1
|
Hossain MK, Chae HJ. Calcium balance through mutual orchestrated inter-organelle communication: A pleiotropic target for combating Alzheimer's disease. Neurochem Int 2025; 182:105905. [PMID: 39566580 DOI: 10.1016/j.neuint.2024.105905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 11/22/2024]
Abstract
Dysfunctional intraneuronal organelles in Alzheimer's Disease (AD) propel aberrant calcium handling, triggering molecular miscommunication within organelles such as mitochondria, endoplasmic reticulum, and lysosomes. This disruption in organelle function not only impairs cellular homeostasis but also exacerbates neurodegenerative processes involving the accumulation of amyloid-β (Aβ) and hyperphosphorylated tau, amplifying the disease's vicious cycle. In this review, the concept of Mutual Orchestrated Inter-organelle Communication (MOIC) proposes potential therapeutic avenues for restoring Ca2+ homeostasis in AD, offering a theoretical framework for developing disease-modifying treatments. The intricate nature of AD necessitates a shift towards combination therapies targeting MOIC-associated pathways, presenting a more effective approach than monotherapy.
Collapse
Affiliation(s)
| | - Han Jung Chae
- Non-Clinical Evaluation Center, Biomedical Research Institute, Jeonbuk National University Hospital, Jeonju, Republic of Korea.
| |
Collapse
|
2
|
Tedeschi V, Sapienza S, Ciancio R, Canzoniero LMT, Pannaccione A, Secondo A. Lysosomal Channels as New Molecular Targets in the Pharmacological Therapy of Neurodegenerative Diseases via Autophagy Regulation. Curr Neuropharmacol 2025; 23:375-383. [PMID: 38766825 DOI: 10.2174/1570159x22666240517101846] [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: 11/20/2023] [Revised: 12/20/2023] [Accepted: 01/16/2024] [Indexed: 05/22/2024] Open
Abstract
Besides controlling several organellar functions, lysosomal channels also guide the catabolic "self-eating" process named autophagy, which is mainly involved in protein and organelle quality control. Neuronal cells are particularly sensitive to the rate of autophagic flux either under physiological conditions or during the degenerative process. Accordingly, neurodegeneration occurring in Parkinson's (PD), Alzheimer's (AD), and Huntington's Diseases (HD), and Amyotrophic Lateral Sclerosis (ALS) as well as Lysosomal Storage Diseases (LSD) is partially due to defective autophagy and accumulation of toxic aggregates. In this regard, dysfunction of lysosomal ionic homeostasis has been identified as a putative cause of aberrant autophagy. From a therapeutic perspective, Transient Receptor Potential Channel Mucolipin 1 (TRPML1) and Two-Pore Channel isoform 2 (TPC2), regulating lysosomal homeostasis, are now considered promising druggable targets in neurodegenerative diseases. Compelling evidence suggests that pharmacological modulation of TRPML1 and TPC2 may rescue the pathological phenotype associated with autophagy dysfunction in AD, PD, HD, ALS, and LSD. Although pharmacological repurposing has identified several already used drugs with the ability to modulate TPC2, and several tools are already available for the modulation of TRPML1, many efforts are necessary to design and test new entities with much higher specificity in order to reduce dysfunctional autophagy during neurodegeneration.
Collapse
Affiliation(s)
- Valentina Tedeschi
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, Federico II University of Naples, Via S. Pansini 5, 80131 Naples, Italy
| | - Silvia Sapienza
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, Federico II University of Naples, Via S. Pansini 5, 80131 Naples, Italy
| | - Raffaella Ciancio
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, Federico II University of Naples, Via S. Pansini 5, 80131 Naples, Italy
| | | | - Anna Pannaccione
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, Federico II University of Naples, Via S. Pansini 5, 80131 Naples, Italy
| | - Agnese Secondo
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, Federico II University of Naples, Via S. Pansini 5, 80131 Naples, Italy
| |
Collapse
|
3
|
Tedeschi V, Nele V, Valsecchi V, Anzilotti S, Vinciguerra A, Zucaro L, Sisalli MJ, Cassiano C, De Iesu N, Pignataro G, Canzoniero LMT, Pannaccione A, De Rosa G, Secondo A. Nanoparticles encapsulating phosphatidylinositol derivatives promote neuroprotection and functional improvement in preclinical models of ALS via a long-lasting activation of TRPML1 lysosomal channel. Pharmacol Res 2024; 210:107491. [PMID: 39491634 DOI: 10.1016/j.phrs.2024.107491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease currently incurable, in which motor neuron degeneration leads to voluntary skeletal muscle atrophy. Molecularly, ALS is characterized by protein aggregation, synaptic and organellar dysfunction, and Ca2+ dyshomeostasis. Of interest, autophagy dysfunction is emerging as one of the main putative targets of ALS therapy. A tune regulation of this cleansing process is affordable by a proper stimulation of TRPML1, one of the main lysosomal channels. However, TRPML1 activation by PI(3,5)P2 has low open probability to remain in an active conformation. To overcome this drawback we developed a lipid-based formulation of PI(3,5)P2 whose putative therapeutic potential has been tested in in vitro and in vivo ALS models. Pharmacodynamic properties of PI(3,5)P2 lipid-based formulations (F1 and F2) on TRPML1 activity have been characterized by means of patch-clamp electrophysiology and Fura-2AM video-imaging in motor neuronal cells. Once selected for the ability to stabilize TRPML1 activity, the most effective preparation F1 was studied in vivo to measure neuromuscular function and survival of SOD1G93A ALS mice, thereby establishing its therapeutic profile. F1, but not PI(3,5)P2 alone, stabilized the open state of the lysosomal channel TRPML1 and increased the persistence of intracellular calcium concentration ([Ca2+]i). Then, F1 was effective in delaying motor neuron loss, improving innervated endplants and muscle performance in SOD1G93A mice, extending overall lifespan by an average of 10 days. Of note F1 prevented gliosis and autophagy dysfunction in ALS mice by restoring PI(3,5)P2 level. Our novel self-assembling lipidic formulation for PI(3,5)P2 delivery exerts a neuroprotective effect in preclinical models of ALS mainly regulating dysfunctional autophagy through TRPML1 activity stabilization.
Collapse
Affiliation(s)
- Valentina Tedeschi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, University of Naples "Federico II", Via S. Pansini 5, Naples 80131, Italy
| | - Valeria Nele
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, Naples 80131, Italy
| | - Valeria Valsecchi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, University of Naples "Federico II", Via S. Pansini 5, Naples 80131, Italy
| | - Serenella Anzilotti
- Department of Science and Technology-DST, University of Sannio, Via Port'Arsa 11, Benevento 82100, Italy
| | - Antonio Vinciguerra
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica Delle Marche", Via Tronto 10/A, Ancona 60126, Italy
| | - Laura Zucaro
- Biogem Scarl, Istituto di Ricerche Genetiche, Ariano Irpino, AV, Italy; Department of Translational Medical Sciences, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Josè Sisalli
- Department of Translational Medical Sciences, University of Naples "Federico II", Via S. Pansini 5, Naples 80131, Italy
| | - Chiara Cassiano
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, Naples 80131, Italy
| | | | - Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, University of Naples "Federico II", Via S. Pansini 5, Naples 80131, Italy
| | | | - Anna Pannaccione
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, University of Naples "Federico II", Via S. Pansini 5, Naples 80131, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, Naples 80131, Italy.
| | - Agnese Secondo
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica Delle Marche", Via Tronto 10/A, Ancona 60126, Italy.
| |
Collapse
|
4
|
Kumar A, Zhao Y, Xie L, Chadda R, Abraham N, Hong J, Feng E, Tranter JD, Rawnsley D, Liu H, Henry KM, Meyer G, Hu M, Xu H, Hinton A, Grueter CE, Abel ED, Norris AW, Diwan A, Sah R. Lysosomal LRRC8 complex regulates lysosomal pH, morphology and systemic glucose metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614256. [PMID: 39386592 PMCID: PMC11463514 DOI: 10.1101/2024.09.22.614256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The lysosome integrates anabolic signalling and nutrient-sensing to regulate intracellular growth pathways. The leucine-rich repeat containing 8 (LRRC8) channel complex forms a lysosomal anion channel and regulates PI3K-AKT-mTOR signalling, skeletal muscle differentiation, growth, and systemic glucose metabolism. Here, we define the endogenous LRRC8 subunits localized to a subset of lysosomes in differentiated myotubes. We show LRRC8A regulates leucine-stimulated mTOR, lysosome size, number, pH, and expression of lysosomal proteins LAMP2, P62, LC3B, suggesting impaired autophagic flux. Mutating a LRRC8A lysosomal targeting dileucine motif sequence (LRRC8A-L706A;L707A) in myotubes recapitulates the abnormal AKT signalling and altered lysosomal morphology and pH observed in LRRC8A KO cells. In vivo , LRRC8A-L706A;L707A KI mice exhibit increased adiposity, impaired glucose tolerance and insulin resistance characterized by reduced skeletal muscle glucose-uptake, and impaired incorporation of glucose into glycogen. These data reveal a lysosomal LRRC8 mediated metabolic signalling function that regulates lysosomal activity, systemic glucose homeostasis and insulin-sensitivity.
Collapse
|
5
|
Hou M, Zhang Z, Fan Z, Huang L, Wang L. The mechanisms of Ca2+ regulating autophagy and its research progress in neurodegenerative diseases: A review. Medicine (Baltimore) 2024; 103:e39405. [PMID: 39183424 PMCID: PMC11346841 DOI: 10.1097/md.0000000000039405] [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: 01/23/2024] [Revised: 07/23/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024] Open
Abstract
Neurodegenerative diseases are complex disorders that significantly challenge human health, with their incidence increasing with age. A key pathological feature of these diseases is the accumulation of misfolded proteins. The underlying mechanisms involve an imbalance in calcium homeostasis and disturbances in autophagy, indicating a likely correlation between them. As the most important second messenger, Ca2+ plays a vital role in regulating various cell activities, including autophagy. Different organelles within cells serve as Ca2+ storage chambers and regulate Ca2+ levels under different conditions. Ca2+ in these compartments can affect autophagy via Ca2+ channels or other related signaling proteins. Researchers propose that Ca2+ regulates autophagy through distinct signal transduction mechanisms, under normal or stressful conditions, and thereby contributing to the occurrence and development of neurodegenerative diseases. This review provides a systematic examination of the regulatory mechanisms of Ca2+ in cell membranes and different organelles, as well as its downstream pathways that influence autophagy and its implications for neurodegenerative diseases. This comprehensive analysis may facilitate the development of new drugs and provide more precise treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Meng Hou
- Department of Neurology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhixiao Zhang
- Department of Neurology, Shanxi Provincial People’s Hospital, Taiyuan, Shanxi, China
| | - Zexin Fan
- Department of Neurology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lei Huang
- Department of Cardiology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Li Wang
- Department of Neurology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
6
|
Chen Y, Xie B, Hu Y, Sun J, Xu J, Shen Y, Zhu Y, Song X. Transient receptor potential mucolipin 1 circumvents oxidative stress in primary human melanocytes. Skin Res Technol 2024; 30:e13772. [PMID: 38899729 PMCID: PMC11187810 DOI: 10.1111/srt.13772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Transient Receptor Potential Mucolipin 1 (TRPML1) serves as a pivotal reactive oxygen species (ROS) sensor in cells, which is implicated in the regulation of autophagy. However, its function in melanocyte autophagy under oxidative stress remains elusive. METHODS The expression and ion channel function of TRPML1 were investigated using immunofluorescence and calcium imaging in primary human melanocytes (MCs). After activating TRPML1 with MLSA1 (TRPML1 agonist), autophagy-related molecules were investigated via western blot. ROS level, apoptosis- and autophagy-related molecules were investigated after pretreatment with MLSA1. After interference with TRPML1 expression, mitochondrial structures were visualized by electron microscopy with hydrogen peroxide (H2O2)treatment. RESULTS TRPML1 was expressed and functionally active in primary human MCs, and its activation promotes elevated expression of LC3-II and reduced apoptosis and ROS levels under oxidative stress. TRPML1 downregulation caused mitochondrial swelling and disruption of cristae structures under oxidative stress in primary human MCs. CONCLUSIONS TRPML1 might mediate lysosomal autophagy in primary human MCs under oxidative stress, participating in mechanisms that maintain the oxidative and antioxidant systems in balance.
