1
|
Kraus F, He Y, Swarup S, Overmyer KA, Jiang Y, Brenner J, Capitanio C, Bieber A, Jen A, Nightingale NM, Anderson BJ, Lee C, Paulo JA, Smith IR, Plitzko JM, Gygi SP, Schulman BA, Wilfling F, Coon JJ, Harper JW. Global cellular proteo-lipidomic profiling of diverse lysosomal storage disease mutants using nMOST. SCIENCE ADVANCES 2025; 11:eadu5787. [PMID: 39841834 PMCID: PMC11753374 DOI: 10.1126/sciadv.adu5787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025]
Abstract
Lysosomal storage diseases (LSDs) comprise ~50 monogenic disorders marked by the buildup of cellular material in lysosomes, yet systematic global molecular phenotyping of proteins and lipids is lacking. We present a nanoflow-based multiomic single-shot technology (nMOST) workflow that quantifies HeLa cell proteomes and lipidomes from over two dozen LSD mutants. Global cross-correlation analysis between lipids and proteins identified autophagy defects, notably the accumulation of ferritinophagy substrates and receptors, especially in NPC1-/- and NPC2-/- mutants, where lysosomes accumulate cholesterol. Autophagic and endocytic cargo delivery failures correlated with elevated lysophosphatidylcholine species and multilamellar structures visualized by cryo-electron tomography. Loss of mitochondrial cristae, MICOS complex components, and OXPHOS components rich in iron-sulfur cluster proteins in NPC2-/- cells was largely alleviated when iron was provided through the transferrin system. This study reveals how lysosomal dysfunction affects mitochondrial homeostasis and underscores nMOST as a valuable discovery tool for identifying molecular phenotypes across LSDs.
Collapse
Affiliation(s)
- Felix Kraus
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Yuchen He
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- Department of Biomolecular Chemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Sharan Swarup
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Katherine A. Overmyer
- Department of Biomolecular Chemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Yizhi Jiang
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Johann Brenner
- Mechanisms of Cellular Quality Control, Max Planck Institute of Biophysics, Frankfurt, Germany
- CryoEM Technology, Max Planck Institute of Biochemistry, Munich, Germany
| | - Cristina Capitanio
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Anna Bieber
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Annie Jen
- Department of Biomolecular Chemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Nicole M. Nightingale
- Department of Biomolecular Chemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Benton J. Anderson
- Department of Biomolecular Chemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Chan Lee
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Joao A. Paulo
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Ian R. Smith
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jürgen M. Plitzko
- CryoEM Technology, Max Planck Institute of Biochemistry, Munich, Germany
| | - Steven P. Gygi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Brenda A. Schulman
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Florian Wilfling
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Mechanisms of Cellular Quality Control, Max Planck Institute of Biophysics, Frankfurt, Germany
| | - Joshua J. Coon
- Department of Biomolecular Chemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Chemistry, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - J. Wade Harper
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| |
Collapse
|
2
|
Cheetham-Wilkinson IJ, Sivalingam B, Flitton C, Flottmann F, Vehling L, Drechsler M, Stojchevska M, Raimondi A, Paululat A, Fröhlich F, Swan LE, Stagi M. RpH-ILV: Probe for lysosomal pH and acute LLOMe-induced membrane permeabilization in cell lines and Drosophila. SCIENCE ADVANCES 2025; 11:eadr7325. [PMID: 39752501 PMCID: PMC11698090 DOI: 10.1126/sciadv.adr7325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 12/02/2024] [Indexed: 01/06/2025]
Abstract
Lysosomal pH dysregulation is a critical element of the pathophysiology of neurodegenerative diseases, cancers, and lysosomal storage disorders (LSDs). To study the role of lysosomes in pathophysiology, probes to analyze lysosomal size, positioning, and pH are indispensable tools. Here, we developed and characterized a ratiometric genetically encoded lysosomal pH probe, RpH-ILV, targeted to a subpopulation of lysosomal intraluminal vesicles. This subpopulation behaves similarly to the general population of LAMP1-positive vesicles in terms of pH response to pharmacological stresses. In addition, RpH-ILV, which is trafficked to the lysosome via a different cytosolic motif than our previous ratiometric sensor, RpH-LAMP1, is well tolerated by the model organism Drosophila melanogaster, exhibits minimal plasma membrane fluorescence, and reveals sensitivity to the lysosomal damaging agent LLOMe, adding a valuable tool to our repertoire of lysosomal pH sensors.
Collapse
Affiliation(s)
- Izaak J. Cheetham-Wilkinson
- Department of Biochemistry Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Bhavya Sivalingam
- Division of Molecular Membrane Biology, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
| | - Chloe Flitton
- Department of Biochemistry Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Franziska Flottmann
- Department of Zoology & Developmental Biology, Osnabrück University, 49076 Osnabrück, Germany
| | - Luisa Vehling
- Department of Zoology & Developmental Biology, Osnabrück University, 49076 Osnabrück, Germany
| | - Maik Drechsler
- Department of Zoology & Developmental Biology, Osnabrück University, 49076 Osnabrück, Germany
| | - Marija Stojchevska
- Department of Biochemistry Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Andrea Raimondi
- Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy
- Università della Svizzera Italiana (USI), Faculty of Biomedical Sciences, Institute for Research in Biomedicine, CH-6500 Bellinzona, Switzerland
| | - Achim Paululat
- Department of Zoology & Developmental Biology, Osnabrück University, 49076 Osnabrück, Germany
- Center of Cellular Nanoanalytics Osnabrück – CellNanOs, Osnabrück University, 49076 Osnabrück, Germany
| | - Florian Fröhlich
- Division of Molecular Membrane Biology, Department of Biology/Chemistry, Osnabrück University, 49076 Osnabrück, Germany
- Center of Cellular Nanoanalytics Osnabrück – CellNanOs, Osnabrück University, 49076 Osnabrück, Germany
| | - Laura E. Swan
- Department of Biochemistry Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Massimiliano Stagi
- Department of Biochemistry Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| |
Collapse
|
3
|
Domingues N, Pires J, Milosevic I, Raimundo N. Role of lipids in interorganelle communication. Trends Cell Biol 2025; 35:46-58. [PMID: 38866684 PMCID: PMC11632148 DOI: 10.1016/j.tcb.2024.04.008] [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: 02/06/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 06/14/2024]
Abstract
Cell homeostasis and function rely on well-orchestrated communication between different organelles. This communication is ensured by signaling pathways and membrane contact sites between organelles. Many players involved in organelle crosstalk have been identified, predominantly proteins and ions. The role of lipids in interorganelle communication remains poorly understood. With the development and broader availability of methods to quantify lipids, as well as improved spatiotemporal resolution in detecting different lipid species, the contribution of lipids to organelle interactions starts to be evident. However, the specific roles of various lipid molecules in intracellular communication remain to be studied systematically. We summarize new insights in the interorganelle communication field from the perspective of organelles and discuss the roles played by lipids in these complex processes.
Collapse
Affiliation(s)
- Neuza Domingues
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
| | - Joana Pires
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
| | - Ira Milosevic
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal; Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nuno Raimundo
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal; Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA; Penn State Cancer Institute, Hershey, PA, USA.
| |
Collapse
|
4
|
Ludlaim AM, Waddington SN, McKay TR. Unifying biology of neurodegeneration in lysosomal storage diseases. J Inherit Metab Dis 2025; 48:e12833. [PMID: 39822020 PMCID: PMC11739831 DOI: 10.1002/jimd.12833] [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: 06/06/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 01/19/2025]
Abstract
There are currently at least 70 characterised lysosomal storage diseases (LSD) resultant from inherited single-gene defects. Of these, at least 30 present with central nervous system (CNS) neurodegeneration and overlapping aetiology. Substrate accumulation and dysfunctional neuronal lysosomes are common denominator, but how variants in 30 different genes converge on this central cellular phenotype is unclear. Equally unresolved is how the accumulation of a diverse spectrum of substrates in the neuronal lysosomes results in remarkably similar neurodegenerative outcomes. Conversely, how is it that many other monogenic LSDs cause only visceral disease? Lysosomal substance accumulation in LSDs with CNS neurodegeneration (nLSD) includes lipofuscinoses, mucopolysaccharidoses, sphingolipidoses and glycoproteinoses. Here, we review the latest discoveries in the fundamental biology of four classes of nLSDs, comparing and contrasting new insights into disease mechanism with emerging evidence of unifying convergence.
