1
|
Umargamwala R, Nicolson S, Manning J, Carosi JM, Kumar S, Denton D. Identification of new candidates regulating autophagy-dependent midgut degradation in Drosophila melanogaster. Cell Death Discov 2025; 11:181. [PMID: 40240351 PMCID: PMC12003636 DOI: 10.1038/s41420-025-02474-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/24/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
Autophagy-dependent cell death (ADCD) is a context-specific form of programmed cell death that plays an important role in development and homeostasis. During Drosophila metamorphosis, hormonal cues modulate growth and other signalling cascades which results in autophagy-dependent degradation of the obsolete larval midgut. While this process does not require caspase activity or apoptotic machinery, several canonical autophagy-related proteins are also dispensable, suggesting additional regulators may be involved in effectively eliminating the larval midgut. Ubiquitination, a process that attaches one or more ubiquitin moieties to a substrate through sequential reactions involving a cascade of enzymes, plays a critical role in autophagy. As the specific role(s) of ubiquitination in ADCD has not been explored, we previously performed a RNAi-mediated knockdown screen of over 250 ubiquitin machinery genes in GFP-labelled Drosophila larval midguts and identified 18 candidate regulators of midgut degradation. In this work, we screened candidate genes for a role in autophagy-dependent midgut degradation by analysing mosaic clones and genetic interactions with Atg1. Validation and further studies into the ubiquitin conjugating enzyme, Effete (Eff), and two ubiquitin ligases, Cullin-4 (Cul4) and Supernumerary limbs (Slmb), demonstrated interplay between ubiquitination and the autophagy machinery in coordinating autophagy-dependent midgut degradation.
Collapse
Affiliation(s)
- Ruchi Umargamwala
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Shannon Nicolson
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Jantina Manning
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Julian M Carosi
- South Australian Health and Medical Research Institute, Adelaide, Australia
- School of Biological Sciences, The University of Adelaide, Adelaide, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia.
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.
| | - Donna Denton
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| |
Collapse
|
2
|
Henn D, Yang X, Li M. Lysosomal quality control Review. Autophagy 2025:1-20. [PMID: 39968899 DOI: 10.1080/15548627.2025.2469206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025] Open
Abstract
Healthy cells need functional lysosomes to degrade cargo delivered by autophagy and endocytosis. Defective lysosomes can lead to severe conditions such as lysosomal storage diseases (LSDs) and neurodegeneration. To maintain lysosome integrity and functionality, cells have evolved multiple quality control pathways corresponding to different types of stress and damage. These can be divided into five levels: regulation, reformation, repair, removal, and replacement. The different levels of lysosome quality control often work together to maintain the integrity of the lysosomal network. This review summarizes the different quality control pathways and discusses the less-studied area of lysosome membrane protein regulation and degradation, highlighting key unanswered questions in the field.Abbreviation: ALR: autophagic lysosome reformation; CASM: conjugation of ATG8 to single membranes: ER: endoplasmic reticulum; ESCRT: endosomal sorting complexes required for transport; ILF: intralumenal fragment; LSD: lysosomal storage disease; LYTL: lysosomal tubulation/sorting driven by LRRK2; PITT: phosphoinositide-initiated membrane tethering and lipid transport; PE: phosphatidylethanolamine; PLR: phagocytic lysosome reformation; PS: phosphatidylserine; PtdIns3P: phosphatidylinositol-3-phosphate; PtdIns4P: phosphatidylinositol-4-phosphate; PtdIns(4,5)P2: phosphatidylinositol-4,5-bisphosphate; V-ATPase: vacuolar-type H+-translocating ATPase.
Collapse
Affiliation(s)
- Danielle Henn
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Xi Yang
- Department of Biological Sciences, Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, USA
| | - Ming Li
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
3
|
Gopal Krishnan PD, Lee WX, Goh KY, Choy SM, Turqueza LRR, Lim ZH, Tang HW. Transcriptional regulation of autophagy in skeletal muscle stem cells. Dis Model Mech 2025; 18:DMM052007. [PMID: 39925192 PMCID: PMC11849978 DOI: 10.1242/dmm.052007] [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] [Indexed: 02/11/2025] Open
Abstract
Muscle stem cells (MuSCs) are essential for the regenerative capabilities of skeletal muscles. MuSCs are maintained in a quiescent state, but, when activated, can undergo proliferation and differentiation into myocytes, which fuse and mature to generate muscle fibers. The maintenance of MuSC quiescence and MuSC activation are processes that are tightly regulated by autophagy, a conserved degradation system that removes unessential or dysfunctional cellular components via lysosomes. Both the upregulation and downregulation of autophagy have been linked to impaired muscle regeneration, causing myopathies such as cancer cachexia, sarcopenia and Duchenne muscular dystrophy. In this Review, we highlight the importance of autophagy in regulating MuSC activity during muscle regeneration. Additionally, we summarize recent studies that link the transcriptional dysregulation of autophagy to muscle atrophy, emphasizing the dominant roles that transcription factors play in myogenic programs. Deciphering and understanding the roles of these transcription factors in the regulation of autophagy during myogenesis could advance the development of regenerative medicine.
Collapse
Affiliation(s)
- Priya D. Gopal Krishnan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Wen Xing Lee
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Kah Yong Goh
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Sze Mun Choy
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | | | - Zhuo Han Lim
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Hong-Wen Tang
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore
| |
Collapse
|
4
|
Mikhail A, Ng S, Xhuti D, Lesinski M, Chhor J, Deguise M, De Repentigny Y, Nederveen J, Kothary R, Tarnopolsky M, Ljubicic V. Skeletal Muscle Mitochondrial and Autophagic Dysregulation Are Modifiable in Spinal Muscular Atrophy. J Cachexia Sarcopenia Muscle 2025; 16:e13701. [PMID: 39901351 PMCID: PMC11790611 DOI: 10.1002/jcsm.13701] [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: 04/23/2024] [Revised: 11/18/2024] [Accepted: 11/29/2024] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Spinal muscular atrophy (SMA) is a health- and life-limiting neuromuscular disorder. Although varying degrees of mitochondrial abnormalities have been documented in SMA skeletal muscle, the influence of disease progression on pathways that govern organelle turnover and dynamics are poorly understood. Thus, the purpose of this study was to investigate skeletal muscle mitochondria during SMA disease progression and determine the effects of therapeutic modalities on organelle biology. METHODS Smn2B/+ and Smn2B/- severe SMA-like mice were used to investigate mitochondrial turnover and dynamics signalling. Muscles were analysed at postnatal day 9 (P9), P13 or P21 to address pre-symptomatic, early symptomatic and late symptomatic periods of the disorder. Additionally, we utilized an acute dose of exercise and urolithin A (UA) to stimulate organelle remodelling in skeletal muscle of SMA mice in vivo and in SMA patient-derived myotubes in vitro, respectively. RESULTS Smn2B/+ and Smn2B/- mice demonstrated similar levels of muscle mitochondrial oxidative phosphorylation (OxPhos) proteins throughout disease progression. In contrast, at P21 the mRNA levels of upstream factors important for the transcription of mitochondrial genes encoded by the nuclear and mitochondrial DNA, including nuclear respiratory factor 2, sirtuin 1, mitochondrial transcription factor A and tumour protein 53, were upregulated (+31%-195%, p < 0.05) in Smn2B/- mice relative to Smn2B/+. Early and late symptomatic skeletal muscle from SMA-like mice showed greater autophagosome formation as denoted by more phosphorylated autophagy related 16-like 1 (p-ATG16L1Ser278) puncta (+60%-80%, p < 0.05), along with a build-up of molecules indicative of damaged mitochondria such as BCL2 interacting protein 3, Parkin and PTEN-induced kinase 1 (+100%-195%, p < 0.05). Furthermore, we observed a fragmented mitochondrial phenotype at P21 that was concomitant with abnormal splicing of Optic atrophy 1 transcripts (-53%, p < 0.05). A single dose of exercise augmented the expression of citrate synthase (+43%, p < 0.05) and corrected the over-assembly of autophagosomes (-64%, p < 0.05). In patient muscle cells, UA treatment stimulated autophagic flux, increased the expression of OxPhos proteins (+15%-47%, p < 0.05) and improved maximal oxygen consumption (+84%, p < 0.05). CONCLUSIONS Abnormal skeletal muscle mitochondrial turnover and dynamics are associated with disease progression in Smn2B/- mice despite compensatory elevations in upstream factors important for organelle synthesis and recycling. Exercise and UA enhance mitochondrial health in skeletal muscle, which indicates that lifestyle-based and pharmacological interventions may be effective countermeasures targeting the organelle for therapeutic remodelling in SMA.
Collapse
Affiliation(s)
| | - Sean Y. Ng
- Department of KinesiologyMcMaster UniversityHamiltonOntarioCanada
| | - Donald Xhuti
- Department of PediatricsMcMaster University Medical CenterHamiltonOntarioCanada
| | | | - Jennifer Chhor
- Department of KinesiologyMcMaster UniversityHamiltonOntarioCanada
| | - Marc‐Olivier Deguise
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawaOntarioCanada
- Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
- Division of Neonatology, Department of PediatricsChildren's Hospital of Eastern OntarioOttawaOntarioCanada
| | - Yves De Repentigny
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Joshua P. Nederveen
- Department of PediatricsMcMaster University Medical CenterHamiltonOntarioCanada
| | - Rashmi Kothary
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawaOntarioCanada
- Department of MedicineUniversity of OttawaOttawaOntarioCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- Canada and Centre for Neuromuscular DiseaseUniversity of OttawaOttawaOntarioCanada
| | - Mark A. Tarnopolsky
- Department of PediatricsMcMaster University Medical CenterHamiltonOntarioCanada
| | | |
Collapse
|
5
|
Acheson J, Joanisse S, Sale C, Hodson N. Recycle, repair, recover: the role of autophagy in modulating skeletal muscle repair and post-exercise recovery. Biosci Rep 2025; 45:1-30. [PMID: 39670455 PMCID: PMC12096956 DOI: 10.1042/bsr20240137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 12/14/2024] Open
Abstract
Skeletal muscle is a highly plastic tissue that can adapt relatively rapidly to a range of stimuli. In response to novel mechanical loading, e.g. unaccustomed resistance exercise, myofibers are disrupted and undergo a period of ultrastructural remodeling to regain full physiological function, normally within 7 days. The mechanisms that underpin this remodeling are believed to be a combination of cellular processes including ubiquitin-proteasome/calpain-mediated degradation, immune cell infiltration, and satellite cell proliferation/differentiation. A relatively understudied system that has the potential to be a significant contributing mechanism to repair and recovery is the autophagolysosomal system, an intracellular process that degrades damaged and redundant cellular components to provide constituent metabolites for the resynthesis of new organelles and cellular structures. This review summarizes our current understanding of the autophagolysosomal system in the context of skeletal muscle repair and recovery. In addition, we also provide hypothetical models of how this system may interact with other processes involved in skeletal muscle remodeling and provide avenues for future research to improve our understanding of autophagy in human skeletal muscle.
Collapse
Affiliation(s)
- Jordan Acheson
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Institute of Sport, Manchester, U.K.
| | - Sophie Joanisse
- School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham, U.K.
| | - Craig Sale
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Institute of Sport, Manchester, U.K.
| | - Nathan Hodson
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Institute of Sport, Manchester, U.K.
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Shan XQ, Zhou N, Pei CX, Lu X, Chen CP, Chen HQ. Tetrandrine induces muscle atrophy involving ROS-mediated inhibition of Akt and FoxO3. Mol Med 2024; 30:218. [PMID: 39548359 PMCID: PMC11566300 DOI: 10.1186/s10020-024-00981-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024] Open
Abstract
Tetrandrine (Tet), a well-known drug of calcium channel blocker, has been broadly applied for anti-inflammatory and anti-fibrogenetic therapy. However, due to the functional diversity of ubiquitous calcium channels, potential side-effects may be expected. Our previous report revealed an inhibitory effect of Tet on myogenesis of skeletal muscle. Here, we found that Tet induced protein degradation resulting in the myofibril atrophy. Upon administration with a relative high dose (40 mg/kg) of Tet for 28 days, the mice displayed significantly reduced muscle mass, strength force, and myosin heavy chain (MyHC) protein levels. The MyHC reduction was further detected in C2C12 myotubes after treating with Tet. Interestingly, the expression of Atrogin-1 and Murf-1, the skeletal muscle specific E3 ligases of protein ubiquitin-proteasome system (UPS), was accordingly up-regulated, and the reduced MyHC was significantly mitigated by MG132, a 26S proteasome inhibitor, indicating a key role of UPS in the protein degradation of muscle cells. Further study showed that Tet induced autophagy also participated in the protein degradation. Mechanistically, Tet treatment caused ROS production in myotubes that in turn targeted on FoxO3/AKT signaling, resulting in the activation of UPS and autophagy processes that were involved in the protein degradation. Our study reveals a potential side-effect of Tet on skeletal muscle atrophy, particularly when the drug dose is relatively high.