Collapse
Affiliation(s)
- Yi Chen
- Department of DermatologyHangzhou Third Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouPeople's Republic of China
| | - Bo Xie
- Department of DermatologyHangzhou Third People's HospitalHangzhouPeople's Republic of China
| | - Yebei Hu
- Department of DermatologyHangzhou Third People's HospitalHangzhouPeople's Republic of China
| | - Jiayi Sun
- Department of DermatologyHangzhou Third Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouPeople's Republic of China
| | - Jinhui Xu
- Department of DermatologyHangzhou Third People's HospitalHangzhouPeople's Republic of China
| | - Yuqing Shen
- Department of DermatologyHangzhou Third Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouPeople's Republic of China
| | - Yuqi Zhu
- Department of DermatologyHangzhou Third Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhouPeople's Republic of China
| | - Xiuzu Song
- Department of DermatologyHangzhou Third People's HospitalHangzhouPeople's Republic of China
| |
Collapse
|
7
|
Rezzani R, Favero G, Gianò M, Pinto D, Labanca M, van Noorden CJ, Rinaldi F. Transient Receptor Potential Channels in the Healthy and Diseased Blood-Brain Barrier. J Histochem Cytochem 2024; 72:199-231. [PMID: 38590114 PMCID: PMC11020746 DOI: 10.1369/00221554241246032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 03/18/2024] [Indexed: 04/10/2024] Open
Abstract
The large family of transient receptor potential (TRP) channels are integral membrane proteins that function as environmental sensors and act as ion channels after activation by mechanical (touch), physical (heat, pain), and chemical stimuli (pungent compounds such as capsaicin). Most TRP channels are localized in the plasma membrane of cells but some of them are localized in membranes of organelles and function as intracellular Ca2+-ion channels. TRP channels are involved in neurological disorders but their precise role(s) and relevance in these disorders are not clear. Endothelial cells of the blood-brain barrier (BBB) express TRP channels such as TRP vanilloid 1-4 and are involved in thermal detection by regulating BBB permeability. In neurological disorders, TRP channels in the BBB are responsible for edema formation in the brain. Therefore, drug design to modulate locally activity of TRP channels in the BBB is a hot topic. Today, the application of TRP channel antagonists against neurological disorders is still limited.
Collapse
Affiliation(s)
- Rita Rezzani
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdipartimental University Center of Research Adaption and Regeneration of Tissues and Organs - ARTO, University of Brescia, Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale - SISDO), Brescia, Italy
| | - Gaia Favero
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Interdipartimental University Center of Research Adaption and Regeneration of Tissues and Organs - ARTO, University of Brescia, Brescia, Italy
| | - Marzia Gianò
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniela Pinto
- Human Microbiome Advanced Project Institute, Milan, Italy
| | - Mauro Labanca
- Division of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale - SISDO), Brescia, Italy
| | - Cornelis J.F. van Noorden
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Fabio Rinaldi
- Human Microbiome Advanced Project Institute, Milan, Italy
| |
Collapse
|
8
|
Nguyen HT, Wiederkehr A, Wollheim CB, Park KS. Regulation of autophagy by perilysosomal calcium: a new player in β-cell lipotoxicity. Exp Mol Med 2024; 56:273-288. [PMID: 38297165 PMCID: PMC10907728 DOI: 10.1038/s12276-024-01161-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: 05/03/2023] [Revised: 10/16/2023] [Accepted: 11/09/2023] [Indexed: 02/02/2024] Open
Abstract
Autophagy is an essential quality control mechanism for maintaining organellar functions in eukaryotic cells. Defective autophagy in pancreatic beta cells has been shown to be involved in the progression of diabetes through impaired insulin secretion under glucolipotoxic stress. The underlying mechanism reveals the pathologic role of the hyperactivation of mechanistic target of rapamycin (mTOR), which inhibits lysosomal biogenesis and autophagic processes. Moreover, accumulating evidence suggests that oxidative stress induces Ca2+ depletion in the endoplasmic reticulum (ER) and cytosolic Ca2+ overload, which may contribute to mTOR activation in perilysosomal microdomains, leading to autophagic defects and β-cell failure due to lipotoxicity. This review delineates the antagonistic regulation of autophagic flux by mTOR and AMP-dependent protein kinase (AMPK) at the lysosomal membrane, and both of these molecules could be activated by perilysosomal calcium signaling. However, aberrant and persistent Ca2+ elevation upon lipotoxic stress increases mTOR activity and suppresses autophagy. Therefore, normalization of autophagy is an attractive therapeutic strategy for patients with β-cell failure and diabetes.
Collapse
Affiliation(s)
- Ha Thu Nguyen
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Korea
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea
| | | | - Claes B Wollheim
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland.
- Department of Clinical Sciences, Lund University, Malmö, Sweden.
| | - Kyu-Sang Park
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Korea.
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Korea.
| |
Collapse
|
9
|
Wahl-Schott C, Freichel M, Hennis K, Philippaert K, Ottenheijm R, Tsvilovskyy V, Varbanov H. Characterization of Endo-Lysosomal Cation Channels Using Calcium Imaging. Handb Exp Pharmacol 2023; 278:277-304. [PMID: 36894791 DOI: 10.1007/164_2023_637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Endo-lysosomes are membrane-bound acidic organelles that are involved in endocytosis, recycling, and degradation of extracellular and intracellular material. The membranes of endo-lysosomes express several Ca2+-permeable cation ion channels, including two-pore channels (TPC1-3) and transient receptor potential mucolipin channels (TRPML1-3). In this chapter, we will describe four different state-of-the-art Ca2+ imaging approaches, which are well-suited to investigate the function of endo-lysosomal cation channels. These techniques include (1) global cytosolic Ca2+ measurements, (2) peri-endo-lysosomal Ca2+ imaging using genetically encoded Ca2+ sensors that are directed to the cytosolic endo-lysosomal membrane surface, (3) Ca2+ imaging of endo-lysosomal cation channels, which are engineered in order to redirect them to the plasma membrane in combination with approaches 1 and 2, and (4) Ca2+ imaging by directing Ca2+ indicators to the endo-lysosomal lumen. Moreover, we will review useful small molecules, which can be used as valuable tools for endo-lysosomal Ca2+ imaging. Rather than providing complete protocols, we will discuss specific methodological issues related to endo-lysosomal Ca2+ imaging.
Collapse
Affiliation(s)
- Christian Wahl-Schott
- Institut für Kardiovaskuläre Physiologie und Pathophysiologie, Lehrstuhl für Vegetative Physiologie, Biomedizinisches Zentrum, Ludwig-Maximilians-Universität München, München, Germany.
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany. .,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany.
| | - Konstantin Hennis
- Institut für Kardiovaskuläre Physiologie und Pathophysiologie, Lehrstuhl für Vegetative Physiologie, Biomedizinisches Zentrum, Ludwig-Maximilians-Universität München, München, Germany
| | - Koenraad Philippaert
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Roger Ottenheijm
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Volodymyr Tsvilovskyy
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Hristo Varbanov
- Institut für Neurophysiologie, Medizinische Hochschule Hannover(MHH), Hannover, Germany
| |
Collapse
|
10
|
Abstract
Ca2+ is a universal second messenger that plays a wide variety of fundamental roles in cellular physiology. Thus, to warrant selective responses and to allow rapid mobilization upon specific stimuli, Ca2+ is accumulated in organelles to keep it at very low levels in the cytoplasm during resting conditions. Major Ca2+ storage organelles include the endoplasmic reticulum (ER), the mitochondria, and as recently demonstrated, the lysosome (Xu and Ren, Annu Rev Physiol 77:57-80, 2015). The importance of Ca2+ signaling deregulation in human physiology is underscored by its involvement in several human diseases, including lysosomal storage disorders, neurodegenerative disease and cancer (Shen et al., Nat Commun 3:731, 2012; Bae et al., J Neurosci 34:11485-11503, 2014). Recent evidence strongly suggests that lysosomal Ca2+ plays a major role in the regulation of lysosomal adaptation to nutrient availability through a lysosomal signaling pathway involving the lysosomal Ca2+ channel TRPML1 and the transcription factor TFEB, a master regulator for lysosomal function and autophagy (Sardiello et al., Science 325:473-477, 2009; Settembre et al., Science 332:1429-1433, 2011; Medina et al., Nat Cell Biol 17:288-299, 2015; Di Paola et al., Cell Calcium 69:112-121, 2018). Due to the tight relationship of this lysosomal Ca2+ channel and TFEB, in this chapter, we will focus on the role of the TRPML1/TFEB pathway in the regulation of lysosomal function and autophagy.
Collapse
Affiliation(s)
- Diego Luis Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy.
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy.
| |
Collapse
|
11
|
Li M, An H, Wang W, Wei D. Biomolecular Markers of Brain Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1419:111-126. [PMID: 37418210 DOI: 10.1007/978-981-99-1627-6_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Characterized by the gradual loss of physiological integrity, impaired function, and increased susceptibility to death, aging is considered the primary risk factor for major human diseases, such as cancer, diabetes, cardiovascular disorders, and neurodegenerative diseases. The time-dependent accumulation of cellular damage is widely considered the general cause of aging. While the mechanism of normal aging is still unresolved, researchers have identified different markers of aging, including genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient-sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication. Theories of aging can be divided into two categories: (1) aging is a genetically programmed process, and (2) aging is a random process caused by gradual damage to the organism over time as a result of its vital activities. Aging affects the entire human body, and aging of the brain is undoubtedly different from all other organs, as neurons are highly differentiated postmitotic cells, and the lifespan of most neurons in the postnatal period is equal to the lifespan of the brain. In this chapter, we discuss the conserved mechanisms of aging that may underlie the changes observed in the aging brain, with a focus on mitochondrial function and oxidative stress, autophagy and protein turnover, insulin/IGF signaling, target of rapamycin (TOR) signaling, and sirtuin function.
Collapse
Affiliation(s)
- Min Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Haiting An
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
- Beijing Neurosurgical Institute, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
| | - Wenxiao Wang
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
- School of Systems Science, Beijing Normal University, Beijing, China
| | - Dongfeng Wei
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
12
|
Scotto Rosato A, Krogsaeter EK, Jaślan D, Abrahamian C, Montefusco S, Soldati C, Spix B, Pizzo MT, Grieco G, Böck J, Wyatt A, Wünkhaus D, Passon M, Stieglitz M, Keller M, Hermey G, Markmann S, Gruber-Schoffnegger D, Cotman S, Johannes L, Crusius D, Boehm U, Wahl-Schott C, Biel M, Bracher F, De Leonibus E, Polishchuk E, Medina DL, Paquet D, Grimm C. TPC2 rescues lysosomal storage in mucolipidosis type IV, Niemann-Pick type C1, and Batten disease. EMBO Mol Med 2022; 14:e15377. [PMID: 35929194 PMCID: PMC9449600 DOI: 10.15252/emmm.202115377] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 01/05/2023] Open
Abstract
Lysosomes are cell organelles that degrade macromolecules to recycle their components. If lysosomal degradative function is impaired, e.g., due to mutations in lysosomal enzymes or membrane proteins, lysosomal storage diseases (LSDs) can develop. LSDs manifest often with neurodegenerative symptoms, typically starting in early childhood, and going along with a strongly reduced life expectancy and quality of life. We show here that small molecule activation of the Ca2+‐permeable endolysosomal two‐pore channel 2 (TPC2) results in an amelioration of cellular phenotypes associated with LSDs such as cholesterol or lipofuscin accumulation, or the formation of abnormal vacuoles seen by electron microscopy. Rescue effects by TPC2 activation, which promotes lysosomal exocytosis and autophagy, were assessed in mucolipidosis type IV (MLIV), Niemann–Pick type C1, and Batten disease patient fibroblasts, and in neurons derived from newly generated isogenic human iPSC models for MLIV and Batten disease. For in vivo proof of concept, we tested TPC2 activation in the MLIV mouse model. In sum, our data suggest that TPC2 is a promising target for the treatment of different types of LSDs, both in vitro and in‐vivo.