Collapse
Affiliation(s)
- Anna M Ludlaim
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Simon N Waddington
- Gene Transfer Technology Group, EGA-Institute for Women's Health, University College London, London, UK
- Faculty of Health Sciences, Wits/SAMRC Antiviral Gene Therapy Research Unit, Johannesburg, South Africa
| | - Tristan R McKay
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| |
Collapse
|
5
|
Chung CY, Singh K, Sheshadri P, Valdebenito GE, Chacko AR, Costa Besada MA, Liang XF, Kabir L, Pitceathly RDS, Szabadkai G, Duchen MR. Inhibition of the PI3K-AKT-MTORC1 axis reduces the burden of the m.3243A>G mtDNA mutation by promoting mitophagy and improving mitochondrial function. Autophagy 2024:1-16. [PMID: 39667405 DOI: 10.1080/15548627.2024.2437908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 11/20/2024] [Accepted: 11/29/2024] [Indexed: 12/14/2024] Open
Abstract
Mitochondrial DNA (mtDNA) encodes genes essential for oxidative phosphorylation. The m.3243A>G mutation causes severe disease, including myopathy, lactic acidosis and stroke-like episodes (MELAS) and is the most common pathogenic mtDNA mutation in humans. We have previously shown that the mutation is associated with constitutive activation of the PI3K-AKT-MTORC1 axis. Inhibition of this pathway in patient fibroblasts reduced the mutant load, rescued mitochondrial bioenergetic function and reduced glucose dependence. We have now investigated the mechanisms that select against the mutant mtDNA under these conditions. Basal macroautophagy/autophagy and lysosomal degradation of mitochondria were suppressed in the mutant cells. Pharmacological inhibition of any step of the PI3K-AKT-MTORC1 pathway activated mitophagy and progressively reduced m.3243A>G mutant load over weeks. Inhibition of autophagy with bafilomycin A1 or chloroquine prevented the reduction in mutant load, suggesting that mitophagy was necessary to remove the mutant mtDNA. Inhibition of the pathway was associated with metabolic remodeling - mitochondrial membrane potential and respiratory rate improved even before a measurable fall in mutant load and proved crucial for mitophagy. Thus, maladaptive activation of the PI3K-AKT-MTORC1 axis and impaired autophagy play a major role in shaping the presentation and progression of disease caused by the m.3243A>G mutation. Our findings highlight a potential therapeutic target for this otherwise intractable disease.Abbreviation: ΔΨm: mitochondrial membrane potential; 2DG: 2-deoxy-D-glucose; ANOVA: analysis of variance; ARMS-qPCR: amplification-refractory mutation system quantitative polymerase chain reaction; Baf A1: bafilomycin A1; BSA: bovine serum albumin; CQ: chloroquine; Cybrid: cytoplasmic hybrid; CYCS: cytochrome c, somatic; DCA: dichloroacetic acid; DMEM: Dulbecco's modified Eagle's medium; DMSO: dimethylsulfoxide; EGFP: enhanced green fluorescent protein; LC3B-I: carboxy terminus cleaved microtubule-associated protein 1 light chain 3 beta; LC3B-II: lipidated microtubule-associated protein 1 light chain 3 beta; LY: LY290042; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MELAS: mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes; MFC: mitochondrial fragmentation count; mt-Keima: mitochondrial-targeted mKeima; mtDNA: mitochondrial DNA/mitochondrial genome; MTOR: mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; OA: oligomycin+antimycin A; OxPhos: oxidative phosphorylation; DPBS: Dulbecco's phosphate-buffered saline; PPARGC1A/PGC-1α: PPARG coactivator 1 alpha; PPARGC1B/PGC-1β: PPARG coactivator 1 beta; PI3K: phosphoinositide 3-kinase; PINK1: PTEN induced kinase 1; qPCR: quantitative polymerase chain reaction; RNA-seq: RNA sequencing; RP: rapamycin; SQSTM1/p62: sequestosome 1; TEM: transmission electron microscopy; WT: wild-type.
Collapse
Affiliation(s)
- Chih-Yao Chung
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| | - Kritarth Singh
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| | - Preethi Sheshadri
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| | - Gabriel E Valdebenito
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| | - Anitta R Chacko
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| | - María Alicia Costa Besada
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
- Cellular and Molecular Neurobiology of Parkinson's Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Spain
| | - Xiao Fei Liang
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| | - Lida Kabir
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| | - Robert D S Pitceathly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Gyorgy Szabadkai
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- The Francis Crick Institute, London, UK
| | - Michael R Duchen
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| |
Collapse
|
6
|
Kraus F, He Y, Swarup S, Overmyer KA, Jiang Y, Brenner J, Capitanio C, Bieber A, Jen A, Nightingale NM, Anderson BJ, Lee C, Paulo JA, Smith IR, Plitzko JM, Gygi SP, Schulman BA, Wilfling F, Coon JJ, Harper JW. Global cellular proteo-lipidomic profiling of diverse lysosomal storage disease mutants using nMOST. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586828. [PMID: 38585873 PMCID: PMC10996675 DOI: 10.1101/2024.03.26.586828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Lysosomal storage diseases (LSDs) comprise ~50 monogenic disorders marked by the buildup of cellular material in lysosomes, yet systematic global molecular phenotyping of proteins and lipids is lacking. We present a nanoflow-based multi-omic single-shot technology (nMOST) workflow that quantifies HeLa cell proteomes and lipidomes from over two dozen LSD mutants. Global cross-correlation analysis between lipids and proteins identified autophagy defects, notably the accumulation of ferritinophagy substrates and receptors, especially in NPC1 -/- and NPC2 -/- mutants, where lysosomes accumulate cholesterol. Autophagic and endocytic cargo delivery failures correlated with elevated lyso-phosphatidylcholine species and multi-lamellar structures visualized by cryo-electron tomography. Loss of mitochondrial cristae, MICOS-complex components, and OXPHOS components rich in iron-sulfur cluster proteins in NPC2 -/- cells was largely alleviated when iron was provided through the transferrin system. This study reveals how lysosomal dysfunction affects mitochondrial homeostasis and underscores nMOST as a valuable discovery tool for identifying molecular phenotypes across LSDs.
Collapse
Affiliation(s)
- Felix Kraus
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- equal contribution
| | - Yuchen He
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- equal contribution
| | - Sharan Swarup
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Katherine A Overmyer
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yizhi Jiang
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Johann Brenner
- Mechanisms of Cellular Quality Control, Max Planck Institute of Biophysics, Frankfurt, Germany
- CryoEM Technology, Max Planck Institute of Biochemistry, Munich, Germany
| | - Cristina Capitanio
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Anna Bieber
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Annie Jen
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Nicole M Nightingale
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Benton J Anderson
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Chan Lee
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Joao A Paulo
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Ian R Smith
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jürgen M Plitzko
- CryoEM Technology, Max Planck Institute of Biochemistry, Munich, Germany
| | - Steven P Gygi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Brenda A Schulman
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Florian Wilfling
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Mechanisms of Cellular Quality Control, Max Planck Institute of Biophysics, Frankfurt, Germany
| | - Joshua J Coon
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
- Morgridge Institute for Research, Madison, WI 53715, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - J Wade Harper
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| |
Collapse
|
7
|
Lee Y, Hwang Y, Kim M, Jeon H, Joo S, Fang S, Kim JW. DGAT2 Plays a Crucial Role to Control ESRRA-PROX1 Transcriptional Network to Maintain Hepatic Mitochondrial Sustainability. Diabetes Metab J 2024; 48:901-914. [PMID: 38644620 PMCID: PMC11449812 DOI: 10.4093/dmj.2023.0368] [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: 10/13/2023] [Accepted: 12/11/2023] [Indexed: 04/23/2024] Open
Abstract
BACKGRUOUND Diacylglycerol O-acyltransferase 2 (DGAT2) synthesizes triacylglycerol (TG) from diacylglycerol; therefore, DGAT2 is considered as a therapeutic target for steatosis. However, the consequence of inhibiting DGAT2 is not fully investigated due to side effects including lethality and lipotoxicity. In this article, we observed the role of DGAT2 in hepatocarcinoma. METHODS The role of DGAT2 is analyzed via loss-of-function assay. DGAT2 knockdown (KD) and inhibitor treatment on HepG2 cell line was analyzed. Cumulative analysis of cell metabolism with bioinformatic data were assessed, and further compared with different cohorts of liver cancer patients and non-alcoholic fatty liver disease (NAFLD) patients to elucidate how DGAT2 is regulating cancer metabolism. RESULTS Mitochondrial function is suppressed in DGAT2 KD HepG2 cell along with the decreased lipid droplets. In the aspect of the cancer, DGAT2 KD upregulates cell proliferation. Analyzing transcriptome of NAFLD and hepatocellular carcinoma (HCC) patients highlights negatively correlating expression patterns of 73 lipid-associated genes including DGAT2. Cancer patients with the lower DGAT2 expression face lower survival rate. DGAT2 KD cell and patients' transcriptome show downregulation in estrogen- related receptor alpha (ESRRA) via integrated system for motif activity response analysis (ISMARA), with increased dimerization with corepressor prospero homeobox 1 (PROX1). CONCLUSION DGAT2 sustains the stability of mitochondria in hepatoma via suppressing ESRRA-PROX1 transcriptional network and hinders HCC from shifting towards glycolytic metabolism, which lowers cell proliferation.