Collapse
Affiliation(s)
- Xin-Qi Shan
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Na Zhou
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Chuang-Xin Pei
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Xue Lu
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Cai-Ping Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
| | - Hua-Qun Chen
- The Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Sciences, Nanjing Normal University, Nanjing, 210023, China.
| |
Collapse
|
7
|
Singh A, Mahapatra KK, Praharaj PP, Patra S, Mishra SR, Patil S, Bhutia SK. Prolonged glutamine starvation reactivates mTOR to inhibit autophagy and initiate autophagic lysosome reformation to maintain cell viability. Int J Biochem Cell Biol 2024; 177:106694. [PMID: 39547430 DOI: 10.1016/j.biocel.2024.106694] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 11/11/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Autophagy, a cellular recycling mechanism, utilizes lysosomes for cellular degradation. Prolonged autophagy reduces the pool of functional lysosomes in the cell. However, lysosomal homeostasis is maintained through the regeneration of functional lysosomes during the terminal stage of autophagy, i.e. Autophagic lysosome reformation (ALR). Through confocal microscopy during glutamine starvation, we unravel the regeneration of tubules from autolysosomes by undertaking significant membrane remodeling, which majorly depends on mTOR reactivation, RAB7 dissociation, phosphatidyl inositol 3 phosphate (PI3P) dependent-dynamin 2 and clathrin recruitment. In glutamine-starved cells, we found mTOR is the central modulator in regulating ALR initiation, and its pharmacological inhibition with rapamycin leads to a decrease in lysosomal tubulation. Moreover, RAB7 and Clathrin are essential for tubule elongation and it showed that siRNA targeting RAB7 and Clathrin restricts tubule initiation under glutamine starvation. In this setting, we examined the physiological relevance of ALR during prolonged glutamine deprivation and found that genetic and pharmacological inhibition of critical proteins involved in ALR promotes cell death in oral cancer cells, establishing ALR is essential for maintaining cell survival during stress.
Collapse
Affiliation(s)
- Amruta Singh
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Kewal Kumar Mahapatra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Prakash Priyadarshi Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Soumya Ranjan Mishra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Sankargouda Patil
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT 84095, USA
| | - Sujit Kumar Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India.
| |
Collapse
|
8
|
He Y, Fan Y, Ahmadpoor X, Wang Y, Li ZA, Zhu W, Lin H. Targeting lysosomal quality control as a therapeutic strategy against aging and diseases. Med Res Rev 2024; 44:2472-2509. [PMID: 38711187 DOI: 10.1002/med.22047] [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: 08/19/2023] [Revised: 04/04/2024] [Accepted: 04/21/2024] [Indexed: 05/08/2024]
Abstract
Previously, lysosomes were primarily referred to as the digestive organelles and recycling centers within cells. Recent discoveries have expanded the lysosomal functional scope and revealed their critical roles in nutrient sensing, epigenetic regulation, plasma membrane repair, lipid transport, ion homeostasis, and cellular stress response. Lysosomal dysfunction is also found to be associated with aging and several diseases. Therefore, function of macroautophagy, a lysosome-dependent intracellular degradation system, has been identified as one of the updated twelve hallmarks of aging. In this review, we begin by introducing the concept of lysosomal quality control (LQC), which is a cellular machinery that maintains the number, morphology, and function of lysosomes through different processes such as lysosomal biogenesis, reformation, fission, fusion, turnover, lysophagy, exocytosis, and membrane permeabilization and repair. Next, we summarize the results from studies reporting the association between LQC dysregulation and aging/various disorders. Subsequently, we explore the emerging therapeutic strategies that target distinct aspects of LQC for treating diseases and combatting aging. Lastly, we underscore the existing knowledge gap and propose potential avenues for future research.
Collapse
Affiliation(s)
- Yuchen He
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yishu Fan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xenab Ahmadpoor
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yumin Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, China
| | - Weihong Zhu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Orthopaedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
9
|
Jiang J, Ren R, Fang W, Miao J, Wen Z, Wang X, Xu J, Jin H. Lysosomal biogenesis and function in osteoclasts: a comprehensive review. Front Cell Dev Biol 2024; 12:1431566. [PMID: 39170917 PMCID: PMC11335558 DOI: 10.3389/fcell.2024.1431566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024] Open
Abstract
Lysosomes serve as catabolic centers and signaling hubs in cells, regulating a multitude of cellular processes such as intracellular environment homeostasis, macromolecule degradation, intracellular vesicle trafficking and autophagy. Alterations in lysosomal level and function are crucial for cellular adaptation to external stimuli, with lysosome dysfunction being implicated in the pathogenesis of numerous diseases. Osteoclasts (OCs), as multinucleated cells responsible for bone resorption and maintaining bone homeostasis, have a complex relationship with lysosomes that is not fully understood. Dysregulated function of OCs can disrupt bone homeostasis leading to the development of various bone disorders. The regulation of OC differentiation and bone resorption for the treatment of bone disease have received considerable attention in recent years, yet the role and regulation of lysosomes in OCs, as well as the potential therapeutic implications of intervening in lysosomal biologic behavior for the treatment of bone diseases, remain relatively understudied. This review aims to elucidate the mechanisms involved in lysosomal biogenesis and to discuss the functions of lysosomes in OCs, specifically in relation to differentiation, bone resorption, and autophagy. Finally, we explore the potential therapeutic implication of targeting lysosomes in the treatment of bone metabolic disorders.
Collapse
Affiliation(s)
- Junchen Jiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Rufeng Ren
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Weiyuan Fang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jiansen Miao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zijun Wen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xiangyang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Haiming Jin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
10
|
Gao C, Wan Q, Yan J, Zhu Y, Tian L, Wei J, Feng B, Niu L, Jiao K. Exploring the Link Between Autophagy-Lysosomal Dysfunction and Early Heterotopic Ossification in Tendons. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400790. [PMID: 38741381 PMCID: PMC11267276 DOI: 10.1002/advs.202400790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/26/2024] [Indexed: 05/16/2024]
Abstract
Heterotopic ossification (HO), the pathological formation of bone within soft tissues such as tendon and muscle, is a notable complication resulting from severe injury. While soft tissue injury is necessary for HO development, the specific molecular pathology responsible for trauma-induced HO remains a mystery. The previous study detected abnormal autophagy function in the early stages of tendon HO. Nevertheless, it remains to be determined whether autophagy governs the process of HO generation. Here, trauma-induced tendon HO model is used to investigate the relationship between autophagy and tendon calcification. In the early stages of tenotomy, it is observed that autophagic flux is significantly impaired and that blocking autophagic flux promoted the development of more rampant calcification. Moreover, Gt(ROSA)26sor transgenic mouse model experiments disclosed lysosomal acid dysfunction as chief reason behind impaired autophagic flux. Stimulating V-ATPase activity reinstated both lysosomal acid functioning and autophagic flux, thereby reversing tendon HO. This present study demonstrates that autophagy-lysosomal dysfunction triggers HO in the stages of tendon injury, with potential therapeutic targeting implications for HO.
Collapse
Affiliation(s)
- Chang‐He Gao
- Department of StomatologyTangdu HospitalState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- Department of StomatologyThe Third Affiliated Hospital of Xinxiang Medical UniversityXinxiangHenan453000P. R. China
| | - Qian‐Qian Wan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Jan‐Fei Yan
- Department of StomatologyTangdu HospitalState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Yi‐Na Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Lei Tian
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Jian‐Hua Wei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Bin Feng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Li‐Na Niu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Kai Jiao
- Department of StomatologyTangdu HospitalState Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationSchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| |
Collapse
|
11
|
Jang JH, Jung HH, Oh NS. Enhanced protective effect of whey protein fermented with Lacticaseibacillus rhamnosus IM36 on dexamethasone-induced myotube atrophy. Food Sci Biotechnol 2024; 33:2243-2254. [PMID: 39130659 PMCID: PMC11315874 DOI: 10.1007/s10068-024-01640-x] [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: 05/10/2024] [Revised: 05/30/2024] [Accepted: 06/10/2024] [Indexed: 08/13/2024] Open
Abstract
This study investigated the preventive potential of whey protein fermented with Lacticaseibacillus rhamnosus IM36 (FWP) against muscle atrophy induced by dexamethasone (DEX). FWP exhibited enhanced antioxidant activities compared with those of unfermented whey protein, effectively suppressing DEX-induced reactive oxygen species production. FWP was treated before the administration of 100 μM DEX on C2C12 myotubes and compared to unfermented whey (WP). DEX significantly inhibited myotube viability and muscle protein synthesis and enhanced degradation. FWP exhibited a dose-dependent attenuation of cell viability loss compared with that of WP. Additionally, FWP stimulated the formation of myotubes and muscle protein synthesis by upregulating myogenesis and insulin-like growth factor-1 expression. Furthermore, FWP significantly attenuated forkhead box protein O3a-mediated ubiquitin ligases and autophagy of lysosomes activated by DEX, inhibiting pathways that lead to muscle protein breakdown. These findings suggest that FWP enhances antioxidant activity and prevented DEX-induced muscle atrophy by regulating muscle protein homeostasis. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-024-01640-x.
Collapse
Affiliation(s)
- Ji Hun Jang
- Department of Food and Biotechnology, Korea University, Sejong, 30019 Korea
| | - Hyeon Ho Jung
- Department of Food and Biotechnology, Korea University, Sejong, 30019 Korea
| | - Nam Su Oh
- Department of Food and Biotechnology, Korea University, Sejong, 30019 Korea
| |
Collapse
|
12
|
Zhou H, Lin X, Feng S, Zhu S, Zhou H, Chen H, Youwu H, Wang Z, Wang R, Shao X, Wang J. Metformin mitigates adipogenesis of fibro-adipogenic progenitors after rotator cuff tears via activating mTOR/ULK1-mediated autophagy. Am J Physiol Cell Physiol 2024; 326:C1590-C1603. [PMID: 38586878 PMCID: PMC11371331 DOI: 10.1152/ajpcell.00034.2024] [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: 01/16/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/09/2024]
Abstract
Muscular fatty infiltration is a common issue after rotator cuff tears (RCTs), which impair shoulder function. Females suffer a higher prevalence and a more severe degree of muscular fatty infiltration after RCT when compared with males, with the underlying mechanisms remaining unclear. Fibro-adipogenic progenitors (FAPs) are the primary source of muscular fatty infiltration following RCT. Our findings disclose that gender-specific disparities in muscular fatty infiltration are linked to mTOR/ULK1-mediated autophagy of FAPs. Decreased autophagic activity contributes to adipogenic differentiation in female FAPs after RCT. Furthermore, metformin could enhance mTOR/ULK1-mediated autophagic processes of FAPs, thereby alleviating fatty infiltration and improving shoulder functionality after RCT. Together, our study reveals that gender differences in muscular fatty infiltration arise from distinct autophagic activities. Metformin could be a promising noninvasive intervention to ameliorate muscular fatty infiltration of RCT.NEW & NOTEWORTHY The current study demonstrated that gender-specific disparities in muscular fatty infiltration are attributed to mTOR/ULK1-mediated autophagy of FAPs. Decreased autophagic activity contributes to adipogenic differentiation in female FAPs after RCT. Moreover, metformin could enhance mTOR/ULK1-mediated autophagic processes of FAPs, thereby alleviating fatty infiltration and improving shoulder functionality after RCT. Therefore, metformin could be a promising noninvasive intervention to ameliorate muscular fatty infiltration of RCT.
Collapse
Affiliation(s)
- Hao Zhou
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xingzuan Lin
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Shujing Feng
- Department of Sports Medicine, School of Exercise and Health, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Siyuan Zhu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Han Zhou
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Huifang Chen
- Department of Sports Medicine, School of Exercise and Health, Shanghai University of Sport, Shanghai, People's Republic of China
| | - He Youwu
- Department of Hand Plastic Surgery, The First People's Hospital of Linping District, Hangzhou, People's Republic of China
| | - Zekai Wang
- Department of Life Science, University of Toronto, Toronto, Ontario, Canada
| | - Ru Wang
- Department of Sports Medicine, School of Exercise and Health, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Xiexiang Shao
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jianhua Wang
- Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
13
|
Bao L, Liu Q, Wang J, Shi L, Pang Y, Niu Y, Zhang R. The interactions of subcellular organelles in pulmonary fibrosis induced by carbon black nanoparticles: a comprehensive review. Arch Toxicol 2024; 98:1629-1643. [PMID: 38536500 DOI: 10.1007/s00204-024-03719-0] [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: 12/11/2023] [Accepted: 02/29/2024] [Indexed: 05/21/2024]
Abstract
Owing to the widespread use and improper emissions of carbon black nanoparticles (CBNPs), the adverse effects of CBNPs on human health have attracted much attention. In toxicological research, carbon black is frequently utilized as a negative control because of its low toxicity and poor solubility. However, recent studies have indicated that inhalation exposure to CBNPs could be a risk factor for severe and prolonged pulmonary inflammation and fibrosis. At present, the pathogenesis of pulmonary fibrosis induced by CBNPs is still not fully elucidated, but it is known that with small particle size and large surface area, CBNPs are more easily ingested by cells, leading to organelle damage and abnormal interactions between organelles. Damaged organelle and abnormal organelles interactions lead to cell structure and function disorders, which is one of the important factors in the development and occurrence of various diseases, including pulmonary fibrosis. This review offers a comprehensive analysis of organelle structure, function, and interaction mechanisms, while also summarizing the research advancements in organelles and organelle interactions in CBNPs-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Lei Bao
- Department of Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China
| | - Qingping Liu
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China
- Department of Toxicology, Hebei Medical University, 361 Zhongshan East Rd, Shijiazhuang, 050017, Hebei, China
| | - Jingyuan Wang
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China
- Department of Toxicology, Hebei Medical University, 361 Zhongshan East Rd, Shijiazhuang, 050017, Hebei, China
| | - Lili Shi
- Department of Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China
| | - Yaxian Pang
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China
- Department of Toxicology, Hebei Medical University, 361 Zhongshan East Rd, Shijiazhuang, 050017, Hebei, China
| | - Yujie Niu
- Department of Occupational Health and Environmental Health, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China
| | - Rong Zhang
- Hebei Key Laboratory of Environment and Human Health, Shijiazhuang, 050017, China.