Collapse
Affiliation(s)
- Anna Scotto Rosato
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Einar K Krogsaeter
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Dawid Jaślan
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Carla Abrahamian
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | | | - Chiara Soldati
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | - Barbara Spix
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | | | | | - Julia Böck
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Amanda Wyatt
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | | | - Marcel Passon
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Marc Stieglitz
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität, Munich, Germany
| | - Marco Keller
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität, Munich, Germany
| | - Guido Hermey
- Center for Molecular Neurobiology Hamburg (ZMNH), Institute of Molecular and Cellular Cognition, UKE, Hamburg, Germany
| | | | | | - Susan Cotman
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ludger Johannes
- Cellular and Chemical Biology Department, Institut Curie, U1143 INSERM, UMR3666 CNRS, PSL Research University, Paris, France
| | - Dennis Crusius
- Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-University (LMU) Hospital, Munich, Germany
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | | | - Martin Biel
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität, Munich, Germany
| | - Franz Bracher
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität, Munich, Germany
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine, Naples, Italy.,Institute of Biochemistry and Cell Biology (IBBC), CNR, Rome, Italy
| | | | - Diego L Medina
- Telethon Institute of Genetics and Medicine, Naples, Italy.,Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy
| | - Dominik Paquet
- Institute for Stroke and Dementia Research (ISD), Ludwig-Maximilians-University (LMU) Hospital, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Christian Grimm
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| |
Collapse
|
13
|
Pollmanns MR, Beer J, Rosignol I, Rodriguez-Muela N, Falkenburger BH, Dinter E. Activated Endolysosomal Cation Channel TRPML1 Facilitates Maturation of α-Synuclein-Containing Autophagosomes. Front Cell Neurosci 2022; 16:861202. [PMID: 35875350 PMCID: PMC9296810 DOI: 10.3389/fncel.2022.861202] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/08/2022] [Indexed: 11/21/2022] Open
Abstract
Background: Protein aggregates are degraded via the autophagy-lysosome pathway and alterations in the lysosomal system leading to the accumulation of pathogenic proteins, including aggregates of α-synuclein in Parkinson’s disease (PD). The importance of the endolysosomal transient receptor potential cation channel, mucolipin subfamily 1 (TRPML1) for the lysosomal function is highlighted by the fact that TRPML1 mutations cause the lysosomal storage disease mucolipidosis type IV. In this study, we investigated the mechanism by which activation of TRPML1 affects the degradation of α-synuclein. Methods: As a model of α-synuclein pathology, we expressed the pathogenic A53Tα-synuclein mutant in HEK293T cells. These cells were treated with the synthetic TRPML1 agonist ML-SA1. The amount of α-synuclein protein was determined by immunoblots. The abundance of aggregates and autolysosomal vesicles was determined by fluorescence microscopy and immunocytochemistry. Findings were confirmed by life-cell imaging and by application of ML-SA1 and the TRPML1 antagonist ML-SI3 to human dopaminergic neurons and human stem cell-derived neurons. Results: ML-SA1 reduced the percentage of HEK293T cells with α-synuclein aggregates and the amount of α-synuclein protein. The effect of ML-SA1 was blocked by pharmacological and genetic inhibition of autophagy. Consistent with TRPML function, it required the membrane lipid PI(3,5)P2, and cytosolic calcium. ML-SA1 shifted the composition of autophagosomes towards a higher fraction of mature autolysosomes, also in presence of α-synuclein. In neurons, inhibition of TRPML1 by its antagonist ML-SI3 blocked autophagosomal clearance, whereas the agonist ML-SA1 shifted the composition of a-synuclein particles towards a higher fraction of acidified particles. ML-SA1 was able to override the effect of Bafilomycin A1, which blocks the fusion of the autophagosome and lysosome and its acidification. Conclusion: These findings suggest, that activating TRPML1 with ML-SA1 facilitates clearance of α-synuclein aggregates primarily by affecting the late steps of the autophagy, i.e., by promoting autophagosome maturation. In agreement with recent work by others, our findings indicate that TRPML1 might constitute a plausible therapeutic target for PD, that warrants further validation in rodent models of α-synuclein pathology.
Collapse
Affiliation(s)
| | - Judith Beer
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ines Rosignol
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
| | - Natalia Rodriguez-Muela
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
| | - Björn H. Falkenburger
- Department of Neurology, RWTH University Aachen, Aachen, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Dresden, Germany
- JARA-Institute Molecular Neuroscience and Neuroimaging, Forschungsszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
- *Correspondence: Björn H. Falkenburger
| | - Elisabeth Dinter
- Department of Neurology, RWTH University Aachen, Aachen, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Dresden, Germany
| |
Collapse
|
14
|
Hu S, Zou Y, Jiang Y, Zhang Q, Cheng H, Wang H, Li X. Scutellarin‐mediated autophagy activates exosome release of rat nucleus pulposus cells by positively regulating Rab8a via the PI3K/PTEN/Akt pathway. Cell Biol Int 2022; 46:1588-1603. [PMID: 35762224 DOI: 10.1002/cbin.11838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/13/2022] [Accepted: 05/23/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Shun‐Qi Hu
- Department of Orthopaedic Surgery, Zhongshan Hospital Fudan University Shanghai China
| | - Yan‐Pei Zou
- Department of Orthopaedic Surgery, Zhongshan Hospital Fudan University Shanghai China
| | - Yun‐Qi Jiang
- Department of Orthopaedic Surgery, Zhongshan Hospital Fudan University Shanghai China
| | - Qi‐Chen Zhang
- Department of Orthopaedic Surgery, Zhongshan Hospital Fudan University Shanghai China
| | - Hong‐Xia Cheng
- Liver Cancer Institute, Zhongshan Hospital Fudan University Shanghai China
| | - Hui‐Ren Wang
- Department of Orthopaedic Surgery, Zhongshan Hospital Fudan University Shanghai China
| | - Xi‐Lei Li
- Department of Orthopaedic Surgery, Zhongshan Hospital Fudan University Shanghai China
| |
Collapse
|
15
|
Vrijsen S, Vrancx C, Del Vecchio M, Swinnen JV, Agostinis P, Winderickx J, Vangheluwe P, Annaert W. Inter-organellar Communication in Parkinson's and Alzheimer's Disease: Looking Beyond Endoplasmic Reticulum-Mitochondria Contact Sites. Front Neurosci 2022; 16:900338. [PMID: 35801175 PMCID: PMC9253489 DOI: 10.3389/fnins.2022.900338] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 05/05/2022] [Indexed: 01/13/2023] Open
Abstract
Neurodegenerative diseases (NDs) are generally considered proteinopathies but whereas this may initiate disease in familial cases, onset in sporadic diseases may originate from a gradually disrupted organellar homeostasis. Herein, endolysosomal abnormalities, mitochondrial dysfunction, endoplasmic reticulum (ER) stress, and altered lipid metabolism are commonly observed in early preclinical stages of major NDs, including Parkinson's disease (PD) and Alzheimer's disease (AD). Among the multitude of underlying defective molecular mechanisms that have been suggested in the past decades, dysregulation of inter-organellar communication through the so-called membrane contact sites (MCSs) is becoming increasingly apparent. Although MCSs exist between almost every other type of subcellular organelle, to date, most focus has been put on defective communication between the ER and mitochondria in NDs, given these compartments are critical in neuronal survival. Contributions of other MCSs, notably those with endolysosomes and lipid droplets are emerging, supported as well by genetic studies, identifying genes functionally involved in lysosomal homeostasis. In this review, we summarize the molecular identity of the organelle interactome in yeast and mammalian cells, and critically evaluate the evidence supporting the contribution of disturbed MCSs to the general disrupted inter-organellar homeostasis in NDs, taking PD and AD as major examples.
Collapse
Affiliation(s)
- Stephanie Vrijsen
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, KU Leuven, Leuven, Belgium
| | - Céline Vrancx
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Mara Del Vecchio
- Laboratory of Functional Biology, Department of Biology, KU Leuven, Heverlee, Belgium
| | - Johannes V. Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Laboratory of Cell Death Research and Therapy, VIB-Center for Cancer Research, KU Leuven, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Joris Winderickx
- Laboratory of Functional Biology, Department of Biology, KU Leuven, Heverlee, Belgium
| | - Peter Vangheluwe
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, KU Leuven, Leuven, Belgium
| | - Wim Annaert
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, KU Leuven, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
16
|
Allan CY, Fisher PR. The Dictyostelium Model for Mucolipidosis Type IV. Front Cell Dev Biol 2022; 10:741967. [PMID: 35493081 PMCID: PMC9043695 DOI: 10.3389/fcell.2022.741967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 03/21/2022] [Indexed: 12/02/2022] Open
Abstract
Mucolipidosis type IV, a devastating neurological lysosomal disease linked to mutations in the transient receptor potential channel mucolipin 1, TRPML1, a calcium permeable channel in the membranes of vesicles in endolysosomal system. TRPML1 function is still being elucidated and a better understanding of the molecular pathogenesis of Mucolipidosis type IV, may facilitate development of potential treatments. We have created a model to study mucolipin function in the eukaryotic slime mould Dictyostelium discoideum by altering expression of its single mucolipin homologue, mcln. We show that in Dictyostelium mucolipin overexpression contributes significantly to global chemotactic calcium responses in vegetative and differentiated cells. Knockdown of mucolipin also enhances calcium responses in vegetative cells but does not affect responses in 6–7 h developed cells, suggesting that in developed cells mucolipin may help regulate local calcium signals rather than global calcium waves. We found that both knocking down and overexpressing mucolipin often, but not always, presented the same phenotypes. Altering mucolipin expression levels caused an accumulation or increased acidification of Lysosensor Blue stained vesicles in vegetative cells. Nutrient uptake by phagocytosis and macropinocytosis were increased but growth rates were not, suggesting defects in catabolism. Both increasing and decreasing mucolipin expression caused the formation of smaller slugs and larger numbers of fruiting bodies during multicellular development, suggesting that mucolipin is involved in initiation of aggregation centers. The fruiting bodies that formed from these smaller aggregates had proportionately larger basal discs and thickened stalks, consistent with a regulatory role for mucolipin-dependent Ca2+ signalling in the autophagic cell death pathways involved in stalk and basal disk differentiation in Dictyostelium. Thus, we have provided evidence that mucolipin contributes to chemotactic calcium signalling and that Dictyostelium is a useful model to study the molecular mechanisms involved in the cytopathogenesis of Mucolipidosis type IV.