Collapse
Affiliation(s)
- Yoseob Lee
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Korea
| | - Yeseong Hwang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Minki Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyeonuk Jeon
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seyeon Joo
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Sungsoon Fang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Korea
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jae-Woo Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Korea
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
8
|
Domingues N, Catarino S, Cristóvão B, Rodrigues L, Carvalho FA, Sarmento MJ, Zuzarte M, Almeida J, Ribeiro-Rodrigues T, Correia-Rodrigues Â, Fernandes F, Rodrigues-Santos P, Aasen T, Santos NC, Korolchuk VI, Gonçalves T, Milosevic I, Raimundo N, Girão H. Connexin43 promotes exocytosis of damaged lysosomes through actin remodelling. EMBO J 2024; 43:3627-3649. [PMID: 39044100 PMCID: PMC11377567 DOI: 10.1038/s44318-024-00177-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024] Open
Abstract
A robust and efficient cellular response to lysosomal membrane damage prevents leakage from the lysosome lumen into the cytoplasm. This response is understood to happen through either lysosomal membrane repair or lysophagy. Here we report exocytosis as a third response mechanism to lysosomal damage, which is further potentiated when membrane repair or lysosomal degradation mechanisms are impaired. We show that Connexin43 (Cx43), a protein canonically associated with gap junctions, is recruited from the plasma membrane to damaged lysosomes, promoting their secretion and accelerating cell recovery. The effects of Cx43 on lysosome exocytosis are mediated by a reorganization of the actin cytoskeleton that increases plasma membrane fluidity and decreases cell stiffness. Furthermore, we demonstrate that Cx43 interacts with the actin nucleator Arp2, the activity of which was shown to be necessary for Cx43-mediated actin rearrangement and lysosomal exocytosis following damage. These results define a novel mechanism of lysosomal quality control whereby Cx43-mediated actin remodelling potentiates the secretion of damaged lysosomes.
Collapse
Affiliation(s)
- Neuza Domingues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
| | - Steve Catarino
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Beatriz Cristóvão
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Lisa Rodrigues
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Filomena A Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Maria João Sarmento
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Mónica Zuzarte
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Jani Almeida
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Teresa Ribeiro-Rodrigues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Ânia Correia-Rodrigues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
| | - Fábio Fernandes
- Institute for Bioengineering and Biosciences (IBB) and Associate Laboratory i4HB-Institute for Health and Bioeconomy, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Paulo Rodrigues-Santos
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Trond Aasen
- Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Viktor I Korolchuk
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Teresa Gonçalves
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Univ Coimbra, Center for Neurosciences and Cell Biology (CNC), Coimbra, Portugal
| | - Ira Milosevic
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
- University of Oxford, Centre for Human Genetics, Nuffield Department of Medicine, Oxford, UK
| | - Nuno Raimundo
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Henrique Girão
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal.
- Univ Coimbra, Faculty of Medicine, Coimbra, Portugal.
- Univ Coimbra, Centre for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.
- Clinical and Academic Centre of Coimbra, Coimbra, Portugal.
| |
Collapse
|
9
|
Sayehmiri F, Motamedi F, Batool Z, Naderi N, Shaerzadeh F, Zoghi A, Rezaei O, Khodagholi F, Pourbadie HG. Mitochondrial plasticity and synaptic plasticity crosstalk; in health and Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14897. [PMID: 39097920 PMCID: PMC11298206 DOI: 10.1111/cns.14897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/19/2024] [Accepted: 07/18/2024] [Indexed: 08/06/2024] Open
Abstract
Synaptic plasticity is believed to underlie the cellular and molecular basis of memory formation. Mitochondria are one of the main organelles involved in metabolism and energy maintenance as plastic organelles that change morphologically and functionally in response to cellular needs and regulate synaptic function and plasticity through multiple mechanisms, including ATP generation, calcium homeostasis, and biogenesis. An increased neuronal activity enhances synaptic efficiency, during which mitochondria's spatial distribution and morphology change significantly. These organelles build up in the pre-and postsynaptic zones to produce ATP, which is necessary for several synaptic processes like neurotransmitter release and recycling. Mitochondria also regulate calcium homeostasis by buffering intracellular calcium, which ensures proper synaptic activity. Furthermore, mitochondria in the presynaptic terminal have distinct morphological properties compared to dendritic or postsynaptic mitochondria. This specialization enables precise control of synaptic activity and plasticity. Mitochondrial dysfunction has been linked to synaptic failure in many neurodegenerative disorders, like Alzheimer's disease (AD). In AD, malfunctioning mitochondria cause delays in synaptic vesicle release and recycling, ionic gradient imbalances, and mostly synaptic failure. This review emphasizes mitochondrial plasticity's contribution to synaptic function. It also explores the profound effect of mitochondrial malfunction on neurodegenerative disorders, focusing on AD, and provides an overview of how they sustain cellular health under normal conditions and how their malfunction contributes to neurodegenerative diseases, highlighting their potential as a therapeutic target for such conditions.
Collapse
Affiliation(s)
- Fatemeh Sayehmiri
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
- Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological SciencesUniversity of KarachiKarachiPakistan
| | - Nima Naderi
- Department of Pharmacology and Toxicology, Faculty of PharmacyShahid Beheshti University of Medical SciencesTehranIran
| | | | - Anahita Zoghi
- Department of Neurology, Loghman Hakim HospitalShahid Beheshti University of Medical SciencesTehranIran
| | - Omidvar Rezaei
- Skull Base Research CenterLoghman Hakim Hospital, Shahid Beheshti University of Medical SciencesTehranIran
| | - Fariba Khodagholi
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | | |
Collapse
|
10
|
Agostini F, Pereyra L, Dale J, Yambire KF, Maglioni S, Schiavi A, Ventura N, Milosevic I, Raimundo N. Upregulation of cholesterol synthesis by lysosomal defects requires a functional mitochondrial respiratory chain. J Biol Chem 2024; 300:107403. [PMID: 38782205 PMCID: PMC11254723 DOI: 10.1016/j.jbc.2024.107403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Mitochondria and lysosomes are two organelles that carry out both signaling and metabolic roles in cells. Recent evidence has shown that mitochondria and lysosomes are dependent on one another, as primary defects in one cause secondary defects in the other. Although there are functional impairments in both cases, the signaling consequences of primary mitochondrial dysfunction and lysosomal defects are dissimilar. Here, we used RNA sequencing to obtain transcriptomes from cells with primary mitochondrial or lysosomal defects to identify the global cellular consequences associated with mitochondrial or lysosomal dysfunction. We used these data to determine the pathways affected by defects in both organelles, which revealed a prominent role for the cholesterol synthesis pathway. We observed a transcriptional upregulation of this pathway in cellular and murine models of lysosomal defects, while it is transcriptionally downregulated in cellular and murine models of mitochondrial defects. We identified a role for the posttranscriptional regulation of transcription factor SREBF1, a master regulator of cholesterol and lipid biosynthesis, in models of mitochondrial respiratory chain deficiency. Furthermore, we found that retention of Ca2+ in lysosomes of cells with mitochondrial respiratory chain defects contributes to the differential regulation of the cholesterol synthesis pathway in the mitochondrial and lysosomal defects tested. Finally, we verified in vivo, using a model of mitochondria-associated disease in Caenorhabditis elegans that normalization of lysosomal Ca2+ levels results in partial rescue of the developmental delay induced by the respiratory chain deficiency.
Collapse
Affiliation(s)
- Francesco Agostini
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Leonardo Pereyra
- Department of Cellular Biochemistry, University Medical Center, Goettingen, Germany
| | - Justin Dale
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - King Faisal Yambire
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, New York, USA
| | - Silvia Maglioni
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany; Institute for Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Alfonso Schiavi
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Natascia Ventura
- IUF-Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany; Institute for Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University, Duesseldorf, Germany
| | - Ira Milosevic
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Multidisciplinary Institute for Ageing, University of Coimbra, Coimbra, Portugal
| | - Nuno Raimundo
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA; Penn State Cancer Institute, Penn State College of Medicine, Hershey, Pennsylvania, USA.
| |
Collapse
|
11
|
Domingues N, Calcagni’ A, Pires J, Freire SR, Herz NJ, Huynh T, Wieciorek K, Moreno MJ, Outeiro TF, Girão H, Milosevic I, Ballabio A, Raimundo N. Loss of the lysosomal protein CLN3 modifies the lipid content of the nuclear envelope leading to DNA damage and activation of YAP1 pro-apoptotic signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596474. [PMID: 38853929 PMCID: PMC11160784 DOI: 10.1101/2024.05.31.596474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Batten disease is characterized by early-onset blindness, juvenile dementia and death during the second decade of life. The most common genetic causes are mutations in the CLN3 gene encoding a lysosomal protein. There are currently no therapies targeting the progression of the disease, mostly due to the lack of knowledge about the disease mechanisms. To gain insight into the impact of CLN3 loss on cellular signaling and organelle function, we generated CLN3 knock-out cells in a human cell line (CLN3-KO), and performed RNA sequencing to obtain the cellular transcriptome. Following a multi-dimensional transcriptome analysis, we identified the transcriptional regulator YAP1 as a major driver of the transcriptional changes observed in CLN3-KO cells. We further observed that YAP1 pro-apoptotic signaling is hyperactive as a consequence of CLN3 functional loss in retinal pigment epithelia cells, and in the hippocampus and thalamus of CLN3exΔ7/8 mice, an established model of Batten disease. Loss of CLN3 activates YAP1 by a cascade of events that starts with the inability of releasing glycerophosphodiesthers from CLN3-KO lysosomes, which leads to perturbations in the lipid content of the nuclear envelope and nuclear dysmorphism. This results in increased number of DNA lesions, activating the kinase c-Abl, which phosphorylates YAP1, stimulating its pro-apoptotic signaling. Altogether, our results highlight a novel organelle crosstalk paradigm in which lysosomal metabolites regulate nuclear envelope content, nuclear shape and DNA homeostasis. This novel molecular mechanism underlying the loss of CLN3 in mammalian cells and tissues may open new c-Abl-centric therapeutic strategies to target Batten disease.