- Department of Toxicology, Hebei Medical University, 361 Zhongshan East Rd, Shijiazhuang, 050017, Hebei, China.
| |
Collapse
|
14
|
Hu C, Song J, Kwok T, Nguyen EV, Shen X, Daly RJ. Proteome-based molecular subtyping and therapeutic target prediction in gastric cancer. Mol Oncol 2024; 18:1437-1459. [PMID: 38627210 PMCID: PMC11161736 DOI: 10.1002/1878-0261.13654] [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: 08/02/2023] [Revised: 03/12/2024] [Accepted: 04/05/2024] [Indexed: 06/09/2024] Open
Abstract
Different molecular classifications for gastric cancer (GC) have been proposed based on multi-omics platforms with the long-term goal of improved precision treatment. However, the GC (phospho)proteome remains incompletely characterized, particularly at the level of tyrosine phosphorylation. In addition, previous multiomics-based stratification of patient cohorts has lacked identification of corresponding cell line models and comprehensive validation of broad or subgroup-selective therapeutic targets. To address these knowledge gaps, we applied a reverse approach, undertaking the most comprehensive (phospho)proteomic analysis of GC cell lines to date and cross-validating this using publicly available data. Mass spectrometry (MS)-based (phospho)proteomic and tyrosine phosphorylation datasets were subjected to individual or integrated clustering to identify subgroups that were subsequently characterized in terms of enriched molecular processes and pathways. Significant congruence was detected between cell line proteomic and specific patient-derived transcriptomic subclassifications. Many protein kinases exhibiting 'outlier' expression or phosphorylation in the cell line dataset exhibited genomic aberrations in patient samples and association with poor prognosis, with casein kinase I isoform delta/epsilon (CSNK1D/E) being experimentally validated as potential therapeutic targets. Src family kinases were predicted to be commonly hyperactivated in GC cell lines, consistent with broad sensitivity to the next-generation Src inhibitor eCF506. In addition, phosphoproteomic and integrative clustering segregated the cell lines into two subtypes, with epithelial-mesenchyme transition (EMT) and proliferation-associated processes enriched in one, designated the EMT subtype, and metabolic pathways, cell-cell junctions, and the immune response dominating the features of the other, designated the metabolism subtype. Application of kinase activity prediction algorithms and interrogation of gene dependency and drug sensitivity databases predicted that the mechanistic target of rapamycin kinase (mTOR) and dual specificity mitogen-activated protein kinase kinase 2 (MAP2K2) represented potential therapeutic targets for the EMT and metabolism subtypes, respectively, and this was confirmed using selective inhibitors. Overall, our study provides novel, in-depth insights into GC proteomics, kinomics, and molecular taxonomy and reveals potential therapeutic targets that could provide the basis for precision treatments.
Collapse
Affiliation(s)
- Changyuan Hu
- Cancer Program, Biomedicine Discovery InstituteMonash UniversityClaytonAustralia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonAustralia
- Wenzhou Medical University‐Monash BDI Alliance in Clinical and Experimental BiomedicineWenzhou Medical UniversityChina
| | - Jiangning Song
- Cancer Program, Biomedicine Discovery InstituteMonash UniversityClaytonAustralia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonAustralia
| | - Terry Kwok
- Cancer Program, Biomedicine Discovery InstituteMonash UniversityClaytonAustralia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonAustralia
- Infection and Immunity Program, Monash Biomedicine Discovery InstituteMonash UniversityClaytonAustralia
- Department of MicrobiologyMonash UniversityClaytonAustralia
| | - Elizabeth V. Nguyen
- Cancer Program, Biomedicine Discovery InstituteMonash UniversityClaytonAustralia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonAustralia
| | - Xian Shen
- Wenzhou Medical University‐Monash BDI Alliance in Clinical and Experimental BiomedicineWenzhou Medical UniversityChina
- Department of Gastrointestinal Surgery, The First Affiliated HospitalWenzhou Medical UniversityChina
| | - Roger J. Daly
- Cancer Program, Biomedicine Discovery InstituteMonash UniversityClaytonAustralia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonAustralia
| |
Collapse
|
15
|
Zhu Y, Liu F, Jian F, Rong Y. Recent progresses in the late stages of autophagy. CELL INSIGHT 2024; 3:100152. [PMID: 38435435 PMCID: PMC10904915 DOI: 10.1016/j.cellin.2024.100152] [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: 10/18/2023] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 03/05/2024]
Abstract
Autophagy, a lysosome-dependent degradation process, plays a crucial role in maintaining cell homeostasis. It serves as a vital mechanism for adapting to stress and ensuring intracellular quality control. Autophagy deficiencies or defects are linked to numerous human disorders, especially those associated with neuronal degeneration or metabolic diseases. Yoshinori Ohsumi was honored with the Nobel Prize in Physiology or Medicine in 2016 for his groundbreaking discoveries regarding autophagy mechanisms. Over the past few decades, autophagy research has predominantly concentrated on the early stages of autophagy, with relatively limited attention given to the late stages. Nevertheless, recent studies have witnessed substantial advancements in understanding the molecular intricacies of the late stages, which follows autophagosome formation. This review provides a comprehensive summary of the recent progresses in comprehending the molecular mechanisms of the late stages of autophagy.
Collapse
Affiliation(s)
- YanYan Zhu
- School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fengping Liu
- School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fenglei Jian
- School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yueguang Rong
- School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
16
|
Swords SB, Jia N, Norris A, Modi J, Cai Q, Grant BD. A conserved requirement for RME-8/DNAJC13 in neuronal autophagic lysosome reformation. Autophagy 2024; 20:792-808. [PMID: 37942902 PMCID: PMC11062384 DOI: 10.1080/15548627.2023.2269028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 09/16/2023] [Accepted: 10/02/2023] [Indexed: 11/10/2023] Open
Abstract
Autophagosomes fuse with lysosomes, forming autolysosomes that degrade engulfed cargo. To maintain lysosomal capacity, autophagic lysosome reformation (ALR) must regenerate lysosomes from autolysosomes using a membrane tubule-based process. Maintaining lysosomal capacity is required to maintain cellular health, especially in neurons where lysosomal dysfunction has been repeatedly implicated in neurodegenerative disease. The DNA-J domain HSC70 co-chaperone RME-8/DNAJC13 has been linked to endosomal coat protein regulation and to neurological disease. We report new analysis of the requirements for the RME-8/DNAJC13 protein in neurons, focusing on intact C. elegans mechanosensory neurons, and primary mouse cortical neurons in culture. Loss of RME-8/DNAJC13 in both systems results in accumulation of grossly elongated autolysosomal tubules. Further C. elegans analysis revealed a similar autolysosome tubule accumulation defect in mutants known to be required for ALR in mammals, including mutants lacking bec-1/BECN1/Beclin1 and vps-15/PIK3R4/p150 that regulate the class III phosphatidylinositol 3-kinase (PtdIns3K) VPS-34, and dyn-1/dynamin that severs ALR tubules. Clathrin is also an important ALR regulator implicated in autolysosome tubule formation and release. In C. elegans we found that loss of RME-8 causes severe depletion of clathrin from neuronal autolysosomes, a phenotype shared with bec-1 and vps-15 mutants. We conclude that RME-8/DNAJC13 plays a previously unrecognized role in ALR, likely affecting autolysosome tubule severing. Additionally, in both systems, loss of RME-8/DNAJC13 reduced macroautophagic/autophagic flux, suggesting feedback regulation from ALR to autophagy. Our results connecting RME-8/DNAJC13 to ALR and autophagy provide a potential mechanism by which RME-8/DNAJC13 could influence neuronal health and the progression of neurodegenerative disease.Abbreviation: ALR, autophagic lysosome reformation; ATG-13/EPG-1, AuTophaGy (yeast Atg homolog)-13; ATG-18, AuTophaGy (yeast Atg homolog)-18; AV, autophagic vacuole; CLIC-1, Clathrin Light Chain-1; EPG-3, Ectopic P Granules-3; EPG-6, Ectopic P Granules-6; LGG-1, LC3, GABARAP and GATE-16 family-1; MAP1LC3/LC3, microtubule-associated protein 1 light chain 3; PD, Parkinson disease; PtdIns3P, phosphatidylinositol-3-phosphate; PtdIns(4,5)P2, phosphatidylinositol-4,5-bisphosphate; RME-8, Receptor Mediated Endocytosis-8; SNX-1, Sorting NeXin-1; VPS-34, related to yeast Vacuolar Protein Sorting factor-34.
Collapse
Affiliation(s)
- Sierra B. Swords
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, USA
| | - Nuo Jia
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Anne Norris
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, USA
| | - Jil Modi
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, USA
| | - Qian Cai
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Barth D. Grant
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, USA
- Center for Lipid Research, New Brunswick, NJ, USA
| |
Collapse
|
17
|
Tanaka M, Sugimoto K, Akasaka H, Yoshida S, Takahashi T, Fujimoto T, Xie K, Yasunobe Y, Yamamoto K, Hirabayashi T, Nakanishi R, Fujino H, Rakugi H. Effects of interleukin-15 on autophagy regulation in the skeletal muscle of mice. Am J Physiol Endocrinol Metab 2024; 326:E326-E340. [PMID: 38294696 DOI: 10.1152/ajpendo.00311.2023] [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: 09/25/2023] [Revised: 01/19/2024] [Accepted: 01/19/2024] [Indexed: 02/01/2024]
Abstract
This study aimed to evaluate the role of skeletal muscle-derived interleukin (IL)-15 in the regulation of skeletal muscle autophagy using IL-15 knockout (KO) and transgenic (TG) mice. Male C57BL/6 wild-type (WT), IL-15 KO, and IL-15 TG mice were used in this study. Changes in muscle mass, forelimb grip strength, succinate dehydrogenase (SDH) activity, gene and protein expression levels of major regulators and indicators of autophagy, comprehensive gene expression, and DNA methylation in the gastrocnemius muscle were analyzed. Enrichment pathway analyses revealed that the pathology of IL-15 gene deficiency was related to the autophagosome pathway. Moreover, although IL-15 KO mice maintained gastrocnemius muscle mass, they exhibited a decrease in autophagy induction. IL-15 TG mice exhibited a decrease in gastrocnemius muscle mass and an increase in forelimb grip strength and SDH activity in skeletal muscle. In the gastrocnemius muscle, the ratio of phosphorylated adenosine monophosphate-activated protein kinase α (AMPKα) to total AMPKα and unc-51-like autophagy activating kinase 1 and Beclin1 protein expression were higher in the IL-15 TG group than in the WT group. IL-15 gene deficiency induces a decrease in autophagy induction. In contrast, IL-15 overexpression could improve muscle quality by activating autophagy induction while decreasing muscle mass. The regulation of IL-15 in autophagy in skeletal muscles may lead to the development of therapies for the autophagy-induced regulation of skeletal muscle mass and cellular quality control.NEW & NOTEWORTHY IL-15 gene deficiency can decrease autophagy induction. However, although IL-15 overexpression induced a decrease in muscle mass, it led to an improvement in muscle quality. Based on these results, understanding the role of IL-15 in regulating autophagy pathways within skeletal muscle may lead to the development of therapies for the autophagy-induced regulation of skeletal muscle mass and cellular quality control.
Collapse
Affiliation(s)
- Minoru Tanaka
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
- Department of Rehabilitation Science, Osaka Health Science University, Osaka, Japan
| | - Ken Sugimoto
- Department of General and Geriatric Medicine, Kawasaki Medical School, Okayama, Japan
| | - Hiroshi Akasaka
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shino Yoshida
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Toshimasa Takahashi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Taku Fujimoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Keyu Xie
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yukiko Yasunobe
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koichi Yamamoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takumi Hirabayashi
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Ryosuke Nakanishi
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Hiromi Rakugi
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Osaka Rosai Hospital, Osaka, Japan
| |
Collapse
|
18
|
Settembre C, Perera RM. Lysosomes as coordinators of cellular catabolism, metabolic signalling and organ physiology. Nat Rev Mol Cell Biol 2024; 25:223-245. [PMID: 38001393 DOI: 10.1038/s41580-023-00676-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 11/26/2023]
Abstract
Every cell must satisfy basic requirements for nutrient sensing, utilization and recycling through macromolecular breakdown to coordinate programmes for growth, repair and stress adaptation. The lysosome orchestrates these key functions through the synchronised interplay between hydrolytic enzymes, nutrient transporters and signalling factors, which together enable metabolic coordination with other organelles and regulation of specific gene expression programmes. In this Review, we discuss recent findings on lysosome-dependent signalling pathways, focusing on how the lysosome senses nutrient availability through its physical and functional association with mechanistic target of rapamycin complex 1 (mTORC1) and how, in response, the microphthalmia/transcription factor E (MiT/TFE) transcription factors exert feedback regulation on lysosome biogenesis. We also highlight the emerging interactions of lysosomes with other organelles, which contribute to cellular homeostasis. Lastly, we discuss how lysosome dysfunction contributes to diverse disease pathologies and how inherited mutations that compromise lysosomal hydrolysis, transport or signalling components lead to multi-organ disorders with severe metabolic and neurological impact. A deeper comprehension of lysosomal composition and function, at both the cellular and organismal level, may uncover fundamental insights into human physiology and disease.