Collapse
|
17
|
Siow WX, Kabiri Y, Tang R, Chao YK, Plesch E, Eberhagen C, Flenkenthaler F, Fröhlich T, Bracher F, Grimm C, Biel M, Zischka H, Vollmar AM, Bartel K. Lysosomal TRPML1 regulates mitochondrial function in hepatocellular carcinoma cells. J Cell Sci 2022; 135:274242. [PMID: 35274126 PMCID: PMC8977057 DOI: 10.1242/jcs.259455] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/27/2022] [Indexed: 11/20/2022] Open
Abstract
Liver cancers, including hepatocellular carcinoma (HCC), are the second most lethal cancers worldwide and novel therapeutic strategies are still highly needed. Recently, the endolysosomal cation channel TRPML1 has gained focus in cancer research representing an interesting novel target. We utilized the recently developed isoform-selective TRPML1 activator ML1-SA1 and the CRISPR/Cas9 system to generate tools for over-activation and loss-of-function studies on TRPML1 in HCC. After verification of our tools, we investigated the role of TRPML1 in HCC by studying proliferation, apoptosis, and proteomic alterations. Further, we analyzed mitochondrial function in detail, facilitating confocal and transmission electron microscopy, combined with SeahorseTM and Oroboros® functional analysis. We report that TRPML1 over-activation by a novel, isoform-selective, low-molecular activator induces apoptosis by impairing mitochondrial function calcium dependently. Additionally, TRPML1 loss-of-function deregulates mitochondrial renewal, which leads to proliferation impairment. Thus, our study reveals a novel role for TRPML1 as regulator of mitochondrial function and its modulators as promising molecules for novel therapeutic options in HCC therapy.
Collapse
Affiliation(s)
- Wei Xiong Siow
- Department of Pharmacy, Center for Drug Research, Pharmaceutical Biology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Yaschar Kabiri
- Technical University Munich, School of Medicine, Institute of Toxicology and Environmental Hygiene, Biedersteiner Strasse 29, D-80802 Munich, Germany
| | - Rachel Tang
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Yu-Kai Chao
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Eva Plesch
- Department of Pharmacy, Center for Drug Research, Pharmaceutical Chemistry, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Carola Eberhagen
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, D-85764 Neuherberg, Germany
| | - Florian Flenkenthaler
- Gene Center, Laboratory for Functional Genome Analysis, Ludwig Maximilians-University Munich, Munich, Germany
| | - Thomas Fröhlich
- Gene Center, Laboratory for Functional Genome Analysis, Ludwig Maximilians-University Munich, Munich, Germany
| | - Franz Bracher
- Department of Pharmacy, Center for Drug Research, Pharmaceutical Chemistry, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christian Grimm
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Martin Biel
- Department of Pharmacy, Center for Drug Research, Pharmacology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Hans Zischka
- Technical University Munich, School of Medicine, Institute of Toxicology and Environmental Hygiene, Biedersteiner Strasse 29, D-80802 Munich, Germany.,Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Ingolstaedter Landstrasse 1, D-85764 Neuherberg, Germany
| | - Angelika M Vollmar
- Department of Pharmacy, Center for Drug Research, Pharmaceutical Biology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Karin Bartel
- Department of Pharmacy, Center for Drug Research, Pharmaceutical Biology, Ludwig-Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
18
|
Krogsaeter E, Rosato AS, Grimm C. TRPMLs and TPCs: targets for lysosomal storage and neurodegenerative disease therapy? Cell Calcium 2022; 103:102553. [DOI: 10.1016/j.ceca.2022.102553] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/04/2022] [Accepted: 02/04/2022] [Indexed: 12/25/2022]
|
19
|
Defective Cystinosin, Aberrant Autophagy−Endolysosome Pathways, and Storage Disease: Towards Assembling the Puzzle. Cells 2022; 11:cells11030326. [PMID: 35159136 PMCID: PMC8834619 DOI: 10.3390/cells11030326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/03/2022] [Accepted: 01/11/2022] [Indexed: 02/05/2023] Open
Abstract
Epithelial cells that form the kidney proximal tubule (PT) rely on an intertwined ecosystem of vesicular membrane trafficking pathways to ensure the reabsorption of essential nutrients—a key requisite for homeostasis. The endolysosome stands at the crossroads of this sophisticated network, internalizing molecules through endocytosis, sorting receptors and nutrient transporters, maintaining cellular quality control via autophagy, and toggling the balance between PT differentiation and cell proliferation. Dysregulation of such endolysosome-guided trafficking pathways might thus lead to a generalized dysfunction of PT cells, often causing chronic kidney disease and life-threatening complications. In this review, we highlight the biological functions of endolysosome-residing proteins from the perspectives of understanding—and potentially reversing—the pathophysiology of rare inherited diseases affecting the kidney PT. Using cystinosis as a paradigm of endolysosome disease causing PT dysfunction, we discuss how the endolysosome governs the homeostasis of specialized epithelial cells. This review also provides a critical analysis of the molecular mechanisms through which defects in autophagy pathways can contribute to PT dysfunction, and proposes potential interventions for affected tissues. These insights might ultimately accelerate the discovery and development of new therapeutics, not only for cystinosis, but also for other currently intractable endolysosome-related diseases, eventually transforming our ability to regulate homeostasis and health.
Collapse
|
20
|
Chen Q, Fang J, Shen H, Chen L, Shi M, Huang X, Miao Z, Gong Y. Roles, molecular mechanisms, and signaling pathways of TMEMs in neurological diseases. Am J Transl Res 2021; 13:13273-13297. [PMID: 35035675 PMCID: PMC8748174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 11/10/2021] [Indexed: 06/14/2023]
Abstract
Transmembrane protein family members (TMEMs) span the entire lipid bilayer and act as channels that allow the transport of specific substances through biofilms. The functions of most TMEMs are unexplored. Numerous studies have shown that TMEMs are involved in the pathophysiological processes of various nervous system diseases, but the specific mechanisms of TMEMs in the pathogenesis of diseases remain unclear. In this review, we discuss the expression, physiological functions, and molecular mechanisms of TMEMs in brain tumors, psychiatric disorders, abnormal motor activity, cobblestone lissencephaly, neuropathic pain, traumatic brain injury, and other disorders of the nervous system. Additionally, we propose that TMEMs may be used as prognostic markers and potential therapeutic targets in patients with various neurological diseases.
Collapse
Affiliation(s)
- Qinghong Chen
- Affiliated Hospital of Jiangxi University of Traditional Chinese MedicineNanchang 330006, Jiangxi, China
| | - Junlin Fang
- Department of Acupuncture and Moxibustion, Banan Hospital of Traditional Chinese MedicineChongqing 401320, China
| | - Hui Shen
- Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese MedicineSuzhou 215600, Jiangsu, China
| | - Liping Chen
- Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese MedicineSuzhou 215600, Jiangsu, China
| | - Mengying Shi
- Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese MedicineSuzhou 215600, Jiangsu, China
| | - Xianbao Huang
- Affiliated Hospital of Jiangxi University of Traditional Chinese MedicineNanchang 330006, Jiangxi, China
| | - Zhiwei Miao
- Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese MedicineSuzhou 215600, Jiangsu, China
| | - Yating Gong
- Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese MedicineSuzhou 215600, Jiangsu, China
| |
Collapse
|
21
|
Ion Channels and Pumps in Autophagy: A Reciprocal Relationship. Cells 2021; 10:cells10123537. [PMID: 34944044 PMCID: PMC8700256 DOI: 10.3390/cells10123537] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/24/2022] Open
Abstract
Autophagy, the process of cellular self-degradation, is intrinsically tied to the degradative function of the lysosome. Several diseases have been linked to lysosomal degradative defects, including rare lysosomal storage disorders and neurodegenerative diseases. Ion channels and pumps play a major regulatory role in autophagy. Importantly, calcium signaling produced by TRPML1 (transient receptor potential cation channel, mucolipin subfamily) has been shown to regulate autophagic progression through biogenesis of autophagic-lysosomal organelles, activation of mTORC1 (mechanistic target of rapamycin complex 1) and degradation of autophagic cargo. ER calcium channels such as IP3Rs supply calcium for the lysosome, and lysosomal function is severely disrupted in the absence of lysosomal calcium replenishment by the ER. TRPML1 function is also regulated by LC3 (microtubule-associated protein light chain 3) and mTORC1, two critical components of the autophagic network. Here we provide an overview of the current knowledge about ion channels and pumps-including lysosomal V-ATPase (vacuolar proton-ATPase), which is required for acidification and hence proper enzymatic activity of lysosomal hydrolases-in the regulation of autophagy, and discuss how functional impairment of some of these leads to diseases.
Collapse
|
22
|
Prashar A, Capurro MI, Jones NL. Under the Radar: Strategies Used by Helicobacter pylori to Evade Host Responses. Annu Rev Physiol 2021; 84:485-506. [PMID: 34672717 DOI: 10.1146/annurev-physiol-061121-035930] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The body depends on its physical barriers and innate and adaptive immune responses to defend against the constant assault of potentially harmful microbes. In turn, successful pathogens have evolved unique mechanisms to adapt to the host environment and manipulate host defenses. Helicobacter pylori (Hp), a human gastric pathogen that is acquired in childhood and persists throughout life, is an example of a bacterium that is very successful at remodeling the host-pathogen interface to promote a long-term persistent infection. Using a combination of secreted virulence factors, immune subversion, and manipulation of cellular mechanisms, Hp can colonize and persist in the hostile environment of the human stomach. Here, we review the most recent and relevant information regarding how this successful pathogen overcomes gastric epithelial host defense responses to facilitate its own survival and establish a chronic infection. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Akriti Prashar
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada;
| | - Mariana I Capurro
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada;
| | - Nicola L Jones
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada; .,Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, Ontario, Canada.,Departments of Paediatrics and Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Misko A, Wood L, Kiselyov K, Slaugenhaupt S, Grishchuk Y. Progress in elucidating pathophysiology of mucolipidosis IV. Neurosci Lett 2021; 755:135944. [PMID: 33965501 PMCID: PMC8253105 DOI: 10.1016/j.neulet.2021.135944] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 11/19/2022]
Abstract
Mucolipidosis IV (MLIV) is an autosomal-recessive disease caused by loss-of-function mutations in the MCOLN1 gene encoding the non-selective cationic lysosomal channel transient receptor potential mucolipin-1 (TRPML1). Patients with MLIV suffer from severe motor and cognitive deficits that manifest in early infancy and progressive loss of vision leading to blindness in the second decade of life. There are no therapies available for MLIV and the unmet medical need is extremely high. Here we review the spectrum of clinical presentations and the latest research in the MLIV pre-clinical model, with the aim of highlighting the progress in understanding the pathophysiology of the disease. These highlights include elucidation of the neurodevelopmental versus neurodegenerative features over the course of disease, hypomyelination as one of the major brain pathological disease hallmarks, and dysregulation of cytokines, with emerging evidence of IFN-gamma pathway upregulation in response to TRPML1 loss and pro-inflammatory activation of astrocytes and microglia. These scientific advances in the MLIV field provide a basis for future translational research, including biomarker and therapy development, that are desperately needed for this patient population.
Collapse
Affiliation(s)
- Albert Misko
- Center for Genomic Medicine and Department of Neurology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, 02114, United States
| | - Levi Wood
- Georgia W. Woodruff School of Mechanical Engineering, Wallace H. Coulter Department of Biomedical Engineering, and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, United States
| | - Kirill Kiselyov
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, 15260, United States
| | - Susan Slaugenhaupt
- Center for Genomic Medicine and Department of Neurology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, 02114, United States
| | - Yulia Grishchuk
- Center for Genomic Medicine and Department of Neurology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA, 02114, United States.
| |
Collapse
|
24
|
Abstract
Lysosomal calcium is emerging as a modulator of autophagy and lysosomal compartment, an obligatory partner to complete the autophagic pathway. A variety of specific signals such as nutrient deprivation or oxidative stress can trigger lysosomal calcium-mediated nuclear translocation of the transcription factor EB (TFEB), a master regulator of global lysosomal function. Also, lysosomal calcium can promote the formation of autophagosome vesicles (AVs) by a mechanism that requires the production of the phosphoinositide PI3P by the VPS34 autophagic complex and the activation of the energy-sensing kinase AMPK. Additionally, lysosomal calcium plays a role in membrane fusion and fission events involved in cellular processes such as endocytic maturation, autophagosome-lysosome fusion, lysosomal exocytosis, and lysosomal reformation upon autophagy completion. Lysosomal calcium-dependent functions are defective in cellular and animal models of the non-selective cation channel TRPML1, whose mutations in humans cause the neurodegenerative lysosomal storage disease mucolipidosis type IV (MLIV). Lysosomal calcium is not only acting as a positive regulator of autophagy, but it is also responsible for turning-off this process through the reactivation of the mTOR kinase during prolonged starvation. More recently, it has been described the role of lysosomal calcium on an elegant sequence of intracellular signaling events such as membrane repair, lysophagy, and lysosomal biogenesis upon the induction of different grades of lysosomal membrane damage. Here, we will discuss these novel findings that re-define the importance of the lysosome and lysosomal calcium signaling at regulating cellular metabolism.