Collapse
Affiliation(s)
- Neuza Domingues
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
| | - Alessia Calcagni’
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Joana Pires
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
| | - Sofia Roque Freire
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
| | - Niculin Joachim Herz
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, USA
| | - Tuong Huynh
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, USA
| | - Katarzyna Wieciorek
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
| | - Maria João Moreno
- CQC-Biological Chemistry Group, Chemistry Department FCTUC, Coimbra, Portugal
| | - Tiago Fleming Outeiro
- University Medical Center Göttingen, Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Henrique Girão
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Centre for Innovative Biomedicine and Biotechnology, Academic and Clinical Center of Coimbra, Faculty of Medicine, University of Coimbra, Portugal
| | - Ira Milosevic
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, UK
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, USA
- SSM School for Advanced Studies, Federico II University, Naples, Italy
| | - Nuno Raimundo
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
- Penn State Cancer Institute, Hershey, PA, USA
| |
Collapse
|
12
|
Malara M, Prestel M, Tahirovic S. Endo-lysosomal dysfunction and neuronal-glial crosstalk in Niemann-Pick type C disease. Philos Trans R Soc Lond B Biol Sci 2024; 379:20220388. [PMID: 38368932 PMCID: PMC10874755 DOI: 10.1098/rstb.2022.0388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 11/27/2023] [Indexed: 02/20/2024] Open
Abstract
Niemann-Pick type C (NPC) disease is a rare progressive lysosomal lipid storage disorder that manifests with a heterogeneous spectrum of clinical syndromes, including visceral, neurological and psychiatric symptoms. This monogenetic autosomal recessive disease is largely caused by mutations in the NPC1 gene, which controls intracellular lipid homeostasis. Vesicle-mediated endo-lysosomal lipid trafficking and non-vesicular lipid exchange via inter-organelle membrane contact sites are both regulated by the NPC1 protein. Loss of NPC1 function therefore triggers intracellular accumulation of diverse lipid species, including cholesterol, glycosphingolipids, sphingomyelin and sphingosine. The NPC1-mediated dysfunction of lipid transport has severe consequences for all brain cells, leading to neurodegeneration. Besides the cell-autonomous contribution of neuronal NPC1, aberrant NPC1 signalling in other brain cells is critical for the pathology. We discuss here the importance of endo-lysosomal dysfunction and a tight crosstalk between neurons, oligodendrocytes, astrocytes and microglia in NPC pathology. We strongly believe that a cell-specific rescue may not be sufficient to counteract the severity of the NPC pathology, but targeting common mechanisms, such as endo-lysosomal and lipid trafficking dysfunction, may ameliorate NPC pathology. This article is part of a discussion meeting issue 'Understanding the endo-lysosomal network in neurodegeneration'.
Collapse
Affiliation(s)
- Mariagiovanna Malara
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Matthias Prestel
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| | - Sabina Tahirovic
- German Center for Neurodegenerative Diseases (DZNE) Munich, 81377 Munich, Germany
| |
Collapse
|
13
|
Sinha S, Hassan N, Schwartz RE. Organelle stress and alterations in interorganelle crosstalk during liver fibrosis. Hepatology 2024; 79:482-501. [PMID: 36626634 DOI: 10.1097/hep.0000000000000012] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/03/2022] [Indexed: 01/12/2023]
Abstract
The synchronous functioning and quality control of organelles ensure cell survival and function and are essential for maintaining homeostasis. Prolonged exposure to stressors (viruses, bacteria, parasitic infections, alcohol, drugs) or genetic mutations often disrupt the functional integrity of organelles which plays a critical role in the initiation and progression of several diseases including chronic liver diseases. One of the most important pathologic consequences of chronic liver diseases is liver fibrosis, characterized by tissue scarring due to the progressive accumulation of extracellular matrix components. Left untreated, fibrosis may advance to life-threatening complications such as cirrhosis, hepatic decompensation, and HCC, which collectively accounts for ∼1 million deaths per year worldwide. Owing to the lack of treatment options that can regress or reverse cirrhosis, liver transplantation is currently the only available treatment for end-stage liver disease. However, the limited supply of usable donor organs, adverse effects of lifelong immunosuppressive regimes, and financial considerations pose major challenges and limit its application. Hence, effective therapeutic strategies are urgently needed. An improved understanding of the organelle-level regulation of fibrosis can help devise effective antifibrotic therapies focused on reducing organelle stress, limiting organelle damage, improving interorganelle crosstalk, and restoring organelle homeostasis; and could be a potential clinical option to avoid transplantation. This review provides a timely update on the recent findings and mechanisms covering organelle-specific dysfunctions in liver fibrosis, highlights how correction of organelle functions opens new treatment avenues and discusses the potential challenges to clinical application.
Collapse
Affiliation(s)
- Saloni Sinha
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | | | | |
Collapse
|
14
|
Kumar S, Chhabra V, Shenoy S, Daksh R, Ravichandiran V, Swamy RS, Kumar N. Role of Flavonoids in Modulation of Mitochondria Dynamics during Oxidative Stress. Mini Rev Med Chem 2024; 24:908-919. [PMID: 37861054 DOI: 10.2174/0113895575259219230920093214] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/09/2023] [Accepted: 07/27/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Flavonoids are a widespread category of naturally occurring polyphenols distinguished by the flavan nucleus in plant-based foods and beverages, known for their various health benefits. Studies have suggested that consuming 150-500 mg of flavonoids daily is beneficial for health. Recent studies suggest that flavonoids are involved in maintaining mitochondrial activity and preventing impairment of mitochondrial dynamics by oxidative stress. OBJECTIVE This review emphasized the significance of studying the impact of flavonoids on mitochondrial dynamics, oxidative stress, and inflammatory response. METHODS This review analysed and summarised the findings related to the impact of flavonoids on mitochondria from publicly available search engines namely Pubmed, Scopus, and Web of Science. DESCRIPTION Any disruption in mitochondrial dynamics can contribute to cellular dysfunction and diseases, including cancer, cardiac conditions, and neurodegeneration. Flavonoids have been shown to modulate mitochondrial dynamics by regulating protein expression involved in fission and fusion events. Furthermore, flavonoids exhibit potent antioxidant properties by lowering the production of ROS and boosting the performance of antioxidant enzymes. Persistent inflammation is a characteristic of many different disorders. This is because flavonoids also alter the inflammatory response by controlling the expression of numerous cytokines and chemokines involved in the inflammatory process. Flavonoids exhibit an impressive array of significant health effects, making them an effective therapeutic agent for managing various disorders. Further this review summarised available mechanisms underlying flavonoids' actions on mitochondrial dynamics and oxidative stress to recognize the optimal dose and duration of flavonoid intake for therapeutic purposes. CONCLUSION This review may provide a solid foundation for developing targeted therapeutic interventions utilizing flavonoids, ultimately benefiting individuals afflicted with various disorders.
Collapse
Affiliation(s)
- Sachindra Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Industrial Area Hajipur, Vaishali, 844102, India
| | - Vishal Chhabra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Industrial Area Hajipur, Vaishali, 844102, India
| | - Smita Shenoy
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education (MAHE), Manipal, 576104, India
| | - Rajni Daksh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Industrial Area Hajipur, Vaishali, 844102, India
| | - Velayutham Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Industrial Area Hajipur, Vaishali, 844102, India
| | - Ravindra Shantakumar Swamy
- Division of Anatomy, Department of Basic Medical Sciences (DBMS), Manipal Academy of Higher Education (MAHE), Manipal, 576104, India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Industrial Area Hajipur, Vaishali, 844102, India
| |
Collapse
|
15
|
Boya P, Kaarniranta K, Handa JT, Sinha D. Lysosomes in retinal health and disease. Trends Neurosci 2023; 46:1067-1082. [PMID: 37848361 PMCID: PMC10842632 DOI: 10.1016/j.tins.2023.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/06/2023] [Accepted: 09/24/2023] [Indexed: 10/19/2023]
Abstract
Lysosomes play crucial roles in various cellular processes - including endocytosis, phagocytosis, and autophagy - which are vital for maintaining retinal health. Moreover, these organelles serve as environmental sensors and act as central hubs for multiple signaling pathways. Through communication with other cellular components, such as mitochondria, lysosomes orchestrate the cytoprotective response essential for preserving cellular homeostasis. This coordination is particularly critical in the retina, given its high metabolic rate and susceptibility to photo-oxidative stress. Consequently, impaired lysosomal function and dysregulated communication between lysosomes and other organelles contribute significantly to the pathobiology of major retinal degenerative diseases. This review explores the pivotal role of lysosomes in retinal cells and their involvement in retinal degenerative diseases.
