - Antonio Diaz
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ming-Kai Pan
- Department of Neurology, Columbia University , New York, NY 10032, USA
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Ori J Lieberman
- Department of Neurology, Columbia University , New York, NY 10032, USA
| | - Samantha J Hutten
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Roy N Alcalay
- Department of Neurology, Columbia University , New York, NY 10032, USA
| | - Sangjun Kim
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
- Neurodegeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Pilar Ximénez-Embún
- Proteomics Unit, Spanish National Cancer Research Centre (CNIO), ProteoRed-ISCIII, Madrid, Spain
| | - Li Fan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Donghoon Kim
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
- Neurodegeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Han Seok Ko
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
- Neurodegeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Talene Yacoubian
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Ellen Kanter
- Departments of Psychiatry and Pharmacology, Columbia University , New York, NY 10032, USA
| | - Ling Liu
- Department of Neurology, Columbia University , New York, NY 10032, USA
| | - Guomei Tang
- Department of Neurology, Columbia University , New York, NY 10032, USA
| | - Javier Muñoz
- Proteomics Unit, Spanish National Cancer Research Centre (CNIO), ProteoRed-ISCIII, Madrid, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | | | - Aiqun Li
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - David Sulzer
- Department of Neurology, Columbia University , New York, NY 10032, USA
- Departments of Psychiatry and Pharmacology, Columbia University , New York, NY 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| |
Collapse
|
41
|
Kaushik S, Tasset I, Arias E, Pampliega O, Wong E, Martinez-Vicente M, Cuervo AM. Autophagy and the hallmarks of aging. Ageing Res Rev 2021; 72:101468. [PMID: 34563704 DOI: 10.1016/j.arr.2021.101468] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/12/2021] [Accepted: 09/20/2021] [Indexed: 12/13/2022]
Abstract
Autophagy, an essential cellular process that mediates degradation of proteins and organelles in lysosomes, has been tightly linked to cellular quality control for its role as part of the proteostasis network. The current interest in identifying the cellular and molecular determinants of aging, has highlighted the important contribution of malfunctioning of autophagy with age to the loss of proteostasis that characterizes all old organisms. However, the diversity of cellular functions of the different types of autophagy and the often reciprocal interactions of autophagy with other determinants of aging, is placing autophagy at the center of the aging process. In this work, we summarize evidence for the contribution of autophagy to health- and lifespan and provide examples of the bidirectional interplay between autophagic pathways and several of the so-called hallmarks of aging. This central role of autophagy in aging, and the dependence on autophagy of many geroprotective interventions, has motivated a search for direct modulators of autophagy that could be used to slow aging and extend healthspan. Here, we review some of those ongoing therapeutic efforts and comment on the potential of targeting autophagy in aging.
Collapse
|
42
|
Targeting lysosomes in human disease: from basic research to clinical applications. Signal Transduct Target Ther 2021; 6:379. [PMID: 34744168 PMCID: PMC8572923 DOI: 10.1038/s41392-021-00778-y] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/26/2021] [Indexed: 01/18/2023] Open
Abstract
In recent years, accumulating evidence has elucidated the role of lysosomes in dynamically regulating cellular and organismal homeostasis. Lysosomal changes and dysfunction have been correlated with the development of numerous diseases. In this review, we interpreted the key biological functions of lysosomes in four areas: cellular metabolism, cell proliferation and differentiation, immunity, and cell death. More importantly, we actively sought to determine the characteristic changes and dysfunction of lysosomes in cells affected by these diseases, the causes of these changes and dysfunction, and their significance to the development and treatment of human disease. Furthermore, we outlined currently available targeting strategies: (1) targeting lysosomal acidification; (2) targeting lysosomal cathepsins; (3) targeting lysosomal membrane permeability and integrity; (4) targeting lysosomal calcium signaling; (5) targeting mTOR signaling; and (6) emerging potential targeting strategies. Moreover, we systematically summarized the corresponding drugs and their application in clinical trials. By integrating basic research with clinical findings, we discussed the current opportunities and challenges of targeting lysosomes in human disease.