Collapse
|
25
|
Estradiol analogs attenuate autophagy, cell migration and invasion by direct and selective inhibition of TRPML1, independent of estrogen receptors. Sci Rep 2021; 11:8313. [PMID: 33859333 PMCID: PMC8050276 DOI: 10.1038/s41598-021-87817-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 04/05/2021] [Indexed: 12/17/2022] Open
Abstract
The cation channel TRPML1 is an important regulator of lysosomal function and autophagy. Loss of TRPML1 is associated with neurodegeneration and lysosomal storage disease, while temporary inhibition of this ion channel has been proposed to be beneficial in cancer therapy. Currently available TRPML1 channel inhibitors are not TRPML isoform selective and block at least two of the three human isoforms. We have now identified the first highly potent and isoform-selective TRPML1 antagonist, the steroid 17β-estradiol methyl ether (EDME). Two analogs of EDME, PRU-10 and PRU-12, characterized by their reduced activity at the estrogen receptor, have been identified through systematic chemical modification of the lead structure. EDME and its analogs, besides being promising new small molecule tool compounds for the investigation of TRPML1, selectively affect key features of TRPML1 function: autophagy induction and transcription factor EB (TFEB) translocation. In addition, they act as inhibitors of triple-negative breast cancer cell migration and invasion.
Collapse
|
26
|
Huang S, Li S, Feng H, Chen Y. Iron Metabolism Disorders for Cognitive Dysfunction After Mild Traumatic Brain Injury. Front Neurosci 2021; 15:587197. [PMID: 33796002 PMCID: PMC8007909 DOI: 10.3389/fnins.2021.587197] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 02/10/2021] [Indexed: 01/25/2023] Open
Abstract
Traumatic brain injury (TBI) is one of the most harmful forms of acute brain injury and predicted to be one of the three major neurological diseases that cause neurological disabilities by 2030. A series of secondary injury cascades often cause cognitive dysfunction of TBI patients leading to poor prognosis. However, there are still no effective intervention measures, which drive us to explore new therapeutic targets. In this process, the most part of mild traumatic brain injury (mTBI) is ignored because its initial symptoms seemed not serious. Unfortunately, the ignored mTBI accounts for 80% of the total TBI, and a large part of the patients have long-term cognitive dysfunction. Iron deposition has been observed in mTBI patients and accompanies the whole pathological process. Iron accumulation may affect long-term cognitive dysfunction from three pathways: local injury, iron deposition induces tau phosphorylation, the formation of neurofibrillary tangles; neural cells death; and neural network damage, iron deposition leads to axonal injury by utilizing the iron sensibility of oligodendrocytes. Thus, iron overload and metabolism dysfunction was thought to play a pivotal role in mTBI pathophysiology. Cerebrospinal fluid-contacting neurons (CSF-cNs) located in the ependyma have bidirectional communication function between cerebral-spinal fluid and brain parenchyma, and may participate in the pathway of iron-induced cognitive dysfunction through projected nerve fibers and transmitted factor, such as 5-hydroxytryptamine, etc. The present review provides an overview of the metabolism and function of iron in mTBI, and to seek a potential new treatment target for mTBI with a novel perspective through combined iron and CSF-cNs.
Collapse
Affiliation(s)
- Suna Huang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Su Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Third Military Medical University (Army Military Medical University), Chongqing, China
| |
Collapse
|
27
|
Lysosomal Calcium Channels in Autophagy and Cancer. Cancers (Basel) 2021; 13:cancers13061299. [PMID: 33803964 PMCID: PMC8001254 DOI: 10.3390/cancers13061299] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Autophagy is a cellular self-eating process that uses lysosome, the waste disposal system of the cell, to degrade and recycle intracellular materials to maintain cellular homeostasis. Defects in autophagy are linked to a variety of pathological states, including cancer. Calcium is an important cellular messenger that regulates the survival of all animal cells. Alterations to calcium homoeostasis are associated with cancer. While it has long been considered as cellular recycling center, the lysosome is now widely known as an intracellular calcium store that regulates autophagy and cancer progression by releasing calcium via some ion channels residing in the lysosomal membrane. In this review, we summarize existing mechanisms of autophagy regulation by lysosomal calcium channels and their implications in cancer development. We hope to guide readers toward a more in-depth understanding of the importance of lysosomal calcium channels in cancer, and potentially facilitate the development of new therapeutics for some cancers. Abstract Ca2+ is pivotal intracellular messenger that coordinates multiple cell functions such as fertilization, growth, differentiation, and viability. Intracellular Ca2+ signaling is regulated by both extracellular Ca2+ entry and Ca2+ release from intracellular stores. Apart from working as the cellular recycling center, the lysosome has been increasingly recognized as a significant intracellular Ca2+ store that provides Ca2+ to regulate many cellular processes. The lysosome also talks to other organelles by releasing and taking up Ca2+. In lysosomal Ca2+-dependent processes, autophagy is particularly important, because it has been implicated in many human diseases including cancer. This review will discuss the major components of lysosomal Ca2+ stores and their roles in autophagy and human cancer progression.
Collapse
|
28
|
Lee K, Jo YY, Chung G, Jung JH, Kim YH, Park CK. Functional Importance of Transient Receptor Potential (TRP) Channels in Neurological Disorders. Front Cell Dev Biol 2021; 9:611773. [PMID: 33748103 PMCID: PMC7969799 DOI: 10.3389/fcell.2021.611773] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
Transient receptor potential (TRP) channels are transmembrane protein complexes that play important roles in the physiology and pathophysiology of both the central nervous system (CNS) and the peripheral nerve system (PNS). TRP channels function as non-selective cation channels that are activated by several chemical, mechanical, and thermal stimuli as well as by pH, osmolarity, and several endogenous or exogenous ligands, second messengers, and signaling molecules. On the pathophysiological side, these channels have been shown to play essential roles in the reproductive system, kidney, pancreas, lung, bone, intestine, as well as in neuropathic pain in both the CNS and PNS. In this context, TRP channels have been implicated in several neurological disorders, including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and epilepsy. Herein, we focus on the latest involvement of TRP channels, with a special emphasis on the recently identified functional roles of TRP channels in neurological disorders related to the disruption in calcium ion homeostasis.
Collapse
Affiliation(s)
- Kihwan Lee
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon, South Korea
| | - Youn Yi Jo
- Department of Anesthesiology and Pain Medicine, Gil Medical Center, Gachon University, Incheon, South Korea
| | - Gehoon Chung
- Department of Oral Physiology and Program in Neurobiology, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Jung Hoon Jung
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Yong Ho Kim
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon, South Korea
| | - Chul-Kyu Park
- Gachon Pain Center and Department of Physiology, Gachon University College of Medicine, Incheon, South Korea
| |
Collapse
|
29
|
Abstract
Ion exchange between intracellular and extracellular spaces is the basic mechanism for controlling cell metabolism and signal transduction. This process is mediated by ion channels and transporters on the plasma membrane, or intracellular membranes that surround various organelles, in response to environmental stimuli. Macroautophagy (hereafter referred to as autophagy) is one of the lysosomal-dependent degradation pathways that maintains homeostasis through the degradation and recycling of cellular components (e.g., dysfunctional proteins and damaged organelles). Although autophagy-related (ATG) proteins play a central role in regulating the formation of autophagy-related member structures (e.g., phagophores, autophagosomes, and autolysosomes), the autophagic process also involves changes in expression and function of ion channels and transporters. Here we discuss current knowledge of the mechanisms that regulate autophagy in mammalian cells, with special attention to the ion channels and transporters. We also highlight prospects for the development of drugs targeting ion channels and transporters in autophagy.
Collapse
Affiliation(s)
- Ruoxi Zhang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
30
|
Ferroptosis and Its Modulation by Autophagy in Light of the Pathogenesis of Lysosomal Storage Diseases. Cells 2021; 10:cells10020365. [PMID: 33578654 PMCID: PMC7916399 DOI: 10.3390/cells10020365] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/31/2021] [Accepted: 02/06/2021] [Indexed: 12/13/2022] Open
Abstract
Ferroptosis is one of the recently described types of cell death which is dependent on many factors, including the accumulation of iron and lipid peroxidation. Its induction requires various signaling pathways. Recent discovery of ferroptosis induction pathways stimulated by autophagy, so called autophagy-dependent ferroptosis, put our attention on the role of ferroptosis in lysosomal storage diseases (LSD). Lysosome dysfunction, observed in these diseases, may influence ferroptosis efficiency, with as yet unknown consequences for the function of cells, tissues, and organisms, due to the effects of ferroptosis on physiological and pathological metabolic processes. Modulation of levels of ferrous ions and enhanced oxidative stress, which are primary markers of ferroptosis, are often described as processes associated with the pathology of LSD. Inhibition of autophagy flux and resultant accumulation of autophagosomes in neuronopathic LSD may induce autophagy-dependent ferroptosis, indicating a considerable contribution of this process in neurodegeneration. In this review article, we describe molecular mechanisms of ferroptosis in light of LSD, underlining the modulation of levels of ferroptosis markers in these diseases. Furthermore, we propose a hypothesis about the possible involvement of autophagy-dependent ferroptosis in these disorders.
Collapse
|
31
|
Vardi A, Pri-Or A, Wigoda N, Grishchuk Y, Futerman AH. Proteomics analysis of a human brain sample from a mucolipidosis type IV patient reveals pathophysiological pathways. Orphanet J Rare Dis 2021; 16:39. [PMID: 33478506 PMCID: PMC7818904 DOI: 10.1186/s13023-021-01679-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/06/2021] [Indexed: 12/29/2022] Open
Abstract
Background Mucolipidosis type IV (MLIV), an ultra-rare neurodevelopmental and neurodegenerative disorder, is caused by mutations in the MCOLN1 gene, which encodes the late endosomal/lysosomal transient receptor potential channel TRPML1 (mucolipin 1). The precise pathophysiogical pathways that cause neurological disease in MLIV are poorly understood. Recently, the first post-mortem brain sample became available from a single MLIV patient, and in the current study we performed mass spectrometry (MS)-based proteomics on this tissue with a view to delineating pathological pathways, and to compare with previously-published data on MLIV, including studies using the Mcoln1−/− mouse. Results A number of pathways were altered in two brain regions from the MLIV patient, including those related to the lysosome, lipid metabolism, myelination, cellular trafficking and autophagy, mTOR and calmodulin, the complement system and interferon signaling. Of these, levels of some proteins not known previously to be associated with MLIV were altered, including APOD, PLIN4, ATG and proteins related to interferon signaling. Moreover, when proteins detected by proteomics in the human brain were compared with their orthologs detected in the Mcoln1−/− mouse by RNAseq, the results were remarkably similar. Finally, analysis of proteins in human and mouse CSF suggest that calbindin 1 and calbindin 2 might be useful as biomarkers to help chart the course of disease development. Conclusions Despite the sample size limitations, our findings are consistent with the relatively general changes in lysosomal function previously reported in MLIV, and shed light on new pathways of disease pathophysiology, which is required in order to understand the course of disease development and to determine the efficacy of therapies when they become available for this devastating disease.