Collapse
Affiliation(s)
- Patricia Boya
- Department of Neuroscience, University of Fribourg, Fribourg, Switzerland
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland, Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland; Department of Molecular Genetics, University of Lodz, Lodz, Poland
| | - James T Handa
- The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Debasish Sinha
- The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
16
|
Chen Z, Zeng S, Qian L. Quantitative Analysis of Mitochondrial RNA in Living Cells with a Dual-Color Imaging System. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301132. [PMID: 37127881 DOI: 10.1002/smll.202301132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/30/2023] [Indexed: 05/03/2023]
Abstract
Accurate quantification and dynamic expression profiling of mitochondrial RNA (mtRNA for short) are critical for illustrating their cellular functions. However, there lack methods for precise detection of mtRNA in situ due to the delivery restrictions and complicated cellular interferences. Herein, a dual-color imaging system featured with signal amplification and normalization capability for quantitative analysis of specific mtRNA is established. As a proof-of-concept example, an enzyme-free hairpin DNA cascade amplifier fine-tailored to specifically recognize mtRNA encoding NADH dehydrogenase subunit 6 (ND6) is employed as the signal output module and integrated into the biodegradable mitochondria-targeting black phosphorus nanosheet (BP-PEI-TPP) to monitor spatial-temporal dynamics of ND6 mtRNA. An internal reference module targeting β-actin mRNA is sent to the cytoplasm via BP-PEI for signal normalization, facilitating mtRNA quantification inside living cells with a degree of specificity and sensitivity as high as reverse transcription-quantitative polymerase chain reaction (RT-qPCR). With negligible cytotoxicity, this noninvasive "RT-qPCR mimic" can accurately indicate target mtRNA levels across different cells, providing a new strategy for precise analysis of subcellular RNAs in living systems.
Collapse
Affiliation(s)
- Zhiyan Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Linghui Qian
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center, & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310058, P. R. China
| |
Collapse
|
17
|
Kim S, Ochoa K, Melli SE, Yousufzai FAK, Barrera ZD, Williams AA, McIntyre G, Delgado E, Bolish JN, Macleod CM, Boghos M, Lens HP, Ramos AG, Wilson VB, Maloney K, Padron ZM, Khan AH, Blanco RE, Soto I. Disruptive lysosomal-metabolic signaling and neurodevelopmental deficits that precede Purkinje cell loss in a mouse model of Niemann-Pick Type-C disease. Sci Rep 2023; 13:5665. [PMID: 37024714 PMCID: PMC10079843 DOI: 10.1038/s41598-023-32971-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 04/05/2023] [Indexed: 04/08/2023] Open
Abstract
Purkinje cell (PC) loss occurs at an early age in patients and animal models of Niemann-Pick Type C (NPC), a lysosomal storage disease caused by mutations in the Npc1 or Npc2 genes. Although degeneration of PCs occurs early in NPC, little is known about how NPC1 deficiency affects the postnatal development of PCs. Using the Npc1nmf164 mouse model, we found that NPC1 deficiency significantly affected the postnatal development of PC dendrites and synapses. The developing dendrites of Npc1nmf164 PCs were significantly deficient in mitochondria and lysosomes. Furthermore, anabolic (mTORC1) and catabolic (TFEB) signaling pathways were not only perturbed but simultaneously activated in NPC1-deficient PCs, suggesting a loss of metabolic balance. We also found that mice with conditional heterozygous deletion of the Phosphatase and Tensin Homolog Deleted on Chromosome 10 gene (Pten-cHet), an inhibitor of mTORC1, showed similar early dendritic alterations in PCs to those found in Npc1-deficient mice. However, in contrast to Npc1nmf164 mice, Pten-cHet mice exhibited the overactivation of the mTORC1 pathway but with a strong inhibition of TFEB signaling, along with no dendritic mitochondrial reductions by the end of their postnatal development. Our data suggest that disruption of the lysosomal-metabolic signaling in PCs causes dendritic and synaptic developmental deficits that precede and promote their early degeneration in NPC.
Collapse
Affiliation(s)
- Sarah Kim
- Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, NJ, USA
| | - Kathleen Ochoa
- Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, NJ, USA
| | - Sierra E Melli
- Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, NJ, USA
| | - Fawad A K Yousufzai
- Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, NJ, USA
| | - Zerian D Barrera
- Department of Biological Science, Rowan University, Glassboro, NJ, USA
| | - Aela A Williams
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ, USA
| | - Gianna McIntyre
- Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, NJ, USA
| | - Esteban Delgado
- Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, NJ, USA
| | - James N Bolish
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ, USA
| | | | - Mary Boghos
- Department of Biology, Providence College, Providence, RI, USA
| | - Hayden P Lens
- Department of Biology, Providence College, Providence, RI, USA
| | - Alex G Ramos
- Department of Biology, Providence College, Providence, RI, USA
| | - Vincent B Wilson
- Department of Biological Science, Rowan University, Glassboro, NJ, USA
| | - Kelly Maloney
- Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, NJ, USA
| | - Zachary M Padron
- Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, NJ, USA
| | - Amaal H Khan
- Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, NJ, USA
| | - Rosa E Blanco
- The Institute of Neurobiology, University of Puerto Rico, San Juan, PR, USA
| | - Ileana Soto
- Department of Biology, Providence College, Providence, RI, USA.
| |
Collapse
|
18
|
Martins TS, Costa RS, Vilaça R, Lemos C, Teixeira V, Pereira C, Costa V. Iron Limitation Restores Autophagy and Increases Lifespan in the Yeast Model of Niemann-Pick Type C1. Int J Mol Sci 2023; 24:6221. [PMID: 37047194 PMCID: PMC10094029 DOI: 10.3390/ijms24076221] [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: 02/07/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Niemann-Pick type C1 (NPC1) is an endolysosomal transmembrane protein involved in the export of cholesterol and sphingolipids to other cellular compartments such as the endoplasmic reticulum and plasma membrane. NPC1 loss of function is the major cause of NPC disease, a rare lysosomal storage disorder characterized by an abnormal accumulation of lipids in the late endosomal/lysosomal network, mitochondrial dysfunction, and impaired autophagy. NPC phenotypes are conserved in yeast lacking Ncr1, an orthologue of human NPC1, leading to premature aging. Herein, we performed a phosphoproteomic analysis to investigate the effect of Ncr1 loss on cellular functions mediated by the yeast lysosome-like vacuoles. Our results revealed changes in vacuolar membrane proteins that are associated mostly with vesicle biology (fusion, transport, organization), autophagy, and ion homeostasis, including iron, manganese, and calcium. Consistently, the cytoplasm to vacuole targeting (Cvt) pathway was increased in ncr1∆ cells and autophagy was compromised despite TORC1 inhibition. Moreover, ncr1∆ cells exhibited iron overload mediated by the low-iron sensing transcription factor Aft1. Iron deprivation restored the autophagic flux of ncr1∆ cells and increased its chronological lifespan and oxidative stress resistance. These results implicate iron overload on autophagy impairment, oxidative stress sensitivity, and cell death in the yeast model of NPC1.
Collapse
Affiliation(s)
- Telma S. Martins
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Rafaela S. Costa
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Rita Vilaça
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Carolina Lemos
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Vitor Teixeira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Clara Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Vítor Costa
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| |
Collapse
|
19
|
Physiological roles of organelles at the pre-synapse in neurons. Int J Biochem Cell Biol 2023; 154:106345. [PMID: 36521722 DOI: 10.1016/j.biocel.2022.106345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Mitochondria, endoplasmic reticulum and lysosomes are involved in different pathways that can regulate pre-synaptic function. In particular, they could modulate ATP availability in response to rapid changes, could control synaptic protein levels and adjust Ca2+ signalling, which could all impact on neuronal activity. Organelles functions in these processes need to be considered alone when describing the impact of pre-synaptic organelles on neurotransmission. However, the interplay among organelles, which occurs either via signalling pathways or through physical membranous contacts, has to be considered. In this brief review, the physiological role of organelles localized at the pre-synapse in neuronal function is discussed.