Collapse
Affiliation(s)
- Carmine Settembre
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy.
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.
| | - Rushika M Perera
- Department of Anatomy, University of California at San Francisco, San Francisco, CA, USA.
- Department of Pathology, University of California at San Francisco, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA.
| |
Collapse
|
19
|
Lourdes SR, Gurung R, Giri S, Mitchell CA, McGrath MJ. A new role for phosphoinositides in regulating mitochondrial dynamics. Adv Biol Regul 2024; 91:101001. [PMID: 38057188 DOI: 10.1016/j.jbior.2023.101001] [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: 11/17/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
Phosphoinositides are a minor group of membrane-associated phospholipids that are transiently generated on the cytoplasmic leaflet of many organelle membranes and the plasma membrane. There are seven functionally distinct phosphoinositides, each derived via the reversible phosphorylation of phosphatidylinositol in various combinations on the inositol ring. Their generation and termination is tightly regulated by phosphatidylinositol-kinases and -phosphatases. These enzymes can function together in an integrated and coordinated manner, whereby the phosphoinositide product of one enzyme may subsequently serve as a substrate for another to generate a different phosphoinositide species. This regulatory mechanism not only enables the transient generation of phosphoinositides on membranes, but also more complex sequential or bidirectional conversion pathways, and phosphoinositides can also be transferred between organelles via membrane contacts. It is this capacity to fine-tune phosphoinositide signals that makes them ideal regulators of membrane organization and dynamics, through their recruitment of signalling, membrane altering and lipid transfer proteins. Research spanning several decades has provided extensive evidence that phosphoinositides are major gatekeepers of membrane organization, with roles in endocytosis, exocytosis, autophagy, lysosome dynamics, vesicular transport and secretion, cilia, inter-organelle membrane contact, endosome maturation and nuclear function. By contrast, there has been remarkably little known about the role of phosphoinositides at mitochondria - an enigmatic and major knowledge gap, with challenges in reliably detecting phosphoinositides at this site. Here we review recent significant breakthroughs in understanding the role of phosphoinositides in regulating mitochondrial dynamics and metabolic function.
Collapse
Affiliation(s)
- Sonia Raveena Lourdes
- Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Rajendra Gurung
- Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Saveen Giri
- Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Christina A Mitchell
- Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
| | - Meagan J McGrath
- Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
20
|
Du J, Dong Y, Zhu H, Deng Y, Sa C, Yu Q, Li M. DNA damage-induced autophagy is regulated by inositol polyphosphate synthetases in Candida albicans. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119622. [PMID: 37913846 DOI: 10.1016/j.bbamcr.2023.119622] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 10/16/2023] [Accepted: 10/22/2023] [Indexed: 11/03/2023]
Abstract
DNA damage-induced autophagy is a new type of autophagy that differs from traditional macroautophagy; however, this type of autophagy has not been identified in the pathogenic fungus Candida albicans. Inositol polyphosphates are involved in the regulation of DNA damage repair and macroautophagy; however, whether inositol polyphosphates are involved in the regulation of DNA damage-induced autophagy remains unclear. In this study, we identified DNA damage-induced autophagy in C. albicans and systematically investigated the mechanisms of inositol polyphosphate pathway regulation. We found that the core machinery of macro autophagy is also essential for DNA damage-induced autophagy, and that inositol polyphosphate synthetases Kcs1, Ipk1, and Vip1 play a critical role in autophagy. In this study, we focused on Kcs1 and Vip1, which are responsible for the synthesis of inositol pyrophosphate. The kcs1Δ/Δ and vip1Δ/Δ strains exhibited reduced number of phagophore assembly sites (PAS) and autophagic bodies. The recruitment of autophagy-related gene 1 (Atg1) to PAS was significantly affected in the kcs1Δ/Δ and vip1Δ/Δ strains. Target of rapamycin complex 1 kinase activity was elevated in kcs1Δ/Δ and vip1Δ/Δ strains, which significantly inhibited the initiation of autophagy. Atg18 Localization was altered in these mutants. The absence of Kcs1 or Vip1 caused the downregulation of RAD53, a key gene in the DNA damage response. These data provide further understanding of the mechanism of autophagy regulation in C. albicans.
Collapse
Affiliation(s)
- Jiawen Du
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yixuan Dong
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Hangqi Zhu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Ying Deng
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Chula Sa
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Qilin Yu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Mingchun Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
21
|
Srivastav S, van der Graaf K, Singh P, Utama AB, Meyer MD, McNew JA, Stern M. Atl (atlastin) regulates mTor signaling and autophagy in Drosophila muscle through alteration of the lysosomal network. Autophagy 2024; 20:131-150. [PMID: 37649246 PMCID: PMC10761077 DOI: 10.1080/15548627.2023.2249794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 08/08/2023] [Accepted: 08/11/2023] [Indexed: 09/01/2023] Open
Abstract
ABBREVIATIONS atl atlastin; ALR autophagic lysosome reformation; ER endoplasmic reticulum; GFP green fluorescent protein; HSP hereditary spastic paraplegia; Lamp1 lysosomal associated membrane protein 1 PolyUB polyubiquitin; RFP red fluorescent protein; spin spinster; mTor mechanistic Target of rapamycin; VCP valosin containing protein.
Collapse
Affiliation(s)
| | | | - Pratibha Singh
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | | | - Matthew D. Meyer
- Shared Equipment Authority, Rice University, Houston, Texas, USA
| | - James A. McNew
- Department of BioSciences, Rice University, Houston, Texas, USA
| | - Michael Stern
- Department of BioSciences, Rice University, Houston, Texas, USA
| |
Collapse
|
22
|
Rodríguez MP, Cabello-Verrugio C. Soluble Factors Associated with Denervation-induced Skeletal Muscle Atrophy. Curr Protein Pept Sci 2024; 25:189-199. [PMID: 38018212 DOI: 10.2174/0113892037189827231018092036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 08/22/2023] [Accepted: 09/03/2023] [Indexed: 11/30/2023]
Abstract
Skeletal muscle tissue has the critical function of mechanical support protecting the body. In addition, its functions are strongly influenced by the balanced synthesis and degradation processes of structural and regulatory proteins. The inhibition of protein synthesis and/or the activation of catabolism generally determines a pathological state or condition called muscle atrophy, a reduction in muscle mass that results in partial or total loss of function. It has been established that many pathophysiological conditions can cause a decrease in muscle mass. Skeletal muscle innervation involves stable and functional neural interactions with muscles via neuromuscular junctions and is essential for maintaining normal muscle structure and function. Loss of motor innervation induces rapid skeletal muscle fiber degeneration with activation of atrophy-related signaling and subsequent disassembly of sarcomeres, altering normal muscle function. After denervation, an inflammation stage is characterized by the increased expression of pro-inflammatory cytokines that determine muscle atrophy. In this review, we highlighted the impact of some soluble factors on the development of muscle atrophy by denervation.
Collapse
Affiliation(s)
- Marianny Portal Rodríguez
- Laboratory of Muscle Pathology, Fragility, and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility, and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, 8370146, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
23
|
Xie G, Jin H, Mikhail H, Pavel V, Yang G, Ji B, Lu B, Li Y. Autophagy in sarcopenia: Possible mechanisms and novel therapies. Biomed Pharmacother 2023; 165:115147. [PMID: 37473679 DOI: 10.1016/j.biopha.2023.115147] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/23/2023] [Accepted: 07/07/2023] [Indexed: 07/22/2023] Open
Abstract
With global population aging, age-related diseases, especially sarcopenia, have attracted much attention in recent years. Characterized by low muscle strength, low muscle quantity or quality and low physical performance, sarcopenia is one of the major factors associated with an increased risk of falls and disability. Much effort has been made to understand the cellular biological and physiological mechanisms underlying sarcopenia. Autophagy is an important cellular self-protection mechanism that relies on lysosomes to degrade misfolded proteins and damaged organelles. Research designed to obtain new insight into human diseases from the autophagic aspect has been carried out and has made new progress, which encourages relevant studies on the relationship between autophagy and sarcopenia. Autophagy plays a protective role in sarcopenia by modulating the regenerative capability of satellite cells, relieving oxidative stress and suppressing the inflammatory response. This review aims to reveal the specific interaction between sarcopenia and autophagy and explore possible therapies in hopes of encouraging more specific research in need and unlocking novel promising therapies to ameliorate sarcopenia.
Collapse
Affiliation(s)
- Guangyang Xie
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; Xiangya School of Medicine, Central South University, Changsha 410008, Hunan, China
| | - Hongfu Jin
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Herasimenka Mikhail
- Republican Scientific and Practical Center of Traumatology and Orthopedics, Minsk 220024, Belarus
| | - Volotovski Pavel
- Republican Scientific and Practical Center of Traumatology and Orthopedics, Minsk 220024, Belarus
| | - Guang Yang
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Bingzhou Ji
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Bangbao Lu
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha 410008, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.
| |
Collapse
|
24
|
Mutvei AP, Nagiec MJ, Blenis J. Balancing lysosome abundance in health and disease. Nat Cell Biol 2023; 25:1254-1264. [PMID: 37580388 DOI: 10.1038/s41556-023-01197-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 06/28/2023] [Indexed: 08/16/2023]
Abstract
Lysosomes are catabolic organelles that govern numerous cellular processes, including macromolecule degradation, nutrient signalling and ion homeostasis. Aberrant changes in lysosome abundance are implicated in human diseases. Here we outline the mechanisms of lysosome biogenesis and turnover, and discuss how changes in the lysosome pool impact physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Anders P Mutvei
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Huddinge, Sweden.
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | - Michal J Nagiec
- Meyer Cancer Center and Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA
| | - John Blenis
- Meyer Cancer Center and Department of Pharmacology, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
25
|
Abstract
Phosphoinositides (PIs) are phospholipids derived from phosphatidylinositol. PIs are regulated via reversible phosphorylation, which is directed by the opposing actions of PI kinases and phosphatases. PIs constitute a minor fraction of the total cellular lipid pool but play pleiotropic roles in multiple aspects of cell biology. Genetic mutations of PI regulatory enzymes have been identified in rare congenital developmental syndromes, including ciliopathies, and in numerous human diseases, such as cancer and metabolic and neurological disorders. Accordingly, PI regulatory enzymes have been targeted in the design of potential therapeutic interventions for human diseases. Recent advances place PIs as central regulators of membrane dynamics within functionally distinct subcellular compartments. This brief review focuses on the emerging role PIs play in regulating cell signaling within the primary cilium and in directing transfer of molecules at interorganelle membrane contact sites and identifies new roles for PIs in subcellular spaces.
Collapse
Affiliation(s)
- Elizabeth Michele Davies
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Christina Anne Mitchell
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Harald Alfred Stenmark
- Department of Molecular Cell Biology, Institute for Cancer Research. The Norwegian Radium Hospital, Montebello, N-0379 Oslo, Norway
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Montebello, N-0379 Oslo, Norway
| |
Collapse
|
26
|
Yuan W, Weaver YM, Earnest S, Taylor CA, Cobb MH, Weaver BP. Modulating p38 MAPK signaling by proteostasis mechanisms supports tissue integrity during growth and aging. Nat Commun 2023; 14:4543. [PMID: 37507441 PMCID: PMC10382525 DOI: 10.1038/s41467-023-40317-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
The conserved p38 MAPK family is activated by phosphorylation during stress responses and inactivated by phosphatases. C. elegans PMK-1 p38 MAPK initiates innate immune responses and blocks development when hyperactivated. Here we show that PMK-1 signaling is enhanced during early aging by modulating the stoichiometry of non-phospho-PMK-1 to promote tissue integrity and longevity. Loss of pmk-1 function accelerates progressive declines in neuronal integrity and lysosome function compromising longevity which has both cell autonomous and cell non-autonomous contributions. CED-3 caspase cleavage limits phosphorylated PMK-1. Enhancing p38 signaling with caspase cleavage-resistant PMK-1 protects lysosomal and neuronal integrity extending a youthful phase. PMK-1 works through a complex transcriptional program to regulate lysosome formation. During early aging, the absolute phospho-p38 amount is maintained but the reservoir of non-phospho-p38 diminishes to enhance signaling without hyperactivation. Our findings show that modulating the stoichiometry of non-phospho-p38 dynamically supports tissue-homeostasis during aging without hyper-activation of stress response.