Collapse
|
43
|
Pirooznia SK, Rosenthal LS, Dawson VL, Dawson TM. Parkinson Disease: Translating Insights from Molecular Mechanisms to Neuroprotection. Pharmacol Rev 2021; 73:33-97. [PMID: 34663684 DOI: 10.1124/pharmrev.120.000189] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Parkinson disease (PD) used to be considered a nongenetic condition. However, the identification of several autosomal dominant and recessive mutations linked to monogenic PD has changed this view. Clinically manifest PD is then thought to occur through a complex interplay between genetic mutations, many of which have incomplete penetrance, and environmental factors, both neuroprotective and increasing susceptibility, which variably interact to reach a threshold over which PD becomes clinically manifested. Functional studies of PD gene products have identified many cellular and molecular pathways, providing crucial insights into the nature and causes of PD. PD originates from multiple causes and a range of pathogenic processes at play, ultimately culminating in nigral dopaminergic loss and motor dysfunction. An in-depth understanding of these complex and possibly convergent pathways will pave the way for therapeutic approaches to alleviate the disease symptoms and neuroprotective strategies to prevent disease manifestations. This review is aimed at providing a comprehensive understanding of advances made in PD research based on leveraging genetic insights into the pathogenesis of PD. It further discusses novel perspectives to facilitate identification of critical molecular pathways that are central to neurodegeneration that hold the potential to develop neuroprotective and/or neurorestorative therapeutic strategies for PD. SIGNIFICANCE STATEMENT: A comprehensive review of PD pathophysiology is provided on the complex interplay of genetic and environmental factors and biologic processes that contribute to PD pathogenesis. This knowledge identifies new targets that could be leveraged into disease-modifying therapies to prevent or slow neurodegeneration in PD.
Collapse
Affiliation(s)
- Sheila K Pirooznia
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Liana S Rosenthal
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering (S.K.P., V.L.D., T.M.D.), Departments of Neurology (S.K.P., L.S.R., V.L.D., T.M.D.), Departments of Physiology (V.L.D.), Solomon H. Snyder Department of Neuroscience (V.L.D., T.M.D.), Department of Pharmacology and Molecular Sciences (T.M.D.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Adrienne Helis Malvin Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.); and Diana Helis Henry Medical Research Foundation, New Orleans, Louisiana (S.K.P., V.L.D., T.M.D.)
| |
Collapse
|
44
|
Gómez-Sintes R, Arias E. Chaperone-mediated autophagy and disease: Implications for cancer and neurodegeneration. Mol Aspects Med 2021; 82:101025. [PMID: 34629183 DOI: 10.1016/j.mam.2021.101025] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 02/07/2023]
Abstract
Chaperone-mediated autophagy (CMA) is a proteolytic process whereby selected intracellular proteins are degraded inside lysosomes. Owing to its selectivity, CMA participates in the modulation of specific regulatory proteins, thereby playing an important role in multiple cellular processes. Studies conducted over the last two decades have enabled the molecular characterization of this autophagic pathway and the design of specific experimental models, and have underscored the importance of CMA in a range of physiological processes beyond mere protein quality control. Those findings also indicate that decreases in CMA function with increasing age may contribute to the pathogenesis of age-associated diseases, including neurodegeneration and cancer. In the context of neurological diseases, CMA impairment is thought to contribute to the accumulation of misfolded/aggregated proteins, a process central to the pathogenesis of neurodegenerative diseases. CMA therefore constitutes a potential therapeutic target, as its induction accelerates the clearance of pathogenic proteins, promoting cell survival. More recent evidence has highlighted the important and complex role of CMA in cancer biology. While CMA induction may limit tumor development, experimental evidence also indicates that inhibition of this pathway can attenuate the growth of established tumors and improve the response to cancer therapeutics. Here, we describe and discuss the evidence supporting a role of impaired CMA function in neurodegeneration and cancer, as well as future research directions to evaluate the potential of this pathway as a target for the prevention and treatment of these diseases.
Collapse
Affiliation(s)
- Raquel Gómez-Sintes
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas Margarita Salas CIB-CSIC, 28040, Madrid, Spain; Department of Developmental and Molecular Biology & Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| | - Esperanza Arias
- Department of Medicine, Marion Bessin Liver Research Center & Institute for Aging Studies, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
45
|
Einstein-Nathan Shock Center: translating the hallmarks of aging to extend human health span. GeroScience 2021; 43:2167-2182. [PMID: 34463901 DOI: 10.1007/s11357-021-00428-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 10/20/2022] Open
Abstract
The overarching mission of the Einstein-Nathan Shock Center (E-NSC) is to make scientific discoveries in geroscience, leveraging on the expertise in our center in 6 out of the 7 pillars of aging, and to translate their effects towards drug discovery. The relevance of this basic biology of aging discoveries to humans will be confirmed through the unique gero-human resource at E-NSC. This is achieved through services provided by E-NSC, connectivity among its members, attracting worldwide investigators, and providing them with the opportunities to become future leaders. The two central components of the E-NSC are (a) cutting-edge research programs and (b) unique E-NSC research support cores. E-NSC scientists lead NIH-supported cutting-edge research programs that integrate key hallmarks of aging including proteostasis/autophagy, metabolism/inflammaging, genetic/epigenetics, stem cells/regeneration, and translational aging/longevity. Since the inception of the E-NSC, the well-integrated, collaborative, and innovative nature of the multiple supporting state-of-the-art E-NSC research cores form the bedrock of research success at the E-NSC. The three state-of-the-art E-NSC research cores, (i) Proteostasis of Aging Core (PAC), (ii) the Health Span Core (HSC), and (iii) the Human Multi-Omics Core (HMOC), have allowed impressive expansion of translational biological research programs. Expansion was facilitated through the wealth of data coming from genomics/proteomics and metabolomic analysis on human longevity studies, due to access to a variety of biological samples from elderly subjects in clinical trials with aging-targeting drugs, and new drug design services via the PAC to target the hallmarks of aging.