Collapse
|
20
|
Chen FW, Davies JP, Calvo R, Chaudhari J, Dolios G, Taylor MK, Patnaik S, Dehdashti J, Mull R, Dranchack P, Wang A, Xu X, Hughes E, Southall N, Ferrer M, Wang R, Marugan JJ, Ioannou YA. Activation of mitochondrial TRAP1 stimulates mitochondria-lysosome crosstalk and correction of lysosomal dysfunction. iScience 2022; 25:104941. [PMID: 36065186 PMCID: PMC9440283 DOI: 10.1016/j.isci.2022.104941] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/27/2022] [Accepted: 08/11/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Fannie W. Chen
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joanna P. Davies
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Raul Calvo
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Jagruti Chaudhari
- Cell Therapy and Cell Engineering Facility, Memorial Sloan Kettering Cancer Center, 1250 1st Avenue, New York, NY 10065, USA
| | - Georgia Dolios
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mercedes K. Taylor
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA
| | - Samarjit Patnaik
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Jean Dehdashti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rebecca Mull
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Patricia Dranchack
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Amy Wang
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Xin Xu
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Emma Hughes
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Noel Southall
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Marc Ferrer
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Rong Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Juan J. Marugan
- Early Translation Branch, National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
- Corresponding author
| | - Yiannis A. Ioannou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Corresponding author
| |
Collapse
|
21
|
O’Neill KI, Kuo LW, Williams MM, Lind H, Crump LS, Hammond NG, Spoelstra NS, Caino MC, Richer JK. NPC1 Confers Metabolic Flexibility in Triple Negative Breast Cancer. Cancers (Basel) 2022; 14:3543. [PMID: 35884604 PMCID: PMC9319388 DOI: 10.3390/cancers14143543] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/07/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022] Open
Abstract
Triple-negative breast cancer (TNBC) often undergoes at least partial epithelial-to-mesenchymal transition (EMT) to facilitate metastasis. Identifying EMT-associated characteristics can reveal novel dependencies that may serve as therapeutic vulnerabilities in this aggressive breast cancer subtype. We found that NPC1, which encodes the lysosomal cholesterol transporter Niemann-Pick type C1 is highly expressed in TNBC as compared to estrogen receptor-positive (ER+) breast cancer, and is significantly elevated in high-grade disease. We demonstrated that NPC1 is directly targeted by microRNA-200c (miR-200c), a potent suppressor of EMT, providing a mechanism for its differential expression in breast cancer subtypes. The silencing of NPC1 in TNBC causes an accumulation of cholesterol-filled lysosomes, and drives decreased growth in soft agar and invasive capacity. Conversely, overexpression of NPC1 in an ER+ cell line increases invasion and growth in soft agar. We further identified TNBC cell lines as cholesterol auxotrophs, however, they do not solely depend on NPC1 for adequate cholesterol supply. The silencing of NPC1 in TNBC cell lines led to altered mitochondrial function and morphology, suppression of mTOR signaling, and accumulation of autophagosomes. A small molecule inhibitor of NPC1, U18666A, decreased TNBC proliferation and synergized with the chemotherapeutic drug, paclitaxel. This work suggests that NPC1 promotes aggressive characteristics in TNBC, and identifies NPC1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Kathleen I. O’Neill
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (K.I.O.); (L.-W.K.); (M.M.W.); (H.L.); (L.S.C.); (N.G.H.); (N.S.S.)
| | - Li-Wei Kuo
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (K.I.O.); (L.-W.K.); (M.M.W.); (H.L.); (L.S.C.); (N.G.H.); (N.S.S.)
| | - Michelle M. Williams
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (K.I.O.); (L.-W.K.); (M.M.W.); (H.L.); (L.S.C.); (N.G.H.); (N.S.S.)
| | - Hanne Lind
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (K.I.O.); (L.-W.K.); (M.M.W.); (H.L.); (L.S.C.); (N.G.H.); (N.S.S.)
| | - Lyndsey S. Crump
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (K.I.O.); (L.-W.K.); (M.M.W.); (H.L.); (L.S.C.); (N.G.H.); (N.S.S.)
| | - Nia G. Hammond
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (K.I.O.); (L.-W.K.); (M.M.W.); (H.L.); (L.S.C.); (N.G.H.); (N.S.S.)
| | - Nicole S. Spoelstra
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (K.I.O.); (L.-W.K.); (M.M.W.); (H.L.); (L.S.C.); (N.G.H.); (N.S.S.)
| | - M. Cecilia Caino
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Jennifer K. Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (K.I.O.); (L.-W.K.); (M.M.W.); (H.L.); (L.S.C.); (N.G.H.); (N.S.S.)
| |
Collapse
|
22
|
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
|
23
|
Tang H, Huang X, Pang S. Regulation of the lysosome by sphingolipids: potential role in aging. J Biol Chem 2022; 298:102118. [PMID: 35691340 PMCID: PMC9257404 DOI: 10.1016/j.jbc.2022.102118] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/01/2022] [Accepted: 06/04/2022] [Indexed: 02/04/2023] Open
Abstract
Sphingolipids are a class of bioactive complex lipids that have been closely associated with aging and aging-related diseases. However, the mechanism through which sphingolipids control aging has long been a mystery. Emerging studies reveal that sphingolipids exert tight control over lysosomal homeostasis and function, as evidenced by sphingolipid-related diseases, including but not limited to lysosomal storage disorders. These diseases are defined by primary lysosomal defects and a few secondary defects such as mitochondrial dysfunction. Intriguingly, recent research indicates that the majority of these defects are also associated with aging, implying that sphingolipid-related diseases and aging may share common mechanisms. We propose that the lysosome is a pivotal hub for sphingolipid-mediated aging regulation. This review discusses the critical roles of sphingolipid metabolism in regulating various lysosomal functions, with an emphasis on how such regulation may contribute to aging and aging-related diseases.
Collapse
Affiliation(s)
- Haiqing Tang
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| | - Xiaokun Huang
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| | - Shanshan Pang
- School of Life Sciences, Chongqing University, Chongqing, 401331, China.
| |
Collapse
|
24
|
Jain A, Zoncu R. Organelle transporters and inter-organelle communication as drivers of metabolic regulation and cellular homeostasis. Mol Metab 2022; 60:101481. [PMID: 35342037 PMCID: PMC9043965 DOI: 10.1016/j.molmet.2022.101481] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Spatial compartmentalization of metabolic pathways within membrane-separated organelles is key to the ability of eukaryotic cells to precisely regulate their biochemical functions. Membrane-bound organelles such as mitochondria, endoplasmic reticulum (ER) and lysosomes enable the concentration of metabolic precursors within optimized chemical environments, greatly accelerating the efficiency of both anabolic and catabolic reactions, enabling division of labor and optimal utilization of resources. However, metabolic compartmentalization also poses a challenge to cells because it creates spatial discontinuities that must be bridged for reaction cascades to be connected and completed. To do so, cells employ different methods to coordinate metabolic fluxes occurring in different organelles, such as membrane-localized transporters to facilitate regulated metabolite exchange between mitochondria and lysosomes, non-vesicular transport pathways via physical contact sites connecting the ER with both mitochondria and lysosomes, as well as localized regulatory signaling processes that coordinately regulate the activity of all these organelles. SCOPE OF REVIEW This review covers how cells use membrane transporters, membrane contact sites, and localized signaling pathways to mediate inter-organelle communication and coordinate metabolism. We also describe how disruption of inter-organelle communication is an emerging driver in a multitude of diseases, from cancer to neurodegeneration. MAJOR CONCLUSIONS Effective communication among organelles is essential to cellular health and function. Identifying the major molecular players involved in mediating metabolic coordination between organelles will further our understanding of cellular metabolism in health and lead us to design better therapeutics against dysregulated metabolism in disease.
Collapse
Affiliation(s)
- Aakriti Jain
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
25
|
Martelli F, Hernandes NH, Zuo Z, Wang J, Wong CO, Karagas NE, Roessner U, Rupasinghe T, Robin C, Venkatachalam K, Perry T, Batterham P, Bellen HJ. Low doses of the organic insecticide spinosad trigger lysosomal defects, elevated ROS, lipid dysregulation, and neurodegeneration in flies. eLife 2022; 11:e73812. [PMID: 35191376 PMCID: PMC8863376 DOI: 10.7554/elife.73812] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 01/28/2022] [Indexed: 12/14/2022] Open
Abstract
Large-scale insecticide application is a primary weapon in the control of insect pests in agriculture. However, a growing body of evidence indicates that it is contributing to the global decline in population sizes of many beneficial insect species. Spinosad emerged as an organic alternative to synthetic insecticides and is considered less harmful to beneficial insects, yet its mode of action remains unclear. Using Drosophila, we show that low doses of spinosad antagonize its neuronal target, the nicotinic acetylcholine receptor subunit alpha 6 (nAChRα6), reducing the cholinergic response. We show that the nAChRα6 receptors are transported to lysosomes that become enlarged and increase in number upon low doses of spinosad treatment. Lysosomal dysfunction is associated with mitochondrial stress and elevated levels of reactive oxygen species (ROS) in the central nervous system where nAChRα6 is broadly expressed. ROS disturb lipid storage in metabolic tissues in an nAChRα6-dependent manner. Spinosad toxicity is ameliorated with the antioxidant N-acetylcysteine amide. Chronic exposure of adult virgin females to low doses of spinosad leads to mitochondrial defects, severe neurodegeneration, and blindness. These deleterious effects of low-dose exposures warrant rigorous investigation of its impacts on beneficial insects.
Collapse
Affiliation(s)
- Felipe Martelli
- School of BioSciences, The University of MelbourneMelbourneAustralia
| | | | - Zhongyuan Zuo
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Julia Wang
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Ching-On Wong
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences CenterHoustonUnited States
| | - Nicholas E Karagas
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences CenterHoustonUnited States
| | - Ute Roessner
- School of BioSciences, The University of MelbourneMelbourneAustralia
| | | | - Charles Robin
- School of BioSciences, The University of MelbourneMelbourneAustralia
| | - Kartik Venkatachalam
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Sciences CenterHoustonUnited States
| | - Trent Perry
- School of BioSciences, The University of MelbourneMelbourneAustralia
| | - Philip Batterham
- School of BioSciences, The University of MelbourneMelbourneAustralia
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Neurological Research Institute, Texas Children HospitalHoustonUnited States
- Howard Hughes Medical Institute, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
26
|
Zhao M, Lian A, Zhong L, Guo R. The regulatory mechanism between lysosomes and mitochondria in the aetiology of cardiovascular diseases. Acta Physiol (Oxf) 2022; 234:e13757. [PMID: 34978753 DOI: 10.1111/apha.13757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/16/2021] [Accepted: 01/01/2022] [Indexed: 11/28/2022]
Abstract
Coordinated action among various organelles maintains cellular functions. For instance, mitochondria and lysosomes are the main organelles contributing to cellular metabolism and provide energy for cardiomyocyte contraction. They also provide essential signalling platforms in the cell that regulate many key processes such as autophagy, apoptosis, oxidative stress, inflammation and cell death. Often, abnormalities in mitochondrial or lysosomal structures and functions bring about cardiovascular diseases (CVDs). Although the communication between mitochondria and lysosomes throughout the cardiovascular system is intensely studied, the regulatory mechanisms have not been completely understood. Thus, we summarize the most recent studies related to mitochondria and lysosomes' role in CVDs and their potential connections and communications under cardiac pathophysiological conditions. Further, we discuss limitations and future perspectives regarding diagnosis, therapeutic strategies and drug discovery in CVDs.