Collapse
Affiliation(s)
- Wang Yuan
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Yi M Weaver
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Svetlana Earnest
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Clinton A Taylor
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Melanie H Cobb
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Benjamin P Weaver
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
27
|
Calcagni' A, Staiano L, Zampelli N, Minopoli N, Herz NJ, Di Tullio G, Huynh T, Monfregola J, Esposito A, Cirillo C, Bajic A, Zahabiyon M, Curnock R, Polishchuk E, Parkitny L, Medina DL, Pastore N, Cullen PJ, Parenti G, De Matteis MA, Grumati P, Ballabio A. Loss of the batten disease protein CLN3 leads to mis-trafficking of M6PR and defective autophagic-lysosomal reformation. Nat Commun 2023; 14:3911. [PMID: 37400440 DOI: 10.1038/s41467-023-39643-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/21/2023] [Indexed: 07/05/2023] Open
Abstract
Batten disease, one of the most devastating types of neurodegenerative lysosomal storage disorders, is caused by mutations in CLN3. Here, we show that CLN3 is a vesicular trafficking hub connecting the Golgi and lysosome compartments. Proteomic analysis reveals that CLN3 interacts with several endo-lysosomal trafficking proteins, including the cation-independent mannose 6 phosphate receptor (CI-M6PR), which coordinates the targeting of lysosomal enzymes to lysosomes. CLN3 depletion results in mis-trafficking of CI-M6PR, mis-sorting of lysosomal enzymes, and defective autophagic lysosomal reformation. Conversely, CLN3 overexpression promotes the formation of multiple lysosomal tubules, which are autophagy and CI-M6PR-dependent, generating newly formed proto-lysosomes. Together, our findings reveal that CLN3 functions as a link between the M6P-dependent trafficking of lysosomal enzymes and lysosomal reformation pathway, explaining the global impairment of lysosomal function in Batten disease.
Collapse
Affiliation(s)
- Alessia Calcagni'
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA.
| | - Leopoldo Staiano
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Institute for Genetic and Biomedical Research, National Research Council (CNR), Milan, Italy
| | | | - Nadia Minopoli
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Translational Medical Sciences, Federico II University, 80131, Naples, Italy
| | - Niculin J Herz
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | | | - Tuong Huynh
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | | | - Alessandra Esposito
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- SSM School for Advanced Studies, Federico II University, Naples, Italy
| | - Carmine Cirillo
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Aleksandar Bajic
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Mahla Zahabiyon
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Rachel Curnock
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Elena Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Luke Parkitny
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Diego Luis Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Translational Medical Sciences, Federico II University, 80131, Naples, Italy
| | - Nunzia Pastore
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Translational Medical Sciences, Federico II University, 80131, Naples, Italy
| | - Peter J Cullen
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Giancarlo Parenti
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Translational Medical Sciences, Federico II University, 80131, Naples, Italy
| | - Maria Antonietta De Matteis
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, Naples, Italy
| | - Paolo Grumati
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Andrea Ballabio
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA.
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy.
- Department of Translational Medical Sciences, Federico II University, 80131, Naples, Italy.
- SSM School for Advanced Studies, Federico II University, Naples, Italy.
| |
Collapse
|
28
|
Shukla D, Gural BM, Cauley ES, Battula N, Mowla S, Karas BF, Roberts LE, Cavallo L, Turkalj L, Moody SA, Swan LE, Manzini MC. Duplicated zebrafish (Danio rerio) inositol phosphatases inpp5ka and inpp5kb diverged in expression pattern and function. Dev Genes Evol 2023; 233:25-34. [PMID: 37184573 PMCID: PMC10239392 DOI: 10.1007/s00427-023-00703-z] [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: 08/31/2022] [Accepted: 04/27/2023] [Indexed: 05/16/2023]
Abstract
One hurdle in the development of zebrafish models of human disease is the presence of multiple zebrafish orthologs resulting from whole genome duplication in teleosts. Mutations in inositol polyphosphate 5-phosphatase K (INPP5K) lead to a syndrome characterized by variable presentation of intellectual disability, brain abnormalities, cataracts, muscle disease, and short stature. INPP5K is a phosphatase acting at position 5 of phosphoinositides to control their homeostasis and is involved in insulin signaling, cytoskeletal regulation, and protein trafficking. Previously, our group and others have replicated the human phenotypes in zebrafish knockdown models by targeting both INPP5K orthologs inpp5ka and inpp5kb. Here, we show that inpp5ka is the more closely related orthologue to human INPP5K. While both inpp5ka and inpp5kb mRNA expression levels follow a similar trend in the developing head, eyes, and tail, inpp5ka is much more abundantly expressed in these tissues than inpp5kb. In situ hybridization revealed a similar trend, also showing unique localization of inpp5kb in the pineal gland and retina indicating different transcriptional regulation. We also found that inpp5kb has lost its catalytic activity against its preferred substrate, PtdIns(4,5)P2. Since most human mutations are missense changes disrupting phosphatase activity, we propose that loss of inpp5ka alone can be targeted to recapitulate the human presentation. In addition, we show that the function of inpp5kb has diverged from inpp5ka and may play a novel role in the zebrafish.
Collapse
Affiliation(s)
- Dhyanam Shukla
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA
| | - Brian M Gural
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA
| | - Edmund S Cauley
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Namarata Battula
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA
| | - Shorbon Mowla
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA
| | - Brittany F Karas
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA
| | - Llion E Roberts
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Luca Cavallo
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA
| | - Luka Turkalj
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA
| | - Sally A Moody
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Laura E Swan
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - M Chiara Manzini
- Department of Neuroscience and Cell Biology and Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 89 French Street, CHINJ Rm 3274, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
29
|
Daly JL, Danson CM, Lewis PA, Zhao L, Riccardo S, Di Filippo L, Cacchiarelli D, Lee D, Cross SJ, Heesom KJ, Xiong WC, Ballabio A, Edgar JR, Cullen PJ. Multi-omic approach characterises the neuroprotective role of retromer in regulating lysosomal health. Nat Commun 2023; 14:3086. [PMID: 37248224 PMCID: PMC10227043 DOI: 10.1038/s41467-023-38719-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 05/05/2023] [Indexed: 05/31/2023] Open
Abstract
Retromer controls cellular homeostasis through regulating integral membrane protein sorting and transport and by controlling maturation of the endo-lysosomal network. Retromer dysfunction, which is linked to neurodegenerative disorders including Parkinson's and Alzheimer's diseases, manifests in complex cellular phenotypes, though the precise nature of this dysfunction, and its relation to neurodegeneration, remain unclear. Here, we perform an integrated multi-omics approach to provide precise insight into the impact of Retromer dysfunction on endo-lysosomal health and homeostasis within a human neuroglioma cell model. We quantify widespread changes to the lysosomal proteome, indicative of broad lysosomal dysfunction and inefficient autophagic lysosome reformation, coupled with a reconfigured cell surface proteome and secretome reflective of increased lysosomal exocytosis. Through this global proteomic approach and parallel transcriptomic analysis, we provide a holistic view of Retromer function in regulating lysosomal homeostasis and emphasise its role in neuroprotection.
Collapse
Affiliation(s)
- James L Daly
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol, BS8 1TD, UK.
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, Guy's Hospital, King's College London, SE1 9RT, London, UK.
| | - Chris M Danson
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol, BS8 1TD, UK
| | - Philip A Lewis
- Bristol Proteomics Facility, School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, BS8 1TD, Bristol, UK
| | - Lu Zhao
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Sara Riccardo
- Telethon Institute of Genetics and Medicine, Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- Next Generation Diagnostic srl, Pozzuoli, Italy
| | - Lucio Di Filippo
- Telethon Institute of Genetics and Medicine, Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- Next Generation Diagnostic srl, Pozzuoli, Italy
| | - Davide Cacchiarelli
- Telethon Institute of Genetics and Medicine, Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
- School for Advanced Studies, University of Naples "Federico II", Naples, Italy
| | - Daehoon Lee
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Stephen J Cross
- Wolfson Bioimaging Facility, Faculty of Biomedical Sciences, University of Bristol, Bristol, UK
| | - Kate J Heesom
- Bristol Proteomics Facility, School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, BS8 1TD, Bristol, UK
| | - Wen-Cheng Xiong
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli, Italy
- Department of Translational Medicine, University of Naples "Federico II", Naples, Italy
- School for Advanced Studies, University of Naples "Federico II", Naples, Italy
- Department of Molecular and Human Genetics and Neurological Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - James R Edgar
- Department of Pathology, Cambridge University, Tennis Court Road, Cambridge, UK
| | - Peter J Cullen
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
30
|
Davies EM, Gurung R, Le KQ, Roan KT, Harvey RP, Mitchell GM, Schwarz Q, Mitchell CA. PI(4,5)P 2-dependent regulation of endothelial tip cell specification contributes to angiogenesis. SCIENCE ADVANCES 2023; 9:eadd6911. [PMID: 37000875 PMCID: PMC10065449 DOI: 10.1126/sciadv.add6911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 02/24/2023] [Indexed: 06/19/2023]
Abstract
Dynamic positioning of endothelial tip and stalk cells, via the interplay between VEGFR2 and NOTCH signaling, is essential for angiogenesis. VEGFR2 activates PI3K, which phosphorylates PI(4,5)P2 to PI(3,4,5)P3, activating AKT; however, PI3K/AKT does not direct tip cell specification. We report that PI(4,5)P2 hydrolysis by the phosphoinositide-5-phosphatase, INPP5K, contributes to angiogenesis. INPP5K ablation disrupted tip cell specification and impaired embryonic angiogenesis associated with enhanced DLL4/NOTCH signaling. INPP5K degraded a pool of PI(4,5)P2 generated by PIP5K1C phosphorylation of PI(4)P in endothelial cells. INPP5K ablation increased PI(4,5)P2, thereby releasing β-catenin from the plasma membrane, and concurrently increased PI(3,4,5)P3-dependent AKT activation, conditions that licensed DLL4/NOTCH transcription. Suppression of PI(4,5)P2 in INPP5K-siRNA cells by PIP5K1C-siRNA, restored β-catenin membrane localization and normalized AKT signaling. Pharmacological NOTCH or AKT inhibition in vivo or genetic β-catenin attenuation rescued angiogenesis defects in INPP5K-null mice. Therefore, PI(4,5)P2 is critical for β-catenin/DLL4/NOTCH signaling, which governs tip cell specification during angiogenesis.
Collapse
Affiliation(s)
- Elizabeth M. Davies
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Rajendra Gurung
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Kai Qin Le
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Katherine T. T. Roan
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Richard P. Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia
- School of Clinical Medicine and School of Biotechnology and Biomolecular Science, University of New South Wales, Kensington, New South Wales 2052, Australia
| | - Geraldine M. Mitchell
- O’Brien Institute Department of St Vincent’s Institute and University of Melbourne, Department of Surgery, St. Vincent’s Hospital, Fitzroy, Victoria 3065, Australia
- Health Sciences Faculty, Australian Catholic University, Fitzroy, Victoria 3065, Australia
| | - Quenten Schwarz
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia 5001, Australia
| | - Christina A. Mitchell
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| |
Collapse
|
31
|
Rowland LA, Guilherme A, Henriques F, DiMarzio C, Munroe S, Wetoska N, Kelly M, Reddig K, Hendricks G, Pan M, Han X, Ilkayeva OR, Newgard CB, Czech MP. De novo lipogenesis fuels adipocyte autophagosome and lysosome membrane dynamics. Nat Commun 2023; 14:1362. [PMID: 36914626 PMCID: PMC10011520 DOI: 10.1038/s41467-023-37016-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
Adipocytes robustly synthesize fatty acids (FA) from carbohydrate through the de novo lipogenesis (DNL) pathway, yet surprisingly DNL contributes little to their abundant triglyceride stored in lipid droplets. This conundrum raises the hypothesis that adipocyte DNL instead enables membrane expansions to occur in processes like autophagy, which requires an abundant supply of phospholipids. We report here that adipocyte Fasn deficiency in vitro and in vivo markedly impairs autophagy, evident by autophagosome accumulation and severely compromised degradation of the autophagic substrate p62. Our data indicate the impairment occurs at the level of autophagosome-lysosome fusion, and indeed, loss of Fasn decreases certain membrane phosphoinositides necessary for autophagosome and lysosome maturation and fusion. Autophagy dependence on FA produced by Fasn is not fully alleviated by exogenous FA in cultured adipocytes, and interestingly, imaging studies reveal that Fasn colocalizes with nascent autophagosomes. Together, our studies identify DNL as a critical source of FAs to fuel autophagosome and lysosome maturation and fusion in adipocytes.