Collapse
Affiliation(s)
- Ayelet Vardi
- Department of Biomolecular Sciences, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Amir Pri-Or
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Noa Wigoda
- The Life Sciences Core Facilities, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Yulia Grishchuk
- Center for Genomic Medicine and Department of Neurology, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, 02114, USA
| | - Anthony H Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, 76100, Rehovot, Israel.
| |
Collapse
|
32
|
Engin AB, Engin A. Alzheimer's Disease and Protein Kinases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1275:285-321. [PMID: 33539020 DOI: 10.1007/978-3-030-49844-3_11] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and accounts for more than 60-80% of all cases of dementia. Loss of pyramidal neurons, extracellular amyloid beta (Abeta) accumulated senile plaques, and neurofibrillary tangles that contain hyperphosphorylated tau constitute the main pathological alterations in AD.Synaptic dysfunction and extrasynaptic N-methyl-D-aspartate receptor (NMDAR) hyperactivation contributes to excitotoxicity in patients with AD. Amyloid precursor protein (APP) and Abeta promoted neurodegeneration develop through the activation of protein kinase signaling cascade in AD. Furthermore, ultimate neuronal death in AD is under control of protein kinases-related signaling pathways. In this chapter, critical check-points within the cross-talk between neuron and protein kinases have been defined regarding the initiation and progression of AD. In this context, amyloid cascade hypothesis, neuroinflammation, oxidative stress, granulovacuolar degeneration, loss of Wnt signaling, Abeta-related synaptic alterations, prolonged calcium ions overload and NMDAR-related synaptotoxicity, damage signals hypothesis and type-3 diabetes are discussed briefly.In addition to clinical perspective of AD pathology, recommendations that might be effective in the treatment of AD patients have been reviewed.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey.
| | - Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
33
|
Two-pore and TRPML cation channels: Regulators of phagocytosis, autophagy and lysosomal exocytosis. Pharmacol Ther 2020; 220:107713. [PMID: 33141027 DOI: 10.1016/j.pharmthera.2020.107713] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023]
Abstract
The old Greek saying "Panta Rhei" ("everything flows") is true for all life and all living things in general. It also becomes nicely evident when looking closely into cells. There, material from the extracellular space is taken up by endocytic processes and transported to endosomes where it is sorted either for recycling or degradation. Cargo is also packaged for export through exocytosis involving the Golgi network, lysosomes and other organelles. Everything in this system is in constant motion and many proteins are necessary to coordinate transport along the different intracellular pathways to avoid chaos. Among these proteins are ion channels., in particular TRPML channels (mucolipins) and two-pore channels (TPCs) which reside on endosomal and lysosomal membranes to speed up movement between organelles, e.g. by regulating fusion and fission; they help readjust pH and osmolarity changes due to such processes, or they promote exocytosis of export material. Pathophysiologically, these channels are involved in neurodegenerative, metabolic, retinal and infectious diseases, cancer, pigmentation defects, and immune cell function, and thus have been proposed as novel pharmacological targets, e.g. for the treatment of lysosomal storage disorders, Duchenne muscular dystrophy, or different types of cancer. Here, we discuss the similarities but also differences of TPCs and TRPMLs in regulating phagocytosis, autophagy and lysosomal exocytosis, and we address the contradictions and open questions in the field relating to the roles TPCs and TRPMLs play in these different processes.
Collapse
|
34
|
Yan Y, Wang Y, Ding J, Lu L, Ke GJ, Dong K. TRPML1 Inhibited Photoreceptor Apoptosis and Protected the Retina by Activation of Autophagy in Experimental Retinal Detachment. Ophthalmic Res 2020; 64:587-594. [PMID: 33027790 DOI: 10.1159/000512104] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 09/23/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVE In this study, we used a rat model of retinal detachment (RD) to investigate the effects of transient receptor potential mucolipin 1 (TRPML1) on photoreceptor cells and the underlying mechanism. METHODS An RD model was established by subretinal injection of sodium hyaluronate, and mucolipin synthetic agonist 1 (ML-SA1) and dimethyl sulphoxide were subretinally injected after RD induction. Retinal morphology was observed using haematoxylin-eosin staining, and the apoptosis of photoreceptor cells was detected by transmission electron microscopy. Reactive oxygen species (ROS) were examined with an ROS detection kit. The retinal expression levels of TRPML1, the autophagy-related protein microtubule-associated protein 1 light chain 3 (LC3), Beclin 1, and cleaved caspase 3 were detected by Western blotting. The Morris water maze was used to test vision-dependent behaviour. RESULTS We found that retinal structure and the outer nuclear layer were improved and that the apoptosis of photoreceptor cells was reduced after ML-SA1 injection. The expression of ROS was reduced, and the loss of TRPML1 was inhibited after ML-SA1 treatment. The LC3-II to LC3-I ratio and Beclin 1 expression were enhanced, and cleaved caspase 3 expression was decreased after ML-SA1 treatment. Treatment with ML-SA1 also improved vision-dependent behaviour. CONCLUSIONS Our findings suggest that ML-SA1 attenuates photoreceptor apoptosis and improves vision-dependent behaviour by activation of autophagy.
Collapse
Affiliation(s)
- Yuanye Yan
- Department of Ophthalmology, Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yisai Wang
- Department of Ophthalmology, Anhui Medical University, Hefei, China
| | - Jie Ding
- Department of Ophthalmology, The Second People's Hospital of Hefei, Hefei, China
| | - Li Lu
- Department of Ophthalmology, Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Gen-Jie Ke
- Department of Ophthalmology, Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Kai Dong
- Department of Ophthalmology, Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
35
|
Zhong Y, Mohan K, Liu J, Al-Attar A, Lin P, Flight RM, Sun Q, Warmoes MO, Deshpande RR, Liu H, Jung KS, Mitov MI, Lin N, Butterfield DA, Lu S, Liu J, Moseley HNB, Fan TWM, Kleinman ME, Wang QJ. Loss of CLN3, the gene mutated in juvenile neuronal ceroid lipofuscinosis, leads to metabolic impairment and autophagy induction in retinal pigment epithelium. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165883. [PMID: 32592935 DOI: 10.1016/j.bbadis.2020.165883] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 06/08/2020] [Accepted: 06/16/2020] [Indexed: 12/13/2022]
Abstract
Juvenile neuronal ceroid lipofuscinosis (JNCL, aka. juvenile Batten disease or CLN3 disease) is a lysosomal storage disease characterized by progressive blindness, seizures, cognitive and motor failures, and premature death. JNCL is caused by mutations in the Ceroid Lipofuscinosis, Neuronal 3 (CLN3) gene, whose function is unclear. Although traditionally considered a neurodegenerative disease, CLN3 disease displays eye-specific effects: Vision loss not only is often one of the earliest symptoms of JNCL, but also has been reported in non-syndromic CLN3 disease. Here we described the roles of CLN3 protein in maintaining healthy retinal pigment epithelium (RPE) and normal vision. Using electroretinogram, fundoscopy and microscopy, we showed impaired visual function, retinal autofluorescent lesions, and RPE disintegration and metaplasia/hyperplasia in a Cln3 ~ 1 kb-deletion mouse model [1] on C57BL/6J background. Utilizing a combination of biochemical analyses, RNA-Seq, Seahorse XF bioenergetic analysis, and Stable Isotope Resolved Metabolomics (SIRM), we further demonstrated that loss of CLN3 increased autophagic flux, suppressed mTORC1 and Akt activities, enhanced AMPK activity, and up-regulated gene expression of the autophagy-lysosomal system in RPE-1 cells, suggesting autophagy induction. This CLN3 deficiency induced autophagy induction coincided with decreased mitochondrial oxygen consumption, glycolysis, the tricarboxylic acid (TCA) cycle, and ATP production. We also reported for the first time that loss of CLN3 led to glycogen accumulation despite of impaired glycogen synthesis. Our comprehensive analyses shed light on how loss of CLN3 affect autophagy and metabolism. This work suggests possible links among metabolic impairment, autophagy induction and lysosomal storage, as well as between RPE atrophy/degeneration and vision loss in JNCL.
Collapse
Affiliation(s)
- Yu Zhong
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Kabhilan Mohan
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY, United States
| | - Jinpeng Liu
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - Ahmad Al-Attar
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Penghui Lin
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Robert M Flight
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States; Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Qiushi Sun
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Marc O Warmoes
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Rahul R Deshpande
- Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Huijuan Liu
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States
| | - Kyung Sik Jung
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY, United States
| | - Mihail I Mitov
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | | | - D Allan Butterfield
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States; Department of Chemistry, University of Kentucky, Lexington, KY, United States
| | - Shuyan Lu
- Pfizer Inc., San Diego, CA, United States
| | - Jinze Liu
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States; Department of Computer Science, University of Kentucky, Lexington, KY, United States; Institute for Biomedical Informatics, University of Kentucky, Lexington, KY, United States
| | - Hunter N B Moseley
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States; Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY, United States; Institute for Biomedical Informatics, University of Kentucky, Lexington, KY, United States
| | - Teresa W M Fan
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States; Center for Environmental and Systems Biochemistry, University of Kentucky, Lexington, KY, United States; Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, United States
| | - Mark E Kleinman
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY, United States
| | - Qing Jun Wang
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY, United States; Markey Cancer Center, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
36
|
Evolutionary Aspects of TRPMLs and TPCs. Int J Mol Sci 2020; 21:ijms21114181. [PMID: 32545371 PMCID: PMC7312350 DOI: 10.3390/ijms21114181] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/05/2020] [Accepted: 06/10/2020] [Indexed: 01/02/2023] Open
Abstract
Transient receptor potential (TRP) or transient receptor potential channels are a highly diverse family of mostly non-selective cation channels. In the mammalian genome, 28 members can be identified, most of them being expressed predominantly in the plasma membrane with the exception of the mucolipins or TRPMLs which are expressed in the endo-lysosomal system. In mammalian organisms, TRPMLs have been associated with a number of critical endo-lysosomal functions such as autophagy, endo-lysosomal fusion/fission and trafficking, lysosomal exocytosis, pH regulation, or lysosomal motility and positioning. The related non-selective two-pore cation channels (TPCs), likewise expressed in endosomes and lysosomes, have also been found to be associated with endo-lysosomal trafficking, autophagy, pH regulation, or lysosomal exocytosis, raising the question why these two channel families have evolved independently. We followed TRP/TRPML channels and TPCs through evolution and describe here in which species TRP/TRPMLs and/or TPCs are found, which functions they have in different species, and how this compares to the functions of mammalian orthologs.
Collapse
|
37
|
Lloyd-Evans E, Waller-Evans H. Lysosomal Ca 2+ Homeostasis and Signaling in Health and Disease. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035311. [PMID: 31653642 DOI: 10.1101/cshperspect.a035311] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Calcium (Ca2+) signaling is an essential process in all cells that is maintained by a plethora of channels, pumps, transporters, receptors, and intracellular Ca2+ sequestering stores. Changes in cytosolic Ca2+ concentration govern processes as far reaching as fertilization, cell growth, and motility through to cell death. In recent years, lysosomes have emerged as a major intracellular Ca2+ storage organelle with an increasing involvement in triggering or regulating cellular functions such as endocytosis, autophagy, and Ca2+ release from the endoplasmic reticulum. This review will summarize recent work in the area of lysosomal Ca2+ signaling and homeostasis, including newly identified functions, and the involvement of lysosome-derived Ca2+ signals in human disease. In addition, we explore recent controversies in the techniques used for measurement of lysosomal Ca2+ content.