Collapse
|
46
|
Gong Y, Li Z, Zou S, Deng D, Lai P, Hu H, Yao Y, Hu L, Zhang S, Li K, Wei T, Zhao X, Xiao G, Chen Z, Jiang Y, Bai X, Zou Z. Vangl2 limits chaperone-mediated autophagy to balance osteogenic differentiation in mesenchymal stem cells. Dev Cell 2021; 56:2103-2120.e9. [PMID: 34214490 DOI: 10.1016/j.devcel.2021.06.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 04/04/2021] [Accepted: 06/11/2021] [Indexed: 12/11/2022]
Abstract
Lysosomes are the recycling center and nutrient signaling hub of the cell. Here, we show that lysosomes also control mesenchymal stem cell (MSC) differentiation by proteomic reprogramming. The chaperone-mediated autophagy (CMA) lysosome subgroup promotes osteogenesis, while suppressing adipogenesis, by selectively removing osteogenesis-deterring factors, especially master transcriptional factors, such as adipogenic TLE3, ZNF423, and chondrogenic SOX9. The activity of the CMA-committed lysosomes in MSCs are controlled by Van-Gogh-like 2 (Vangl2) at lysosomes. Vangl2 directly binds to lysosome-associated membrane protein 2A (LAMP-2A) and targets it for degradation. MSC-specific Vangl2 ablation in mice increases LAMP-2A expression and CMA-lysosome numbers, promoting bone formation while reducing marrow fat. The Vangl2:LAMP-2A ratio in MSCs correlates inversely with the capacity of the cells for osteoblastic differentiation in humans and mice. These findings demonstrate a critical role for lysosomes in MSC lineage acquisition and establish Vangl2-LAMP-2A signaling as a critical control mechanism.
Collapse
Affiliation(s)
- Yan Gong
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ziqi Li
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shitian Zou
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Daizhao Deng
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Pinglin Lai
- State Key Laboratory of Organ Failure Research, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Hongling Hu
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yongzhou Yao
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Le Hu
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Sheng Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Kai Li
- State Key Laboratory of Organ Failure Research, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Tiantian Wei
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaoyang Zhao
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China
| | - Guozhi Xiao
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment and School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zifeng Chen
- Department of Orthopedic Trauma, Panyu District Central Hospital of Guangzhou, Guangzhou 511400, China
| | - Yu Jiang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Academy of Orthopedics, Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China.
| | - Zhipeng Zou
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
47
|
Khan A, Johnson R, Wittmer C, Maile M, Tatsukawa K, Wong JL, Gill MB, Stocking EM, Natala SR, Paulino AD, Bowden-Verhoek JK, Wrasidlo W, Masliah E, Bonhaus DW, Price DL. NPT520-34 improves neuropathology and motor deficits in a transgenic mouse model of Parkinson's disease. Brain 2021; 144:3692-3709. [PMID: 34117864 DOI: 10.1093/brain/awab214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/12/2021] [Accepted: 05/11/2021] [Indexed: 12/09/2022] Open
Abstract
NPT520-34 is a clinical-stage, small molecule being developed for the treatment of Parkinson's disease and other neurodegenerative disorders. The therapeutic potential of NPT520-34 was first suggested by findings from cell-based assays of alpha-synuclein (ASYN) clearance. As reported here, NPT520-34 was subsequently evaluated for therapeutically relevant actions in a transgenic animal model of Parkinson's disease that overexpresses human ASYN and in an acute lipopolysaccharide (LPS)-challenge model using wild-type mice. Daily administration of NPT520-34 to mThy1-ASYN (Line 61) transgenic mice for one or three months resulted in reduced ASYN pathology, reduced expression of markers of neuroinflammation, and improvements in multiple indices of motor function. In an LPS-challenge model using wild-type mice, a single-dose of NPT520-34 reduced LPS-evoked increases in the expression of several pro-inflammatory cytokines in plasma. These findings demonstrate the beneficial effects of NPT520-34 on both inflammation and protein-pathology endpoints, with consequent improvements in motor function in an animal model of Parkinson's disease. These findings further suggest that NPT520-34 may have two complementary actions: (1) to increase the clearance of neurotoxic protein aggregates and (2) to directly attenuate inflammation. NPT520-34 treatment may thereby address two of the predominate underlying pathophysiological aspects of neurodegenerative disorders such as Parkinson's disease.