Collapse
Affiliation(s)
- Leslie A Rowland
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| | - Adilson Guilherme
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Felipe Henriques
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Chloe DiMarzio
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Sean Munroe
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Nicole Wetoska
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Mark Kelly
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Keith Reddig
- Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Gregory Hendricks
- Department of Radiology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Meixia Pan
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Olga R Ilkayeva
- Duke Molecular Physiology Institute, Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Christopher B Newgard
- Duke Molecular Physiology Institute, Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
32
|
Swords S, Jia N, Norris A, Modi J, Cai Q, Grant BD. A Conserved Requirement for RME-8/DNAJC13 in Neuronal Autolysosome Reformation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.27.530319. [PMID: 36909501 PMCID: PMC10002637 DOI: 10.1101/2023.02.27.530319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Autophagosomes fuse with lysosomes, forming autolysosomes that degrade engulfed cargo. To maintain lysosomal capacity, autolysosome reformation (ALR) must regenerate lysosomes from autolysosomes using a membrane tubule-based process. Maintaining lysosomal capacity is required to maintain proteostasis and cellular health, especially in neurons where lysosomal dysfunction has been repeatedly implicated in neurodegenerative disease. Cell biological studies have linked the DNA-J domain Hsc70 co-chaperone RME-8/DNAJC13 to endosomal coat protein regulation, while human genetics studies have linked RME-8/DNAJC13 to neurological disease, including Parkinsonism and Essential Tremor. We report new analysis of the requirements for the RME-8/DNAJC13 protein in neurons, focusing on C. elegans mechanosensory neurons in the intact animal, and in primary mouse cortical neurons in culture. We find that loss of RME-8/DNAJC13 in both systems results in accumulation of grossly elongated autolysosomal tubules. Further C. elegans analysis revealed a similar autolysosome tubule accumulation defect in mutants known to be required for ALR in mammals, including bec-1/beclin and vps-15/PIK3R4/p150 that regulate type-III PI3-kinase VPS-34, and dyn-1/dynamin that severs ALR tubules. Clathrin is also an important ALR regulator implicated in autolysosome tubule formation and release. In C. elegans we found that loss of RME-8 causes severe depletion of clathrin from neuronal autolysosomes, a phenotype shared with bec-1 and vps-15 mutants. We conclude that RME-8/DNAJC13 plays a conserved but previously unrecognized role in autolysosome reformation, likely affecting ALR tubule initiation and/or severing. Additionally, in both systems, we found that loss of RME-8/DNAJC13 appeared to reduce autophagic flux, suggesting feedback regulation from ALR to autophagy. Our results connecting RME-8/DNAJC13 to ALR and autophagy provide a potential mechanism by which RME-8/DNAJC13 could influence neuronal health and the progression of neurodegenerative disease.
Collapse
Affiliation(s)
- Sierra Swords
- Department of Molecular Biology and Biochemistry Rutgers University, Piscataway, NJ USA, 08854
| | - Nuo Jia
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ USA, 08854
| | - Anne Norris
- Department of Molecular Biology and Biochemistry Rutgers University, Piscataway, NJ USA, 08854
| | - Jil Modi
- Department of Molecular Biology and Biochemistry Rutgers University, Piscataway, NJ USA, 08854
| | - Qian Cai
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ USA, 08854
| | - Barth D. Grant
- Department of Molecular Biology and Biochemistry Rutgers University, Piscataway, NJ USA, 08854
- Center for Lipid Research, New Brunswick, NJ USA 08901
| |
Collapse
|
33
|
Sugar transporter Slc37a2 regulates bone metabolism in mice via a tubular lysosomal network in osteoclasts. Nat Commun 2023; 14:906. [PMID: 36810735 PMCID: PMC9945426 DOI: 10.1038/s41467-023-36484-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 02/01/2023] [Indexed: 02/23/2023] Open
Abstract
Osteoclasts are giant bone-digesting cells that harbor specialized lysosome-related organelles termed secretory lysosomes (SLs). SLs store cathepsin K and serve as a membrane precursor to the ruffled border, the osteoclast's 'resorptive apparatus'. Yet, the molecular composition and spatiotemporal organization of SLs remains incompletely understood. Here, using organelle-resolution proteomics, we identify member a2 of the solute carrier 37 family (Slc37a2) as a SL sugar transporter. We demonstrate in mice that Slc37a2 localizes to the SL limiting membrane and that these organelles adopt a hitherto unnoticed but dynamic tubular network in living osteoclasts that is required for bone digestion. Accordingly, mice lacking Slc37a2 accrue high bone mass owing to uncoupled bone metabolism and disturbances in SL export of monosaccharide sugars, a prerequisite for SL delivery to the bone-lining osteoclast plasma membrane. Thus, Slc37a2 is a physiological component of the osteoclast's unique secretory organelle and a potential therapeutic target for metabolic bone diseases.
Collapse
|
34
|
Serra-Vinardell J, Sandler MB, De Pace R, Manzella-Lapeira J, Cougnoux A, Keyvanfar K, Introne WJ, Brzostowski JA, Ward ME, Gahl WA, Sharma P, Malicdan MCV. LYST deficiency impairs autophagic lysosome reformation in neurons and alters lysosome number and size. Cell Mol Life Sci 2023; 80:53. [PMID: 36707427 PMCID: PMC11072721 DOI: 10.1007/s00018-023-04695-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/11/2022] [Accepted: 01/08/2023] [Indexed: 01/29/2023]
Abstract
Chediak-Higashi syndrome (CHS) is a rare, autosomal recessive disorder caused by biallelic mutations in the lysosomal trafficking regulator (LYST) gene. Even though enlarged lysosomes and/or lysosome-related organelles (LROs) are the typical cellular hallmarks of CHS, they have not been investigated in human neuronal models. Moreover, how and why the loss of LYST function causes a lysosome phenotype in cells has not been elucidated. We report that the LYST-deficient human neuronal model exhibits lysosome depletion accompanied by hyperelongated tubules extruding from enlarged autolysosomes. These results have also been recapitulated in neurons differentiated from CHS patients' induced pluripotent stem cells (iPSCs), validating our model system. We propose that LYST ensures the correct fission/scission of the autolysosome tubules during autophagic lysosome reformation (ALR), a crucial process to restore the number of free lysosomes after autophagy. We further demonstrate that LYST is recruited to the lysosome membrane, likely to facilitate the fission of autolysosome tubules. Together, our results highlight the key role of LYST in maintaining lysosomal homeostasis following autophagy and suggest that ALR dysregulation is likely associated with the neurodegenerative CHS phenotype.
Collapse
Affiliation(s)
- Jenny Serra-Vinardell
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Maxwell B Sandler
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Raffaella De Pace
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Javier Manzella-Lapeira
- Twinbrook Imaging Facility, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | - Antony Cougnoux
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Keyvan Keyvanfar
- National Heart, Lung, and Blood Institute, Flow Cytometry Facility, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Wendy J Introne
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Joseph A Brzostowski
- Twinbrook Imaging Facility, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | - Michael E Ward
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| | - William A Gahl
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Undiagnosed Diseases Program, National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Common Fund, Office of the Director, NIH, Bethesda, MD, 20892, USA
| | - Prashant Sharma
- Undiagnosed Diseases Program, National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Common Fund, Office of the Director, NIH, Bethesda, MD, 20892, USA.
| | - May Christine V Malicdan
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Undiagnosed Diseases Program, National Human Genome Research Institute (NHGRI), National Institutes of Health (NIH), Common Fund, Office of the Director, NIH, Bethesda, MD, 20892, USA
| |
Collapse
|
35
|
Zhang M, Chen M, Li Y, Rao M, Wang D, Wang Z, Zhang L, Yin P, Tang P. Delayed denervation-induced muscle atrophy in Opg knockout mice. Front Physiol 2023; 14:1127474. [PMID: 36909232 PMCID: PMC9992212 DOI: 10.3389/fphys.2023.1127474] [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: 12/19/2022] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Recent evidence has shown a crucial role for the osteoprotegerin/receptor activator of nuclear factor κ-B ligand/RANK (OPG/RANKL/RANK) signaling axis not only in bone but also in muscle tissue; however, there is still a lack of understanding of its effects on muscle atrophy. Here, we found that denervated Opg knockout mice displayed better functional recovery and delayed muscle atrophy, especially in a specific type IIB fiber. Moreover, OPG deficiency promoted milder activation of the ubiquitin-proteasome pathway, which further verified the protective role of Opg knockout in denervated muscle damage. Furthermore, transcriptome sequencing indicated that Opg knockout upregulated the expression of Inpp5k, Rbm3, and Tet2 and downregulated that of Deptor in denervated muscle. In vitro experiments revealed that satellite cells derived from Opg knockout mice displayed a better differentiation ability than those acquired from wild-type littermates. Higher expression levels of Tet2 were also observed in satellite cells derived from Opg knockout mice, which provided a possible mechanistic basis for the protective effects of Opg knockout on muscle atrophy. Taken together, our findings uncover the novel role of Opg in muscle atrophy process and extend the current understanding in the OPG/RANKL/RANK signaling axis.
Collapse
Affiliation(s)
- Mingming Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Ming Chen
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Yi Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Man Rao
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Duanyang Wang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhongqi Wang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Licheng Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Pengbin Yin
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| | - Peifu Tang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, China
| |
Collapse
|
36
|
Qin Y, Medina MW. Mechanism of the Regulation of Plasma Cholesterol Levels by PI(4,5)P 2. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:89-119. [PMID: 36988878 DOI: 10.1007/978-3-031-21547-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Elevated low-density lipoprotein (LDL) cholesterol (LDLc) is one of the most well-established risk factors for cardiovascular disease, while high levels of high-density lipoprotein (HDL) cholesterol (HDLc) have been associated with protection from cardiovascular disease. Cardiovascular disease remains one of the leading causes of death worldwide; thus it is important to understand mechanisms that impact LDLc and HDLc metabolism. In this chapter, we will discuss molecular processes by which phosphatidylinositol-(4,5)-bisphosphate, PI(4,5)P2, is thought to modulate LDLc or HDLc. Section 1 will provide an overview of cholesterol in the circulation, discussing processes that modulate the various forms of lipoproteins (LDL and HDL) carrying cholesterol. Section 2 will describe how a PI(4,5)P2 phosphatase, transmembrane protein 55B (TMEM55B), impacts circulating LDLc levels through its ability to regulate lysosomal decay of the low-density lipoprotein receptor (LDLR), the primary receptor for hepatic LDL uptake. Section 3 will discuss how PI(4,5)P2 interacts with apolipoprotein A-I (apoA1), the key apolipoprotein on HDL. In addition to direct mechanisms of PI(4,5)P2 action on circulating cholesterol, Sect. 4 will review how PI(4,5)P2 may indirectly impact LDLc and HDLc by affecting insulin action. Last, as cholesterol is controlled through intricate negative feedback loops, Sect. 5 will describe how PI(4,5)P2 is regulated by cholesterol.
Collapse
Affiliation(s)
- Yuanyuan Qin
- Department of Pediatrics, Division of Cardiology, University of California, San Francisco, Oakland, CA, USA
| | - Marisa W Medina
- Department of Pediatrics, Division of Cardiology, University of California, San Francisco, Oakland, CA, USA.
| |
Collapse
|
37
|
Nanayakkara R, Gurung R, Rodgers SJ, Eramo MJ, Ramm G, Mitchell CA, McGrath MJ. Autophagic lysosome reformation in health and disease. Autophagy 2022:1-18. [DOI: 10.1080/15548627.2022.2128019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
- Randini Nanayakkara
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Monash Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, Victoria, Australia
| | - Rajendra Gurung
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Samuel J. Rodgers
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Matthew J. Eramo
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Georg Ramm
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
- Monash Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, Victoria, Australia
| | - Christina A. Mitchell
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Meagan J. McGrath
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
38
|
Functional Nutrients to Ameliorate Neurogenic Muscle Atrophy. Metabolites 2022; 12:metabo12111149. [DOI: 10.3390/metabo12111149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Neurogenic muscle atrophy is a debilitating condition that occurs from nerve trauma in association with diseases or during aging, leading to reduced interaction between motoneurons and skeletal fibers. Current therapeutic approaches aiming at preserving muscle mass in a scenario of decreased nervous input include physical activity and employment of drugs that slow down the progression of the condition yet provide no concrete resolution. Nutritional support appears as a precious tool, adding to the success of personalized medicine, and could thus play a relevant part in mitigating neurogenic muscle atrophy. We herein summarize the molecular pathways triggered by denervation of the skeletal muscle that could be affected by functional nutrients. In this narrative review, we examine and discuss studies pertaining to the use of functional ingredients to counteract neurogenic muscle atrophy, focusing on their preventive or curative means of action within the skeletal muscle. We reviewed experimental models of denervation in rodents and in amyotrophic lateral sclerosis, as well as that caused by aging, considering the knowledge generated with use of animal experimental models and, also, from human studies.
Collapse
|
39
|
Jani RA, Di Cicco A, Keren-Kaplan T, Vale-Costa S, Hamaoui D, Hurbain I, Tsai FC, Di Marco M, Macé AS, Zhu Y, Amorim MJ, Bassereau P, Bonifacino JS, Subtil A, Marks MS, Lévy D, Raposo G, Delevoye C. PI4P and BLOC-1 remodel endosomal membranes into tubules. J Biophys Biochem Cytol 2022; 221:213508. [PMID: 36169638 PMCID: PMC9524204 DOI: 10.1083/jcb.202110132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 07/25/2022] [Accepted: 08/31/2022] [Indexed: 12/11/2022] Open
Abstract
Intracellular trafficking is mediated by transport carriers that originate by membrane remodeling from donor organelles. Tubular carriers contribute to the flux of membrane lipids and proteins to acceptor organelles, but how lipids and proteins impose a tubular geometry on the carriers is incompletely understood. Using imaging approaches on cells and in vitro membrane systems, we show that phosphatidylinositol-4-phosphate (PI4P) and biogenesis of lysosome-related organelles complex 1 (BLOC-1) govern the formation, stability, and functions of recycling endosomal tubules. In vitro, BLOC-1 binds and tubulates negatively charged membranes, including those containing PI4P. In cells, endosomal PI4P production by type II PI4-kinases is needed to form and stabilize BLOC-1-dependent recycling endosomal tubules. Decreased PI4KIIs expression impairs the recycling of endosomal cargoes and the life cycles of intracellular pathogens such as Chlamydia bacteria and influenza virus that exploit the membrane dynamics of recycling endosomes. This study demonstrates how a phospholipid and a protein complex coordinate the remodeling of cellular membranes into functional tubules.