Collapse
Affiliation(s)
- Mengxue Zhao
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
| | - Andrew Lian
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona California USA
| | - Li Zhong
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
- College of Osteopathic Medicine of the Pacific Western University of Health Sciences Pomona California USA
| | - Rui Guo
- College of Life Sciences Institute of Life Science and Green Development Hebei University Baoding China
- The Key Laboratory of Zoological Systematics and Application College of Life Sciences Hebei University Baoding China
| |
Collapse
|
27
|
Dudek J, Maack C. Mechano-energetic aspects of Barth syndrome. J Inherit Metab Dis 2022; 45:82-98. [PMID: 34423473 DOI: 10.1002/jimd.12427] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/28/2021] [Accepted: 08/19/2021] [Indexed: 12/22/2022]
Abstract
Energy-demanding organs like the heart are strongly dependent on oxidative phosphorylation in mitochondria. Oxidative phosphorylation is governed by the respiratory chain located in the inner mitochondrial membrane. The inner mitochondrial membrane is the only cellular membrane with significant amounts of the phospholipid cardiolipin, and cardiolipin was found to directly interact with a number of essential protein complexes, including respiratory chain complexes I to V. An inherited defect in the biogenesis of cardiolipin causes Barth syndrome, which is associated with cardiomyopathy, skeletal myopathy, neutropenia and growth retardation. Energy conversion is dependent on reducing equivalents, which are replenished by oxidative metabolism in the Krebs cycle. Cardiolipin deficiency in Barth syndrome also affects Krebs cycle activity, metabolite transport and mitochondrial morphology. During excitation-contraction coupling, calcium (Ca2+ ) released from the sarcoplasmic reticulum drives sarcomeric contraction. At the same time, Ca2+ influx into mitochondria drives the activation of Krebs cycle dehydrogenases and the regeneration of reducing equivalents. Reducing equivalents are essential not only for energy conversion, but also for maintaining a redox buffer, which is required to detoxify reactive oxygen species (ROS). Defects in CL may also affect Ca2+ uptake into mitochondria and thereby hamper energy supply and demand matching, but also detoxification of ROS. Here, we review the impact of cardiolipin deficiency on mitochondrial function in Barth syndrome and discuss potential therapeutic strategies.
Collapse
Affiliation(s)
- Jan Dudek
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
| |
Collapse
|
28
|
Zhao R, Zhao X, Wang X, Liu Y, Yang J, Jiang S, Zhou X, Jiao B, Zhang L, Liu Y, Yu Z. Fasting promotes acute hypoxic adaptation by suppressing mTOR-mediated pathways. Cell Death Dis 2021; 12:1045. [PMID: 34732698 PMCID: PMC8566556 DOI: 10.1038/s41419-021-04351-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 11/09/2022]
Abstract
Rapid adaptation to a hypoxic environment is an unanswered question that we are committed to exploring. At present, there is no suitable strategy to achieve rapid hypoxic adaptation. Here, we demonstrate that fasting preconditioning for 72 h reduces tissue injuries and maintains cardiac function, consequently significantly improving the survival rates of rats under extreme hypoxia, and this strategy can be used for rapid hypoxic adaptation. Mechanistically, fasting reduces blood glucose and further suppresses tissue mTOR activity. On the one hand, fasting-induced mTOR inhibition reduces unnecessary ATP consumption and increases ATP reserves under acute hypoxia as a result of decreased protein synthesis and lipogenesis; on the other hand, fasting-induced mTOR inhibition improves mitochondrial oxygen utilization efficiency to ensure ATP production under acute hypoxia, which is due to the significant decrease in ROS generation induced by enhanced mitophagy. Our findings highlight the important role of mTOR in acute hypoxic adaptation, and targeted regulation of mTOR could be a new strategy to improve acute hypoxic tolerance in the body.
Collapse
Affiliation(s)
- Ruzhou Zhao
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Xingcheng Zhao
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Xiaobo Wang
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Yanqi Liu
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Jie Yang
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Shuai Jiang
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Xiang Zhou
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
- Department of Nuclear Medicine, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Bo Jiao
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Lin Zhang
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China
| | - Yong Liu
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China.
| | - Zhibin Yu
- Department of Aerospace Physiology, Air Force Medical University, Xi'an, China.
| |
Collapse
|
29
|
In situ observation of mitochondrial biogenesis as the early event of apoptosis. iScience 2021; 24:103038. [PMID: 34553131 PMCID: PMC8441175 DOI: 10.1016/j.isci.2021.103038] [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: 05/06/2021] [Revised: 07/28/2021] [Accepted: 08/22/2021] [Indexed: 12/25/2022] Open
Abstract
Mitochondrial biogenesis is a cell response to external stimuli which is generally believed to suppress apoptosis. However, during the process of apoptosis, whether mitochondrial biogenesis occurs in the early stage of the apoptotic cells remains unclear. To address this question, we constructed the COX8-EGFP-ACTIN-mCherry HeLa cells with recombinant fluorescent proteins respectively tagged on the nucleus and mitochondria and monitored the mitochondrial changes in the living cells exposed to gamma-ray radiation. Besides in situ detection of mitochondrial fluorescence changes, we also examined the cell viability, nuclear DNA damage, reactive oxygen species (ROS), mitochondrial superoxide, citrate synthase activity, ATP, cytoplasmic and mitochondrial calcium, mitochondrial mass, mitochondrial morphology, and protein expression related to mitochondrial biogenesis, as well as the apoptosis biomarkers. As a result, we confirmed that significant mitochondrial biogenesis took place preceding the radiation-induced apoptosis, and it was closely correlated with the apoptotic cells at late stage. The involved mechanism was also discussed. Dual fluorescence approach was used for in situ observation of living cell processes Radiation-induced effects of mitochondrial biogenesis and apoptosis were observed Relationship between mitochondrial biogenesis and apoptosis was revisited Assessing early mitochondrial biogenesis is critical for predicting later fate of cells
Collapse
|
30
|
Han S, Ren M, Kuang T, Pang M, Guan D, Liu Y, Wang Y, Zhang W, Ye Z. Cerebellar Long Noncoding RNA Expression Profile in a Niemann-Pick C Disease Mouse Model. Mol Neurobiol 2021; 58:5826-5836. [PMID: 34410604 PMCID: PMC8599378 DOI: 10.1007/s12035-021-02526-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/05/2021] [Indexed: 02/08/2023]
Abstract
Niemann-Pick type C (NP-C) disease is a neurodegenerative lysosomal storage disorder primarily caused by mutations in NPC1. However, its pathogenesis remains poorly understood. While mounting evidence has demonstrated the involvement of long noncoding RNAs (lncRNAs) in the pathogenesis of neurodegenerative disorders, the lncRNA expression profile in NP-C has not been determined. Here, we used RNA-seq analysis to determine lncRNA and mRNA expression profiles of the cerebella of NPC1−/− mice. We found that 272 lncRNAs and 856 mRNAs were significantly dysregulated in NPC1−/− mice relative to controls (≥ 2.0-fold, p < 0.05). Quantitative real-time PCR (qRT‐PCR) was utilized to validate the expression of selected lncRNAs and mRNAs. Next, a lncRNA-mRNA coexpression network was employed to examine the potential roles of the differentially expressed (DE) lncRNAs. Functional analysis revealed that mRNAs coexpressed with lncRNAs are mainly linked to immune system–related processes and neuroinflammation. Moreover, knockdown of the lncRNA H19 ameliorated changes in ROS levels and cell viability and suppressed the lipopolysaccharide (LPS)–induced inflammatory response in vitro. Our findings indicate that dysregulated lncRNA expression patterns are associated with NP-C pathogenesis and offer insight into the development of novel therapeutics based on lncRNAs.