Collapse
Affiliation(s)
- Emyr Lloyd-Evans
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, United Kingdom
| | - Helen Waller-Evans
- Medicines Discovery Institute, Cardiff University, Cardiff CF10 3AT, United Kingdom
| |
Collapse
|
38
|
Darios F, Stevanin G. Impairment of Lysosome Function and Autophagy in Rare Neurodegenerative Diseases. J Mol Biol 2020; 432:2714-2734. [PMID: 32145221 PMCID: PMC7232018 DOI: 10.1016/j.jmb.2020.02.033] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 02/07/2023]
Abstract
Rare genetic diseases affect a limited number of patients, but their etiology is often known, facilitating the development of reliable animal models and giving the opportunity to investigate physiopathology. Lysosomal storage disorders are a group of rare diseases due to primary alteration of lysosome function. These diseases are often associated with neurological symptoms, which highlighted the importance of lysosome in neurodegeneration. Likewise, other groups of rare neurodegenerative diseases also present lysosomal alteration. Lysosomes fuse with autophagosomes and endosomes to allow the degradation of their content thanks to hydrolytic enzymes. It has emerged that alteration of the autophagy–lysosome pathway could play a critical role in neuronal death in many neurodegenerative diseases. Using a repertoire of selected rare neurodegenerative diseases, we highlight that a variety of alterations of the autophagy–lysosome pathway are associated with neuronal death. Yet, in most cases, it is still unclear why alteration of this pathway can lead to neurodegeneration. Lysosome function is impaired in many rare neurodegenerative diseases, making it a convergent point for these diseases. Impaired lysosome function is associated with alteration of the autophagy pathway. Autophagy–lysosome pathway can be impaired at various steps in different rare neurodegenerative diseases. The mechanisms linking impaired autophagy–lysosome pathway to neurodegeneration are still not fully elucidated.
Collapse
Affiliation(s)
- Frédéric Darios
- Sorbonne Université, F-75013, Paris, France; Inserm, U1127, F-75013 Paris, France; CNRS, UMR 7225, F-75013 Paris, France; Institut du Cerveau et de la Moelle Epinière, ICM, F-75013 Paris, France.
| | - Giovanni Stevanin
- Sorbonne Université, F-75013, Paris, France; Inserm, U1127, F-75013 Paris, France; CNRS, UMR 7225, F-75013 Paris, France; Institut du Cerveau et de la Moelle Epinière, ICM, F-75013 Paris, France; PSL Research University, Ecole Pratique des Hautes Etudes, Laboratoire de Neurogénétique, F-75013 Paris, France
| |
Collapse
|
39
|
Leong YQ, Ng KY, Chye SM, Ling APK, Koh RY. Mechanisms of action of amyloid-beta and its precursor protein in neuronal cell death. Metab Brain Dis 2020; 35:11-30. [PMID: 31811496 DOI: 10.1007/s11011-019-00516-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/14/2019] [Indexed: 02/08/2023]
Abstract
Extracellular senile plaques and intracellular neurofibrillary tangles are the neuropathological findings of the Alzheimer's disease (AD). Based on the amyloid cascade hypothesis, the main component of senile plaques, the amyloid-beta (Aβ) peptide, and its derivative called amyloid precursor protein (APP) both have been found to place their central roles in AD development for years. However, the recent therapeutics have yet to reverse or halt this disease. Previous evidence demonstrates that the accumulation of Aβ peptides and APP can exert neurotoxicity and ultimately neuronal cell death. Hence, we discuss the mechanisms of excessive production of Aβ peptides and APP serving as pathophysiologic stimuli for the initiation of various cell signalling pathways including apoptosis, necrosis, necroptosis and autophagy which lead to neuronal cell death. Conversely, the activation of such pathways could also result in the abnormal generation of APP and Aβ peptides. An elucidation of actions of APP and its metabolite, Aβ, could be vital in suggesting novel therapeutic opportunities.
Collapse
Affiliation(s)
- Yong Qi Leong
- School of Health Sciences, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia
| | - Soi Moi Chye
- School of Health Sciences, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Anna Pick Kiong Ling
- School of Health Sciences, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Rhun Yian Koh
- School of Health Sciences, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
40
|
Scotto Rosato A, Montefusco S, Soldati C, Di Paola S, Capuozzo A, Monfregola J, Polishchuk E, Amabile A, Grimm C, Lombardo A, De Matteis MA, Ballabio A, Medina DL. TRPML1 links lysosomal calcium to autophagosome biogenesis through the activation of the CaMKKβ/VPS34 pathway. Nat Commun 2019; 10:5630. [PMID: 31822666 PMCID: PMC6904751 DOI: 10.1038/s41467-019-13572-w] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 11/14/2019] [Indexed: 12/24/2022] Open
Abstract
The lysosomal calcium channel TRPML1, whose mutations cause the lysosomal storage disorder (LSD) mucolipidosis type IV (MLIV), contributes to upregulate autophagic genes by inducing the nuclear translocation of the transcription factor EB (TFEB). Here we show that TRPML1 activation also induces autophagic vesicle (AV) biogenesis through the generation of phosphatidylinositol 3-phosphate (PI3P) and the recruitment of essential PI3P-binding proteins to the nascent phagophore in a TFEB-independent manner. Thus, TRPML1 activation of phagophore formation requires the calcium-dependent kinase CaMKKβ and AMPK, which increase the activation of ULK1 and VPS34 autophagic protein complexes. Consistently, cells from MLIV patients show a reduced recruitment of PI3P-binding proteins to the phagophore during autophagy induction, suggesting that altered AV biogenesis is part of the pathological features of this disease. Together, we show that TRPML1 is a multistep regulator of autophagy that may be targeted for therapeutic purposes to treat LSDs and other autophagic disorders. It was known that prolonged TRMPL1 activation induces TFEB translocation and upregulates autophagic gene regulation. Here, the authors show that acute TRMPL1 activation also induces autophagy through VPS34 and by lysosomal calcium release independent of TFEB.
Collapse
Affiliation(s)
- A Scotto Rosato
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy.,Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - S Montefusco
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
| | - C Soldati
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
| | - S Di Paola
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
| | - A Capuozzo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
| | - J Monfregola
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
| | - E Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
| | - A Amabile
- Telethon Institute for Gene Therapy (SR-Tiget), Division of Regenerative Medicine, Stem Cells, and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - C Grimm
- Faculty of Medicine, Walther Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Munich, Germany
| | - A Lombardo
- Telethon Institute for Gene Therapy (SR-Tiget), Division of Regenerative Medicine, Stem Cells, and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - M A De Matteis
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy.,Department of Molecular Medicine and Medical Biotechnology, Federico II University, Naples, Italy
| | - A Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy.,Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy.,Baylor College of Medicine, Houston, Texas, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - D L Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy. .,Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy.
| |
Collapse
|
41
|
Calcium Dyshomeostasis and Lysosomal Ca 2+ Dysfunction in Amyotrophic Lateral Sclerosis. Cells 2019; 8:cells8101216. [PMID: 31597311 PMCID: PMC6829585 DOI: 10.3390/cells8101216] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 09/24/2019] [Accepted: 10/03/2019] [Indexed: 12/11/2022] Open
Abstract
Recent findings in the understanding of amyotrophic lateral sclerosis (ALS) revealed that alteration in calcium (Ca2+) homeostasis may largely contribute to motor neuron demise. A large part of these alterations is due to dysfunctional Ca2+-storing organelles, including the endoplasmic reticulum (ER) and mitochondria. Very recently, lysosomal Ca2+ dysfunction has emerged as an important pathological change leading to neuronal loss in ALS. Remarkably, the Ca2+-storing organelles are interacting with each other at specialized domains controlling mitochondrial dynamics, ER/lysosomal function, and autophagy. This occurs as a result of interaction between specific ionic channels and Ca2+-dependent proteins located in each structure. Therefore, the dysregulation of these ionic mechanisms could be considered as a key element in the neurodegenerative process. This review will focus on the possible role of lysosomal Ca2+ dysfunction in the pathogenesis of several neurodegenerative diseases, including ALS and shed light on the possibility that specific lysosomal Ca2+ channels might represent new promising targets for preventing or at least delaying neurodegeneration in ALS.
Collapse
|
42
|
The activation of Mucolipin TRP channel 1 (TRPML1) protects motor neurons from L-BMAA neurotoxicity by promoting autophagic clearance. Sci Rep 2019; 9:10743. [PMID: 31341250 PMCID: PMC6656764 DOI: 10.1038/s41598-019-46708-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 07/04/2019] [Indexed: 12/14/2022] Open
Abstract
Cellular clearance mechanisms including the autophagy-lysosome pathway are impaired in amyotrophic lateral sclerosis (ALS). One of the most important proteins involved in the regulation of autophagy is the lysosomal Ca2+ channel Mucolipin TRP channel 1 (TRPML1). Therefore, we investigated the role of TRPML1 in a neuronal model of ALS/Parkinson-dementia complex reproduced by the exposure of motor neurons to the cyanobacterial neurotoxin beta-methylamino-L-alanine (L-BMAA). Under these conditions, L-BMAA induces a dysfunction of the endoplasmic reticulum (ER) leading to ER stress and cell death. Therefore we hypothesized a dysfunctional coupling between lysosomes and ER in L-BMAA-treated motor neurons. Here, we showed that in motor neuronal cells TRPML1 as well as the lysosomal protein LAMP1 co-localized with ER. In addition, TRPML1 co-immunoprecipitated with the ER Ca2+ sensor STIM1. Functionally, the TRPML1 agonist ML-SA1 induced lysosomal Ca2+ release in a dose-dependent way in motor neuronal cells. The SERCA inhibitor thapsigargin increased the fluorescent signal associated with lysosomal Ca2+ efflux in the cells transfected with the genetically encoded Ca2+ indicator GCaMP3-ML1, thus suggesting an interplay between the two organelles. Moreover, chronic exposure to L-BMAA reduced TRPML1 protein expression and produced an impairment of both lysosomal and ER Ca2+ homeostasis in primary motor neurons. Interestingly, the preincubation of ML-SA1, by an early activation of AMPK and beclin 1, rescued motor neurons from L-BMAA-induced cell death and reduced the expression of the ER stress marker GRP78. Finally, ML-SA1 reduced the accumulation of the autophagy-related proteins p62/SQSTM1 and LC3-II in L-BMAA-treated motor neurons. Collectively, we propose that the pharmacological stimulation of TRPML1 can rescue motor neurons from L-BMAA-induced toxicity by boosting autophagy and reducing ER stress.
Collapse
|
43
|
Liu EA, Lieberman AP. The intersection of lysosomal and endoplasmic reticulum calcium with autophagy defects in lysosomal diseases. Neurosci Lett 2019; 697:10-16. [PMID: 29704574 PMCID: PMC6202281 DOI: 10.1016/j.neulet.2018.04.049] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 04/13/2018] [Accepted: 04/24/2018] [Indexed: 01/01/2023]
Abstract
The lysosomal storage disorders (LSDs) encompass a group of more than 50 inherited diseases characterized by the accumulation of lysosomal substrates. Two-thirds of patients experience significant neurological symptoms, but the mechanisms of neurodegeneration are not well understood. Interestingly, a wide range of LSDs show defects in both autophagy and Ca2+ homeostasis, which is notable as Ca2+ is a key regulator of autophagy. The crosstalk between these pathways in the context of LSD pathogenesis is not well characterized, but further understanding of this relationship could open up promising therapeutic targets. This review discusses the role of endoplasmic reticulum and lysosomal Ca2+ in autophagy regulation and highlights what is known about defects in autophagy and Ca2+ homeostasis in two LSDs, Niemann-Pick type C disease and Gaucher disease.