Collapse
Affiliation(s)
- Riddhi Atul Jani
- Institut Curie, Université PSL, CNRS, UMR144, Structure and Membrane Compartments, Paris, France
| | - Aurélie Di Cicco
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, Paris, France.,Institut Curie, Université PSL, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), Paris, France
| | - Tal Keren-Kaplan
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Silvia Vale-Costa
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Daniel Hamaoui
- Institut Pasteur, Université de Paris Cité, CNRS UMR3691, Cellular biology of microbial infection, Paris, France
| | - Ilse Hurbain
- Institut Curie, Université PSL, CNRS, UMR144, Structure and Membrane Compartments, Paris, France.,Institut Curie, Université PSL, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), Paris, France
| | - Feng-Ching Tsai
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, Paris, France
| | - Mathilde Di Marco
- Institut Curie, Université PSL, CNRS, UMR144, Structure and Membrane Compartments, Paris, France
| | - Anne-Sophie Macé
- Institut Curie, Université PSL, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), Paris, France
| | - Yueyao Zhu
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA.,Department of Biology, University of Pennsylvania, Philadelphia, PA
| | - Maria João Amorim
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal.,Universidade Católica Portuguesa, Católica Medical School, Católica Biomedical Research Centre, Palma de Cima, Lisboa, Portugal
| | - Patricia Bassereau
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, Paris, France
| | - Juan S Bonifacino
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD
| | - Agathe Subtil
- Institut Pasteur, Université de Paris Cité, CNRS UMR3691, Cellular biology of microbial infection, Paris, France
| | - Michael S Marks
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA.,Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Daniel Lévy
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico-Chimie Curie, Paris, France.,Institut Curie, Université PSL, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), Paris, France
| | - Graça Raposo
- Institut Curie, Université PSL, CNRS, UMR144, Structure and Membrane Compartments, Paris, France.,Institut Curie, Université PSL, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), Paris, France
| | - Cédric Delevoye
- Institut Curie, Université PSL, CNRS, UMR144, Structure and Membrane Compartments, Paris, France.,Institut Curie, Université PSL, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), Paris, France
| |
Collapse
|
40
|
Zhu Y, Li S, Jaume A, Jani RA, Delevoye C, Raposo G, Marks MS. Type II phosphatidylinositol 4-kinases function sequentially in cargo delivery from early endosomes to melanosomes. J Biophys Biochem Cytol 2022; 221:213509. [PMID: 36169639 PMCID: PMC9524207 DOI: 10.1083/jcb.202110114] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/31/2022] [Accepted: 05/04/2022] [Indexed: 12/11/2022] Open
Abstract
Melanosomes are pigment cell-specific lysosome-related organelles in which melanin pigments are synthesized and stored. Melanosome maturation requires delivery of melanogenic cargoes via tubular transport carriers that emanate from early endosomes and that require BLOC-1 for their formation. Here we show that phosphatidylinositol-4-phosphate (PtdIns4P) and the type II PtdIns-4-kinases (PI4KIIα and PI4KIIβ) support BLOC-1-dependent tubule formation to regulate melanosome biogenesis. Depletion of either PI4KIIα or PI4KIIβ with shRNAs in melanocytes reduced melanin content and misrouted BLOC-1-dependent cargoes to late endosomes/lysosomes. Genetic epistasis, cell fractionation, and quantitative live-cell imaging analyses show that PI4KIIα and PI4KIIβ function sequentially and non-redundantly downstream of BLOC-1 during tubule elongation toward melanosomes by generating local pools of PtdIns4P. The data show that both type II PtdIns-4-kinases are necessary for efficient BLOC-1-dependent tubule elongation and subsequent melanosome contact and content delivery during melanosome biogenesis. The independent functions of PtdIns-4-kinases in tubule extension are downstream of likely redundant functions in BLOC-1-dependent tubule initiation.
Collapse
Affiliation(s)
- Yueyao Zhu
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA.,Department of Biology, University of Pennsylvania School of Arts and Sciences, Philadelphia, PA
| | - Shuixing Li
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA.,Department of Pathology and Laboratory Medicine and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Alexa Jaume
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA.,Department of Pathology and Laboratory Medicine and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Riddhi Atul Jani
- Institut Curie, PSL Research University, CNRS, UMR 144, Structure and Membrane Compartments, Paris, France
| | - Cédric Delevoye
- Institut Curie, PSL Research University, CNRS, UMR 144, Structure and Membrane Compartments, Paris, France
| | - Graça Raposo
- Institut Curie, PSL Research University, CNRS, UMR 144, Structure and Membrane Compartments, Paris, France
| | - Michael S Marks
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA.,Department of Pathology and Laboratory Medicine and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
41
|
Li H, Zhang L, Zhang L, Han R. Autophagy in striated muscle diseases. Front Cardiovasc Med 2022; 9:1000067. [PMID: 36312227 PMCID: PMC9606591 DOI: 10.3389/fcvm.2022.1000067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Impaired biomolecules and cellular organelles are gradually built up during the development and aging of organisms, and this deteriorating process is expedited under stress conditions. As a major lysosome-mediated catabolic process, autophagy has evolved to eradicate these damaged cellular components and recycle nutrients to restore cellular homeostasis and fitness. The autophagic activities are altered under various disease conditions such as ischemia-reperfusion cardiac injury, sarcopenia, and genetic myopathies, which impact multiple cellular processes related to cellular growth and survival in cardiac and skeletal muscles. Thus, autophagy has been the focus for therapeutic development to treat these muscle diseases. To develop the specific and effective interventions targeting autophagy, it is essential to understand the molecular mechanisms by which autophagy is altered in heart and skeletal muscle disorders. Herein, we summarize how autophagy alterations are linked to cardiac and skeletal muscle defects and how these alterations occur. We further discuss potential pharmacological and genetic interventions to regulate autophagy activities and their applications in cardiac and skeletal muscle diseases.
Collapse
Affiliation(s)
- Haiwen Li
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Lingqiang Zhang
- State Key Laboratory of Proteomics, National Center of Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Lei Zhang
- Department of Anatomy and Neurobiology, Shanghai Yangzhi Rehabilitation Hospital, Shanghai Sunshine Rehabilitation Center, School of Medicine, Tongji University, Shanghai, China
| | - Renzhi Han
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
42
|
Vantaggiato C, Orso G, Guarato G, Brivio F, Napoli B, Panzeri E, Masotti S, Santorelli FM, Lamprou M, Gumeni S, Clementi E, Bassi MT. Rescue of lysosomal function as therapeutic strategy for SPG15 hereditary spastic paraplegia. Brain 2022; 146:1103-1120. [PMID: 36029068 PMCID: PMC9976989 DOI: 10.1093/brain/awac308] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/22/2022] [Accepted: 08/11/2022] [Indexed: 11/14/2022] Open
Abstract
SPG15 is a hereditary spastic paraplegia subtype caused by mutations in Spastizin, a protein encoded by the ZFYVE26 gene. Spastizin is involved in autophagosome maturation and autophagic lysosome reformation and SPG15-related mutations lead to autophagic lysosome reformation defects with lysosome enlargement, free lysosome depletion and autophagosome accumulation. Symptomatic and rehabilitative treatments are the only therapy currently available for patients. Here, we targeted autophagy and lysosomes in SPG15 patient-derived cells by using a library of autophagy-modulating compounds. We identified a rose of compounds affecting intracellular calcium levels, the calcium-calpain pathway or lysosomal functions, which reduced autophagosome accumulation. The six most effective compounds were tested in vivo in a new SPG15 loss of function Drosophila model that mimicked the reported SPG15 phenotype, with autophagosome accumulation, enlarged lysosomes, reduced free lysosomes, autophagic lysosome reformation defects and locomotor deficit. These compounds, namely verapamil, Bay K8644, 2',5'-dideoxyadenosine, trehalose, Small-Molecule Enhancer of Rapamycin 28 and trifluoperazine, improved lysosome biogenesis and function in vivo, demonstrating that lysosomes are a key pharmacological target to rescue SPG15 phenotype. Among the others, the Small-Molecule Enhancer of Rapamycin 28 was the most effective, rescuing both autophagic lysosome reformation defects and locomotor deficit, and could be considered as a potential therapeutic compound for this hereditary spastic paraplegia subtype.
Collapse
Affiliation(s)
- Chiara Vantaggiato
- Correspondence to: Chiara Vantaggiato, PhD Scientific Institute, IRCCS E. Medea, Laboratory of Molecular Biology, Via D. L. Monza 20 23842 Bosisio Parini, Lecco, Italy E-mail:
| | - Genny Orso
- Correspondence may also be addressed to: Genny Orso, PhD Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo E. Meneghetti 2, Padova, Italy E-mail:
| | - Giulia Guarato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo E. Meneghetti 2, Padova, Italy
| | - Francesca Brivio
- Scientific Institute IRCCS E. Medea, Laboratory of Molecular Biology, 23842 Bosisio Parini, Lecco, Italy
| | - Barbara Napoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo E. Meneghetti 2, Padova, Italy
| | - Elena Panzeri
- Scientific Institute IRCCS E. Medea, Laboratory of Molecular Biology, 23842 Bosisio Parini, Lecco, Italy
| | - Simona Masotti
- Scientific Institute IRCCS E. Medea, Laboratory of Molecular Biology, 23842 Bosisio Parini, Lecco, Italy
| | | | - Maria Lamprou
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens 15784, Greece
| | - Sentiljana Gumeni
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens 15784, Greece
| | - Emilio Clementi
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences L. Sacco, ‘Luigi Sacco’ University Hospital, Università di Milano, Milan, Italy
| | - Maria Teresa Bassi
- Scientific Institute IRCCS E. Medea, Laboratory of Molecular Biology, 23842 Bosisio Parini, Lecco, Italy
| |
Collapse
|
43
|
Rodgers SJ, Jones EI, Arumugam S, Hamila SA, Danne J, Gurung R, Eramo MJ, Nanayakkara R, Ramm G, McGrath MJ, Mitchell CA. Endosome maturation links PI3Kα signaling to lysosome repopulation during basal autophagy. EMBO J 2022; 41:e110398. [PMID: 35968799 PMCID: PMC9531306 DOI: 10.15252/embj.2021110398] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 07/19/2022] [Accepted: 07/21/2022] [Indexed: 12/24/2022] Open
Abstract
Autophagy depends on the repopulation of lysosomes to degrade intracellular components and recycle nutrients. How cells co‐ordinate lysosome repopulation during basal autophagy, which occurs constitutively under nutrient‐rich conditions, is unknown. Here, we identify an endosome‐dependent phosphoinositide pathway that links PI3Kα signaling to lysosome repopulation during basal autophagy. We show that PI3Kα‐derived PI(3)P generated by INPP4B on late endosomes was required for basal but not starvation‐induced autophagic degradation. PI(3)P signals were maintained as late endosomes matured into endolysosomes, and served as the substrate for the 5‐kinase, PIKfyve, to generate PI(3,5)P2. The SNX‐BAR protein, SNX2, was recruited to endolysosomes by PI(3,5)P2 and promoted lysosome reformation. Inhibition of INPP4B/PIKfyve‐dependent lysosome reformation reduced autophagic clearance of protein aggregates during proteotoxic stress leading to increased cytotoxicity. Therefore under nutrient‐rich conditions, PI3Kα, INPP4B, and PIKfyve sequentially contribute to basal autophagic degradation and protection from proteotoxic stress via PI(3,5)P2‐dependent lysosome reformation from endolysosomes. These findings reveal that endosome maturation couples PI3Kα signaling to lysosome reformation during basal autophagy.