Collapse
Affiliation(s)
- Asma Khan
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Robert Johnson
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Carrie Wittmer
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Michelle Maile
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Keith Tatsukawa
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Julian L Wong
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Martin B Gill
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Emily M Stocking
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Srinivasa R Natala
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Amy D Paulino
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Jon K Bowden-Verhoek
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Wolfgang Wrasidlo
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Eliezer Masliah
- Departments of Neuroscience and Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Douglas W Bonhaus
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| | - Diana L Price
- Neuropore Therapies, Inc., 10835 Road to the Cure, Suite 230, San Diego, CA 92121, USA
| |
Collapse
|
48
|
Kim H, Seong J. Fluorescent Protein-Based Autophagy Biosensors. MATERIALS 2021; 14:ma14113019. [PMID: 34199451 PMCID: PMC8199620 DOI: 10.3390/ma14113019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/28/2021] [Accepted: 05/30/2021] [Indexed: 11/16/2022]
Abstract
Autophagy is an essential cellular process of self-degradation for dysfunctional or unnecessary cytosolic constituents and organelles. Dysregulation of autophagy is thus involved in various diseases such as neurodegenerative diseases. To investigate the complex process of autophagy, various biochemical, chemical assays, and imaging methods have been developed. Here we introduce various methods to study autophagy, in particular focusing on the review of designs, principles, and limitations of the fluorescent protein (FP)-based autophagy biosensors. Different physicochemical properties of FPs, such as pH-sensitivity, stability, brightness, spectral profile, and fluorescence resonance energy transfer (FRET), are considered to design autophagy biosensors. These FP-based biosensors allow for sensitive detection and real-time monitoring of autophagy progression in live cells with high spatiotemporal resolution. We also discuss future directions utilizing an optobiochemical strategy to investigate the in-depth mechanisms of autophagy. These cutting-edge technologies will further help us to develop the treatment strategies of autophagy-related diseases.
Collapse
Affiliation(s)
- Heejung Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea;
- Department of Converging Science and Technology, Kyung Hee University, Seoul 02453, Korea
| | - Jihye Seong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea;
- Department of Converging Science and Technology, Kyung Hee University, Seoul 02453, Korea
- Correspondence:
| |
Collapse
|
49
|
Zhang J, Huang J, Gu Y, Xue M, Qian F, Wang B, Yang W, Yu H, Wang Q, Guo X, Ding X, Wang J, Jin M, Zhang Y. Inflammation-induced inhibition of chaperone-mediated autophagy maintains the immunosuppressive function of murine mesenchymal stromal cells. Cell Mol Immunol 2021; 18:1476-1488. [PMID: 31900460 PMCID: PMC8167126 DOI: 10.1038/s41423-019-0345-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 12/04/2019] [Indexed: 12/13/2022] Open
Abstract
Macroautophagy has been implicated in modulating the therapeutic function of mesenchymal stromal cells (MSCs). However, the biological function of chaperone-mediated autophagy (CMA) in MSCs remains elusive. Here, we found that CMA was inhibited in MSCs in response to the proinflammatory cytokines interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α). In addition, suppression of CMA by knocking down the CMA-related lysosomal receptor lysosomal-associated membrane protein 2 (LAMP-2A) in MSCs significantly enhanced the immunosuppressive effect of MSCs on T cell proliferation, and as expected, LAMP-2A overexpression in MSCs exerted the opposite effect on T cell proliferation. This effect of CMA on the immunosuppressive function of MSCs was attributed to its negative regulation of the expression of chemokine C-X-C motif ligand 10 (CXCL10), which recruits inflammatory cells, especially T cells, to MSCs, and inducible nitric oxide synthase (iNOS), which leads to the subsequent inhibition of T cell proliferation via nitric oxide (NO). Mechanistically, CMA inhibition dramatically promoted IFN-γ plus TNF-α-induced activation of NF-κB and STAT1, leading to the enhanced expression of CXCL10 and iNOS in MSCs. Furthermore, we found that IFN-γ plus TNF-α-induced AKT activation contributed to CMA inhibition in MSCs. More interestingly, CMA-deficient MSCs exhibited improved therapeutic efficacy in inflammatory liver injury. Taken together, our findings established CMA inhibition as a critical contributor to the immunosuppressive function of MSCs induced by inflammatory cytokines and highlighted a previously unknown function of CMA.