Collapse
Affiliation(s)
- Elaine A Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, United States; Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI 48109, United States; Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, United States.
| |
Collapse
|
44
|
Boudewyn LC, Walkley SU. Current concepts in the neuropathogenesis of mucolipidosis type IV. J Neurochem 2019; 148:669-689. [PMID: 29770442 PMCID: PMC6239999 DOI: 10.1111/jnc.14462] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/29/2018] [Accepted: 05/02/2018] [Indexed: 12/11/2022]
Abstract
Mucolipidosis type IV (MLIV) is an autosomal recessive, lysosomal storage disorder causing progressively severe intellectual disability, motor and speech deficits, retinal degeneration often culminating in blindness, and systemic disease causing a shortened lifespan. MLIV results from mutations in the gene MCOLN1 encoding the transient receptor potential channel mucolipin-1. It is an ultra-rare disease and is currently known to affect just over 100 diagnosed individuals. The last decade has provided a wealth of research focused on understanding the role of the enigmatic mucolipin-1 protein in cell and brain function and how its absence causes disease. This review explores our current understanding of the mucolipin-1 protein in relation to neuropathogenesis in MLIV and describes recent findings implicating mucolipin-1's important role in mechanistic target of rapamycin and TFEB (transcription factor EB) signaling feedback loops as well as in the function of the greater endosomal/lysosomal system. In addition to addressing the vital role of mucolipin-1 in the brain, we also report new data on the question of whether haploinsufficiency as would be anticipated in MCOLN1 heterozygotes is associated with any evidence of neuron dysfunction or disease. Greater insights into the role of mucolipin-1 in the nervous system can be expected to shed light not only on MLIV disease but also on numerous processes governing normal brain function. This article is part of the Special Issue "Lysosomal Storage Disorders".
Collapse
Affiliation(s)
- Lauren C. Boudewyn
- Dominick P. Purpura Department of Neuroscience, Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, New York
| | - Steven U. Walkley
- Dominick P. Purpura Department of Neuroscience, Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
45
|
Chen LL, Huang YJ, Cui JT, Song N, Xie J. Iron Dysregulation in Parkinson's Disease: Focused on the Autophagy-Lysosome Pathway. ACS Chem Neurosci 2019; 10:863-871. [PMID: 30590010 DOI: 10.1021/acschemneuro.8b00390] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and is characterized by dopaminergic neuron loss in the substantia nigra pars compacta (SNpc). Although both iron accumulation and a defective autophagy-lysosome pathway contribute to the pathological development of PD, the connection between these two causes is poorly documented. The autophagy-lysosome pathway not only responds to regulation by iron chelators and channels but also participates in cellular iron recycling through the degradation of ferritin and other iron-containing components. Previously, ferritin has been posited to be the bridge between iron accumulation and autophagy impairment in PD. In addition, iron directly interacts with α-synuclein in Lewy bodies, which are primarily digested through the autophagy-lysosome pathway. These findings indicate that some link exists between iron deposition and autophagy impairment in PD. In this review, the basic mechanisms of the autophagy-lysosome pathway and iron trafficking are introduced, and then their interaction under physiological conditions is explained. Finally, we finish by discussing the dysfunction of iron deposition and autophagy in PD, as well as their potential relationship, which will provide some insight for further study.
Collapse
Affiliation(s)
- Lei-Lei Chen
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong 266071, China
| | - Yu-Jv Huang
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong 266071, China
| | - Jun-tao Cui
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong 266071, China
| | - Ning Song
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong 266071, China
| | - Junxia Xie
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, Shandong 266071, China
| |
Collapse
|
46
|
Degradation of TRPML1 in Neurons Reduces Neuron Survival in Transient Global Cerebral Ischemia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4612727. [PMID: 30662583 PMCID: PMC6312622 DOI: 10.1155/2018/4612727] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 09/28/2018] [Accepted: 10/15/2018] [Indexed: 11/17/2022]
Abstract
Postcardiac arrest syndrome yields poor neurological outcomes, but the mechanisms underlying this condition remain poorly understood. Autophagy plays an important role in neuronal apoptosis induced by ischemia. However, whether autophagy is involved in neuron apoptosis induced by cardiac arrest has been less studied. This study found that TRPML1 participates in cerebral ischemic reperfusion injury. Primary neurons were isolated and treated with mucolipin synthetic agonist 1 (ML-SA1), as well as infected with the recombinant lentivirus TRPML1 overexpression vector in vitro. ML-SA1 was delivered intracerebroventricularly in transient global ischemia model. Protein expression levels were determined by western blot. Neurological deficit score and the infarct volume were analyzed for the detection of neuronal damage. We found that TRPML1 was significantly downregulated in vivo and in vitro ischemic reperfusion model. We also observed that TRPML1 overexpression or treatment with the ML-SA1 attenuated neuronal death in primary neurons and ameliorated neurological dysfunction in vivo. Our findings suggested that autophagy and apoptosis were activated after transient global ischemia. Administration of ML-SA1 before transient global ischemia ameliorated neurological dysfunction possibly through the promotion of autophagy and the inhibition of apoptosis.
Collapse
|
47
|
Sun W, Yue J. TPC2 mediates autophagy progression and extracellular vesicle secretion in cancer cells. Exp Cell Res 2018; 370:478-489. [PMID: 29990474 DOI: 10.1016/j.yexcr.2018.07.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/04/2018] [Accepted: 07/06/2018] [Indexed: 02/06/2023]
Abstract
Autophagy is an evolutionarily conserved lysosomal degradation process, and is involved in various cellular processes. Here we studied the role of two pore channel 2 (TPC2), a lysosomal non-selective Na+/Ca2+ channel, in autophagy progression. We found that TPC overexpression in 4T1 mouse breast cancer cell line or in HeLa human cervical cancer cell line inhibited the fusion between autophagosome and lysosome, resulting in the accumulation of autophagosomes accompanied with increased lysosomal pH and TFEB nuclear localization. Interestingly, we also found that extracellular vesicle (EV) secretion was markedly decreased in TPC2 overexpressing cells but was induced in TPC2 knockdown cells. In addition, migration of TPC2 knockdown cells, not TPC2 overexpressing cells, was inhibited. Taken together, these results support a role of TPC2 in autophagy progression and EV trafficking in cancer cells.
Collapse
Affiliation(s)
- Wei Sun
- City University of Hong Kong ShenZhen Research Institute, ShenZhen, China; Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Jianbo Yue
- City University of Hong Kong ShenZhen Research Institute, ShenZhen, China; Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
48
|
Weinstock LD, Furness AM, Herron SS, Smith SS, Sankar SB, DeRosa SG, Gao D, Mepyans ME, Scotto Rosato A, Medina DL, Vardi A, Ferreira NS, Cho SM, Futerman AH, Slaugenhaupt SA, Wood LB, Grishchuk Y. Fingolimod phosphate inhibits astrocyte inflammatory activity in mucolipidosis IV. Hum Mol Genet 2018; 27:2725-2738. [PMID: 29771310 PMCID: PMC6915831 DOI: 10.1093/hmg/ddy182] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/09/2018] [Accepted: 05/08/2018] [Indexed: 12/25/2022] Open
Abstract
Mucolipidosis IV (MLIV) is an orphan neurodevelopmental disease that causes severe neurologic dysfunction and loss of vision. Currently there is no therapy for MLIV. It is caused by loss of function of the lysosomal channel mucolipin-1, also known as TRPML1. Knockout of the Mcoln1 gene in a mouse model mirrors clinical and neuropathologic signs in humans. Using this model, we previously observed robust activation of microglia and astrocytes in early symptomatic stages of disease. Here we investigate the consequence of mucolipin-1 loss on astrocyte inflammatory activation in vivo and in vitro and apply a pharmacologic approach to restore Mcoln1-/- astrocyte homeostasis using a clinically approved immunomodulator, fingolimod. We found that Mcoln1-/- mice over-express numerous pro-inflammatory cytokines, some of which were also over-expressed in astrocyte cultures. Changes in the cytokine profile in Mcoln1-/- astrocytes are concomitant with changes in phospho-protein signaling, including activation of PI3K/Akt and MAPK pathways. Fingolimod promotes cytokine homeostasis, down-regulates signaling within the PI3K/Akt and MAPK pathways and restores the lysosomal compartment in Mcoln1-/- astrocytes. These data suggest that fingolimod is a promising candidate for preclinical evaluation in our MLIV mouse model, which, in case of success, can be rapidly translated into clinical trial.
Collapse
Affiliation(s)
- Laura D Weinstock
- George W. Woodruff School of Mechanical Engineering, Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory, Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Dr., Atlanta, GA, USA
| | - Amanda M Furness
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| | - Shawn S Herron
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| | - Sierra S Smith
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| | - Sitara B Sankar
- George W. Woodruff School of Mechanical Engineering, Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory, Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Dr., Atlanta, GA, USA
| | - Samantha G DeRosa
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| | - Dadi Gao
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| | - Molly E Mepyans
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| | - Anna Scotto Rosato
- Telethon Institute of Genetics and Medicine (TIGEM), via Campi Flegrei 34, Pozzuoli (NA), Italy
| | - Diego L Medina
- Telethon Institute of Genetics and Medicine (TIGEM), via Campi Flegrei 34, Pozzuoli (NA), Italy
| | - Ayelet Vardi
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Natalia S Ferreira
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Winterthurerstrasse 260, Zurich, Switzerland
| | - Soo Min Cho
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Anthony H Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Susan A Slaugenhaupt
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| | - Levi B Wood
- George W. Woodruff School of Mechanical Engineering, Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory, Parker H. Petit Institute for Bioengineering & Bioscience, Georgia Institute of Technology, 315 Ferst Dr., Atlanta, GA, USA
| | - Yulia Grishchuk
- Department of Neurology, Center for Genomic Medicine, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA, USA
| |
Collapse
|
49
|
Abstract
One of the fundamental properties of the cell is the capability to digest and remodel its own components according to metabolic and developmental needs. This is accomplished via the autophagy-lysosome system, a pathway of critical importance in the brain, where it contributes to neuronal plasticity and must protect nonreplaceable neurons from the potentially harmful accumulation of cellular waste. The study of lysosomal biogenesis and function in the context of common and rare neurodegenerative diseases has revealed that a dysfunctional autophagy-lysosome system is the shared nexus where multiple, interconnected pathogenic events take place. The characterization of pathways and mechanisms regulating the lysosomal system and autophagic clearance offers unprecedented opportunities for the development of polyvalent therapeutic strategies based on the enhancement of the autophagy-lysosome pathway to maintain cellular homeostasis and achieve neuroprotection.
Collapse
Affiliation(s)
- Jaiprakash Sharma
- Department of Molecular and Human Genetics, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA;
| | - Alberto di Ronza
- Department of Molecular and Human Genetics, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA;
| | - Parisa Lotfi
- Department of Molecular and Human Genetics, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA;
| | - Marco Sardiello
- Department of Molecular and Human Genetics, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas 77030, USA;
| |
Collapse
|
50
|
Li H, Pei W, Vergarajauregui S, Zerfas PM, Raben N, Burgess SM, Puertollano R. Novel degenerative and developmental defects in a zebrafish model of mucolipidosis type IV. Hum Mol Genet 2018; 26:2701-2718. [PMID: 28449103 DOI: 10.1093/hmg/ddx158] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/19/2017] [Indexed: 12/20/2022] Open
Abstract
Mucolipidosis type IV (MLIV) is a lysosomal storage disease characterized by neurologic and ophthalmologic abnormalities. There is currently no effective treatment. MLIV is caused by mutations in MCOLN1, a lysosomal cation channel from the transient receptor potential (TRP) family. In this study, we used genome editing to knockout the two mcoln1 genes present in Danio rerio (zebrafish). Our model successfully reproduced the retinal and neuromuscular defects observed in MLIV patients, indicating that this model is suitable for studying the disease pathogenesis. Importantly, our model revealed novel insights into the origins and progression of the MLIV pathology, including the contribution of autophagosome accumulation to muscle dystrophy and the role of mcoln1 in embryonic development, hair cell viability and cellular maintenance. The generation of a MLIV model in zebrafish is particularly relevant given the suitability of this organism for large-scale in vivo drug screening, thus providing unprecedented opportunities for therapeutic discovery.
Collapse
Affiliation(s)
- Huiqing Li
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wuhong Pei
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sivia Vergarajauregui
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.,Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Patricia M Zerfas
- Office of Research Services, Division of Veterinary Resources, National Institutes of Health, Bethesda, MD, USA
| | - Nina Raben
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shawn M Burgess
- Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rosa Puertollano
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|