Collapse
Affiliation(s)
- Samuel J Rodgers
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Emily I Jones
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Senthil Arumugam
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,European Molecular Biological Laboratory Australia, Monash University, Clayton, VIC, Australia
| | - Sabryn A Hamila
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Jill Danne
- Monash Ramaciotti Centre for Cryo Electron Microscopy, A Node of Microscopy Australia, Monash University, Clayton, VIC, Australia
| | - Rajendra Gurung
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Matthew J Eramo
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Randini Nanayakkara
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Monash Ramaciotti Centre for Cryo Electron Microscopy, A Node of Microscopy Australia, Monash University, Clayton, VIC, Australia
| | - Georg Ramm
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.,Monash Ramaciotti Centre for Cryo Electron Microscopy, A Node of Microscopy Australia, Monash University, Clayton, VIC, Australia
| | - Meagan J McGrath
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Christina A Mitchell
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
44
|
Elkenani M, Barakat AZ, Held T, Rodrigues DM, Mobarak S, Swarnka S, Adham IM, Mohamed BA. Heat shock protein A4 ablation leads to skeletal muscle myopathy associated with dysregulated autophagy and induced apoptosis. J Transl Med 2022; 20:229. [PMID: 35568953 PMCID: PMC9107738 DOI: 10.1186/s12967-022-03418-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/28/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Molecular chaperones assist protein folding, facilitate degradation of misfolded polypeptides, and thereby maintain protein homeostasis. Impaired chaperone activity leads to defective protein quality control that is implicated in multiple skeletal muscle diseases. The heat shock protein A4 (HSPA4) acts as a co-chaperone for HSP70. Previously, we showed that Hspa4 deletion causes impaired protein homeostasis in the heart. However, its functional role in skeletal muscle has not been explored. METHODS We performed a comparative phenotypic and biochemical analyses of Hspa4 knockout (KO) mice with wild-type (WT) littermates. RESULTS HSPA4 is markedly upregulated in regenerating WT muscle in vivo, and in differentiated myoblasts in vitro. Hspa4-KO mice are marked by growth retardation and increased variability in body weight, accompanied by 35% mortality rates during the peri-weaning period. The surviving Hspa4-KO mice experienced progressive skeletal muscle myopathy, characterized by increased number of muscle fibers with centralized nuclei, heterogeneous myofiber size distribution, inflammatory cell infiltrates and upregulation of embryonic and perinatal myosin heavy chain transcripts. Hspa4-KO muscles demonstrated an accumulation of autophagosome-associated proteins including microtubule associated protein1 light chain 3-II (LC3-II) and p62/sequestosome accompanied by increased number of TUNEL-positive nuclei. CONCLUSIONS Our findings underscore the indispensable role of HSPA4 in maintenance of muscle integrity through contribution in skeletal muscle autophagy and apoptosis, which might provide a novel therapeutic strategy for skeletal muscle morbidities.
Collapse
Affiliation(s)
- Manar Elkenani
- Department of Cardiology and Pneumology, Heart Center, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Amal Z Barakat
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany.,Biotechnology Research Institute, National Research Centre, Giza, Egypt
| | - Torsten Held
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Daniel Marques Rodrigues
- Department of Cardiology and Pneumology, Heart Center, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Sherok Mobarak
- Department of Cardiology and Pneumology, Heart Center, University Medical Center Göttingen, Göttingen, Germany
| | - Surabhi Swarnka
- Department of Cardiology and Pneumology, Heart Center, University Medical Center Göttingen, Göttingen, Germany
| | - Ibrahim M Adham
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Belal A Mohamed
- Department of Cardiology and Pneumology, Heart Center, University Medical Center Göttingen, Göttingen, Germany. .,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.
| |
Collapse
|
45
|
Bohnert KA, Johnson AE. Branching Off: New Insight Into Lysosomes as Tubular Organelles. Front Cell Dev Biol 2022; 10:863922. [PMID: 35646899 PMCID: PMC9130654 DOI: 10.3389/fcell.2022.863922] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/25/2022] [Indexed: 12/13/2022] Open
Abstract
Lysosomes are acidic, membrane-bound organelles that play essential roles in cellular quality control, metabolism, and signaling. The lysosomes of a cell are commonly depicted as vesicular organelles. Yet, lysosomes in fact show a high degree of ultrastructural heterogeneity. In some biological contexts, lysosome membranes naturally transform into tubular, non-vesicular morphologies. Though the purpose and regulation of tubular lysosomes has been historically understudied, emerging evidence suggests that tubular lysosomes may carry out unique activities, both degradative and non-degradative, that are critical to cell behavior, function, and viability. Here, we discuss recent advances in understanding the biological significance of tubular lysosomes in cellular physiology, and we highlight a growing number of examples that indicate the centrality of this special class of lysosomes to health and disease.
Collapse
Affiliation(s)
- K. Adam Bohnert
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, United States
| | - Alyssa E. Johnson
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
46
|
Malkeyeva D, Kiseleva E, Fedorova SA. Loss of Hsp67Bc leads to autolysosome enlargement in the Drosophila brain. Cell Biol Int 2021; 46:203-212. [PMID: 34719095 DOI: 10.1002/cbin.11721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 10/18/2021] [Accepted: 10/23/2021] [Indexed: 01/02/2023]
Abstract
Hsp67Bc is a small heat shock protein found in Drosophila melanogaster. Apart from performing a function (common for all small heat shock proteins) of preventing aggregation of misfolded proteins, it is involved in macroautophagy regulation alongside the Starvin protein. Overexpression of the D. melanogaster Hsp67Bc gene has been shown to stimulate macroautophagy in S2 cell culture. Nonetheless, it has been unknown how the absence of the Hsp67Bc gene may affect it. Here, we studied the effect of Hsp67Bc gene deletion on the macroautophagy induced by the pathogenic Wolbachia wMelPop strain in D. melanogaster. We detected Wolbachia inside autophagic vacuoles in fly neurons, thereby proving that these endosymbionts were being eliminated via macroautophagy. Nevertheless, we did not register any difference in brain bacterial load between Hsp67Bc-null and control flies at all tested stages of ontogenesis. Moreover, the abundance of autophagic vacuoles was similar between neurons of the mutant and control flies, yet the cross-sectional area of autolysosomes on ultrathin sections was more than 1.5-fold larger in Hsp67Bc-null fly brains than in the control line. Our findings suggest that the product of the Hsp67Bc gene does not participate in the initiation of endosymbiont-induced macroautophagy but may mediate autophagosome maturation: the deletion of the Hsp67Bc gene leads to the increase in autolysosome size.
Collapse
Affiliation(s)
- Dina Malkeyeva
- Cell Biology Department, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Elena Kiseleva
- Cell Biology Department, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - Svetlana A Fedorova
- Cell Biology Department, Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| |
Collapse
|
47
|
Gómez-Oca R, Cowling BS, Laporte J. Common Pathogenic Mechanisms in Centronuclear and Myotubular Myopathies and Latest Treatment Advances. Int J Mol Sci 2021; 22:11377. [PMID: 34768808 PMCID: PMC8583656 DOI: 10.3390/ijms222111377] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 10/18/2021] [Indexed: 01/18/2023] Open
Abstract
Centronuclear myopathies (CNM) are rare congenital disorders characterized by muscle weakness and structural defects including fiber hypotrophy and organelle mispositioning. The main CNM forms are caused by mutations in: the MTM1 gene encoding the phosphoinositide phosphatase myotubularin (myotubular myopathy), the DNM2 gene encoding the mechanoenzyme dynamin 2, the BIN1 gene encoding the membrane curvature sensing amphiphysin 2, and the RYR1 gene encoding the skeletal muscle calcium release channel/ryanodine receptor. MTM1, BIN1, and DNM2 proteins are involved in membrane remodeling and trafficking, while RyR1 directly regulates excitation-contraction coupling (ECC). Several CNM animal models have been generated or identified, which confirm shared pathological anomalies in T-tubule remodeling, ECC, organelle mispositioning, protein homeostasis, neuromuscular junction, and muscle regeneration. Dynamin 2 plays a crucial role in CNM physiopathology and has been validated as a common therapeutic target for three CNM forms. Indeed, the promising results in preclinical models set up the basis for ongoing clinical trials. Another two clinical trials to treat myotubular myopathy by MTM1 gene therapy or tamoxifen repurposing are also ongoing. Here, we review the contribution of the different CNM models to understanding physiopathology and therapy development with a focus on the commonly dysregulated pathways and current therapeutic targets.
Collapse
Affiliation(s)
- Raquel Gómez-Oca
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France;
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67400 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67400 Illkirch, France
- Strasbourg University, 67081 Strasbourg, France
- Dynacure, 67400 Illkirch, France;
| | | | - Jocelyn Laporte
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France;
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67400 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67400 Illkirch, France
- Strasbourg University, 67081 Strasbourg, France
| |
Collapse
|
48
|
Schechter M, Sharon R. An Emerging Role for Phosphoinositides in the Pathophysiology of Parkinson’s Disease. JOURNAL OF PARKINSON'S DISEASE 2021; 11:1725-1750. [PMID: 34151859 PMCID: PMC8609718 DOI: 10.3233/jpd-212684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Recent data support an involvement of defects in homeostasis of phosphoinositides (PIPs) in the pathophysiology of Parkinson’s disease (PD). Genetic mutations have been identified in genes encoding for PIP-regulating and PIP-interacting proteins, that are associated with familial and sporadic PD. Many of these proteins are implicated in vesicular membrane trafficking, mechanisms that were recently highlighted for their close associations with PD. PIPs are phosphorylated forms of the membrane phospholipid, phosphatidylinositol. Their composition in the vesicle’s membrane of origin, as well as membrane of destination, controls vesicular membrane trafficking. We review the converging evidence that points to the involvement of PIPs in PD. The review describes PD- and PIP-associated proteins implicated in clathrin-mediated endocytosis and autophagy, and highlights the involvement of α-synuclein in these mechanisms.
Collapse
Affiliation(s)
- Meir Schechter
- Department of Biochemistry and Molecular Biology, IMRIC, The Hebrew University-Hadassah Medical School, Ein Kerem, Jerusalem, Israel
| | - Ronit Sharon
- Department of Biochemistry and Molecular Biology, IMRIC, The Hebrew University-Hadassah Medical School, Ein Kerem, Jerusalem, Israel
| |
Collapse
|
49
|
Miyazaki M, Hiramoto M, Takano N, Kokuba H, Takemura J, Tokuhisa M, Hino H, Kazama H, Miyazawa K. Targeted disruption of GAK stagnates autophagic flux by disturbing lysosomal dynamics. Int J Mol Med 2021; 48:195. [PMID: 34468012 PMCID: PMC8416139 DOI: 10.3892/ijmm.2021.5028] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/05/2021] [Indexed: 01/13/2023] Open
Abstract
The autophagy-lysosome system allows cells to adapt to environmental changes by regulating the degradation and recycling of cellular components, and to maintain homeostasis by removing aggregated proteins and defective organelles. Cyclin G-associated kinase (GAK) is involved in the regulation of clathrin-dependent endocytosis and cell cycle progression. In addition, a single nucleotide polymorphism at the GAK locus has been reported as a risk factor for Parkinson's disease. However, the roles of GAK in the autophagy-lysosome system are not completely understood, thus the present study aimed to clarify this. In the present study, under genetic disruption or chemical inhibition of GAK, analyzing autophagic flux and observing morphological changes of autophagosomes and autolysosomes revealed that GAK controlled lysosomal dynamics via actomyosin regulation, resulting in a steady progression of autophagy. GAK knockout (KO) in A549 cells impaired autophagosome-lysosome fusion and autophagic lysosome reformation, which resulted in the accumulation of enlarged autophagosomes and autolysosomes during prolonged starvation. The stagnation of autophagic flux accompanied by these phenomena was also observed with the addition of a GAK inhibitor. Furthermore, the addition of Rho-associated protein kinase (ROCK) inhibitor or ROCK1 knockdown mitigated GAK KO-mediated effects. The results suggested a vital role of GAK in controlling lysosomal dynamics via maintaining lysosomal homeostasis during autophagy.
Collapse
Affiliation(s)
- Masaya Miyazaki
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Masaki Hiramoto
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Naoharu Takano
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Hiroko Kokuba
- Laboratory of Electron Microscopy, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Jun Takemura
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Mayumi Tokuhisa
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Hirotsugu Hino
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Hiromi Kazama
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| | - Keisuke Miyazawa
- Department of Biochemistry, Tokyo Medical University, Tokyo 160‑8402, Japan
| |
Collapse
|
50
|
Yin L, Li N, Jia W, Wang N, Liang M, Yang X, Du G. Skeletal muscle atrophy: From mechanisms to treatments. Pharmacol Res 2021; 172:105807. [PMID: 34389456 DOI: 10.1016/j.phrs.2021.105807] [Citation(s) in RCA: 190] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/03/2021] [Accepted: 08/07/2021] [Indexed: 02/07/2023]
Abstract
Skeletal muscle is a crucial tissue for movement, gestural assistance, metabolic homeostasis, and thermogenesis. It makes up approximately 40% of the total body weight and 50% of total protein. However, several pathological abnormalities (e.g., chronic diseases, cancer, long-term infection, aging) can induce an imbalance in skeletal muscle protein synthesis and degradation, which triggers muscle wasting and even leads to atrophy. Skeletal muscle atrophy is characterized by weakening, shrinking, and decreasing muscle mass and fiber cross-sectional area at the histological level. It manifests as a reduction in force production, easy fatigue and decreased exercise capability, along with a lower quality of life. Mechanistically, there are several pathophysiological processes involved in skeletal muscle atrophy, including oxidative stress and inflammation, which then activate signal transduction, such as the ubiquitin proteasome system, autophagy lysosome system, and mTOR. Considering the great economic and social burden that muscle atrophy can inflict, effective prevention and treatment strategies are essential but still limited. Exercise is widely acknowledged as the most effective therapy for skeletal muscle atrophy; unfortunately, it is not applicable for all patients. Several active substances for skeletal muscle atrophy have been discovered and evaluated in clinical trials, however, they have not been marketed to date. Knowledge is being gained on the underlying mechanisms, highlighting more promising treatment strategies in the future. In this paper, the mechanisms and treatment strategies for skeletal muscle atrophy are briefly reviewed.
Collapse
Affiliation(s)
- Lin Yin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Na Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Weihua Jia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Nuoqi Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Meidai Liang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China
| | - Xiuying Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China.
| | - Guanhua Du
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing Key Laboratory of Drug Target and Screening Research, Institute of Materia Medica of Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, PR China.
| |
Collapse
|