Collapse
Affiliation(s)
- Jie Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jiefang Huang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuting Gu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Mingxing Xue
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Fengtao Qian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bei Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wanlin Yang
- Pediatric Institute of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - Hongshuang Yu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiwei Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Guo
- Pediatric Institute of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - Xinyuan Ding
- Pediatric Institute of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - Jina Wang
- Department of Urology and Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Min Jin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Yanyun Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
- Pediatric Institute of Soochow University, Institutes for Translational Medicine, Soochow University, Suzhou, China.
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
50
|
Bourdenx M, Martín-Segura A, Scrivo A, Rodriguez-Navarro JA, Kaushik S, Tasset I, Diaz A, Storm NJ, Xin Q, Juste YR, Stevenson E, Luengo E, Clement CC, Choi SJ, Krogan NJ, Mosharov EV, Santambrogio L, Grueninger F, Collin L, Swaney DL, Sulzer D, Gavathiotis E, Cuervo AM. Chaperone-mediated autophagy prevents collapse of the neuronal metastable proteome. Cell 2021; 184:2696-2714.e25. [PMID: 33891876 DOI: 10.1016/j.cell.2021.03.048] [Citation(s) in RCA: 164] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 01/03/2021] [Accepted: 03/23/2021] [Indexed: 12/18/2022]
Abstract
Components of the proteostasis network malfunction in aging, and reduced protein quality control in neurons has been proposed to promote neurodegeneration. Here, we investigate the role of chaperone-mediated autophagy (CMA), a selective autophagy shown to degrade neurodegeneration-related proteins, in neuronal proteostasis. Using mouse models with systemic and neuronal-specific CMA blockage, we demonstrate that loss of neuronal CMA leads to altered neuronal function, selective changes in the neuronal metastable proteome, and proteotoxicity, all reminiscent of brain aging. Imposing CMA loss on a mouse model of Alzheimer's disease (AD) has synergistic negative effects on the proteome at risk of aggregation, thus increasing neuronal disease vulnerability and accelerating disease progression. Conversely, chemical enhancement of CMA ameliorates pathology in two different AD experimental mouse models. We conclude that functional CMA is essential for neuronal proteostasis through the maintenance of a subset of the proteome with a higher risk of misfolding than the general proteome.
Collapse
Affiliation(s)
- Mathieu Bourdenx
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Adrián Martín-Segura
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Aurora Scrivo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jose A Rodriguez-Navarro
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Susmita Kaushik
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Inmaculada Tasset
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Antonio Diaz
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Nadia J Storm
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Qisheng Xin
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Yves R Juste
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Erica Stevenson
- Department of Cellular Molecular Pharmacology, School of Medicine and California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA; David Gladstone Institutes, San Francisco, CA 94158, USA
| | - Enrique Luengo
- Department of Pharmacology, School of Medicine, Instituto Teófilo Hernando for Drug Discovery, Universidad Autonoma de Madrid, Madrid 28049, Spain
| | - Cristina C Clement
- Department of Radiation Oncology, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Se Joon Choi
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY 10461, USA
| | - Nevan J Krogan
- Department of Cellular Molecular Pharmacology, School of Medicine and California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA; David Gladstone Institutes, San Francisco, CA 94158, USA
| | - Eugene V Mosharov
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY 10461, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Fiona Grueninger
- Roche Pharma Research and Early Development (pRED), Neuro-Immunology, Roche Innovation Center Basel, CH-4070, Switzerland
| | - Ludovic Collin
- Roche Pharma Research and Early Development (pRED), Neuro-Immunology, Roche Innovation Center Basel, CH-4070, Switzerland
| | - Danielle L Swaney
- Department of Cellular Molecular Pharmacology, School of Medicine and California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA; David Gladstone Institutes, San Francisco, CA 94158, USA
| | - David Sulzer
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY 10461, USA; Departments of Neurology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA
| | - Evripidis Gavathiotis
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|