Collapse
Affiliation(s)
- Shiqian Han
- Department of Tropical Medicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Meng Ren
- Laboratory Animal Research Center, Chongqing University School of Medicine, Chongqing, 400044, China
| | - Tianyin Kuang
- Laboratory Animal Research Center, Chongqing University School of Medicine, Chongqing, 400044, China
| | - Mao Pang
- Laboratory Animal Research Center, Chongqing University School of Medicine, Chongqing, 400044, China
| | - Dongwei Guan
- Laboratory Animal Research Center, Chongqing University School of Medicine, Chongqing, 400044, China
| | - Yesong Liu
- Cornell University, Ithaca, NY, 14853, USA
| | - Yong Wang
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Wengeng Zhang
- Precision Medicine Key Laboratory of Sichuan Province and Precision Medicine Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhijia Ye
- Department of Tropical Medicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China. .,Laboratory Animal Research Center, Chongqing University School of Medicine, Chongqing, 400044, China.
| |
Collapse
|
31
|
Wang YP, Sharda A, Xu SN, van Gastel N, Man CH, Choi U, Leong WZ, Li X, Scadden DT. Malic enzyme 2 connects the Krebs cycle intermediate fumarate to mitochondrial biogenesis. Cell Metab 2021; 33:1027-1041.e8. [PMID: 33770508 PMCID: PMC10472834 DOI: 10.1016/j.cmet.2021.03.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/21/2020] [Accepted: 03/03/2021] [Indexed: 12/13/2022]
Abstract
Mitochondria have an independent genome (mtDNA) and protein synthesis machinery that coordinately activate for mitochondrial generation. Here, we report that the Krebs cycle intermediate fumarate links metabolism to mitobiogenesis through binding to malic enzyme 2 (ME2). Mechanistically, fumarate binds ME2 with two complementary consequences. First, promoting the formation of ME2 dimers, which activate deoxyuridine 5'-triphosphate nucleotidohydrolase (DUT). DUT fosters thymidine generation and an increase of mtDNA. Second, fumarate-induced ME2 dimers abrogate ME2 monomer binding to mitochondrial ribosome protein L45, freeing it for mitoribosome assembly and mtDNA-encoded protein production. Methylation of the ME2-fumarate binding site by protein arginine methyltransferase-1 inhibits fumarate signaling to constrain mitobiogenesis. Notably, acute myeloid leukemia is highly dependent on mitochondrial function and is sensitive to targeting of the fumarate-ME2 axis. Therefore, mitobiogenesis can be manipulated in normal and malignant cells through ME2, an unanticipated governor of mitochondrial biomass production that senses nutrient availability through fumarate.
Collapse
Affiliation(s)
- Yi-Ping Wang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College, Fudan University, Shanghai 20032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 20032, China
| | - Azeem Sharda
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Shuang-Nian Xu
- Department of Hematology, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - Nick van Gastel
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Cheuk Him Man
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Una Choi
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Wei Zhong Leong
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Xi Li
- Department of Hematology, Southwest Hospital, Army Medical University, Chongqing 400038, China
| | - David T Scadden
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
32
|
Xu F, Li Y, Cao Z, Zhang J, Huang W. AFB 1-induced mice liver injury involves mitochondrial dysfunction mediated by mitochondrial biogenesis inhibition. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 216:112213. [PMID: 33838459 DOI: 10.1016/j.ecoenv.2021.112213] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 03/24/2021] [Accepted: 03/29/2021] [Indexed: 06/12/2023]
Abstract
Aflatoxin B1 (AFB1) pollutes foodstuffs and feeds, causing a food safety problem and seriously endangering human and animal health. Liver is the principal organ for AFB1 accumulation and biotransformation, during which AFB1 can cause acute and chronic liver damage, however, the specific mechanism is not completely clear. Mitochondria are the primary organelle of cellular bio-oxidation, providing 95% energy for liver to execute its multiple functions. Therefore, we speculated that mitochondrial dysfunction is involved in AFB1-induced liver injury. To verify the hypothesis, a total of eighty healthy male mice were randomly divided into four groups on average, and exposed with 0, 0.375, 0.75 and 1.5 mg/kg body weight AFB1 by intragastric administration for 30 d. The results displayed that AFB1 triggered liver injury accompanied by oxidative stress. AFB1 exposure also damaged mitochondria structure, decreased mitochondrial membrane potential (MMP), as well as increased cytoplasmic cytochrome c (Cyt-c) protein expression, Bax, p53, Caspase-3/9 protein and/or mRNA expression levels and terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine-5'-triphosphate (dUTP) nick end labeling (TUNEL) staining positive cells in mice liver. Meanwhile, AFB1 exposure elevated pyruvate content, inhibited tricarboxylic acid (TCA) cycle rate-limiting enzymes and electron transport chain (ETC) complexes I-V activities, disturbed ETC complexes I-V subunits mRNA expression levels and reduced adenosine triphosphate (ATP) level in mice liver. These results indicated that AFB1 destroyed mitochondrial structure, activated mitochondrion-dependent apoptosis and induced mitochondrial dysfunction. In addition, AFB1 disrupted mitochondrial biogenesis, presented as the abnormalities of protein and/or gene expression levels of voltage dependent anion channel protein 1 (VDAC1), peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), nuclear respiratory factor 1 (Nrf1) and mitochondrial transcription factor A (Tfam). This may contribute to hepatic and mitochondrial lesions induced by AFB1. These results provide a new perspective for elucidating the mechanisms of AFB1 hepatotoxicity.
Collapse
Affiliation(s)
- Feibo Xu
- Department of Histology and Embryology, College of Basic Medicine, Binzhou Medical University, 346 Guanhai Road, Yantai 246003, Shandong, China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| | - Yanfei Li
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zheng Cao
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jian Zhang
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Wanyue Huang
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
33
|
Yuan L, Yuan Y, Liu F, Li L, Liu J, Chen Y, Cheng J, Lu Y. PGC-1α alleviates mitochondrial dysfunction via TFEB-mediated autophagy in cisplatin-induced acute kidney injury. Aging (Albany NY) 2021; 13:8421-8439. [PMID: 33714196 PMCID: PMC8034953 DOI: 10.18632/aging.202653] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/19/2020] [Indexed: 02/05/2023]
Abstract
Because of the key role of impaired mitochondria in the progression of acute kidney injury (AKI), it is striking that peroxisome proliferator γ coactivator 1-α (PGC-1α), a transcriptional coactivator of genes involved in mitochondrial biogenesis and autophagy, protects from kidney injury. However, the specific mechanism involved in PGC-1α-mediated autophagy remains elusive. In vivo, along with the severe kidney damage, the expression of PGC-1α was decreased in cisplatin-induced AKI mice. Conversely, PGC-1α activator (ZLN005) administration could alleviate kidney injury. Consistently, in vitro overexpression of PGC-1α or ZLN005 treatment inhibited cell apoptosis and mitochondrial dysfunction induced by cisplatin. Moreover, ZLN005 treatment increased the expression of LC3-II and co-localization between LC3 and mitochondria, suggesting that the mitophagy was activated. Furthermore, PGC-1α-mediated the activation of mitophagy was reliant on the increased expression of TFEB, and the protective effects were abrogated in TFEB-knockdown cells. These data suggest that the activation of PGC-1α could alleviate mitochondrial dysfunction and kidney injury in AKI mice via TFEB-mediated autophagy.
Collapse
Affiliation(s)
- Longhui Yuan
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yujia Yuan
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Fei Liu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Lan Li
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jingping Liu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Younan Chen
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jingqiu Cheng
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, Department of Nephrology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Loss of NPC1 enhances phagocytic uptake and impairs lipid trafficking in microglia. Nat Commun 2021; 12:1158. [PMID: 33627648 PMCID: PMC7904859 DOI: 10.1038/s41467-021-21428-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 01/27/2021] [Indexed: 02/08/2023] Open
Abstract
Niemann-Pick type C disease is a rare neurodegenerative disorder mainly caused by mutations in NPC1, resulting in abnormal late endosomal/lysosomal lipid storage. Although microgliosis is a prominent pathological feature, direct consequences of NPC1 loss on microglial function remain not fully characterized. We discovered pathological proteomic signatures and phenotypes in NPC1-deficient murine models and demonstrate a cell autonomous function of NPC1 in microglia. Loss of NPC1 triggers enhanced phagocytic uptake and impaired myelin turnover in microglia that precede neuronal death. Npc1−/− microglia feature a striking accumulation of multivesicular bodies and impaired trafficking of lipids to lysosomes while lysosomal degradation function remains preserved. Molecular and functional defects were also detected in blood-derived macrophages of NPC patients that provide a potential tool for monitoring disease. Our study underscores an essential cell autonomous role for NPC1 in immune cells and implies microglial therapeutic potential. Niemann-Pick type C disease is a rare childhood neurodegenerative disorder predominantly caused by mutations in NPC1, resulting in abnormal late endosomal and lysosomal defects. Here the authors show that NPC1 disruption largely impairs microglial function.
Collapse
|
35
|
Bennett JP, Onyango IG. Energy, Entropy and Quantum Tunneling of Protons and Electrons in Brain Mitochondria: Relation to Mitochondrial Impairment in Aging-Related Human Brain Diseases and Therapeutic Measures. Biomedicines 2021; 9:225. [PMID: 33671585 PMCID: PMC7927033 DOI: 10.3390/biomedicines9020225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 11/16/2022] Open
Abstract
Adult human brains consume a disproportionate amount of energy substrates (2-3% of body weight; 20-25% of total glucose and oxygen). Adenosine triphosphate (ATP) is a universal energy currency in brains and is produced by oxidative phosphorylation (OXPHOS) using ATP synthase, a nano-rotor powered by the proton gradient generated from proton-coupled electron transfer (PCET) in the multi-complex electron transport chain (ETC). ETC catalysis rates are reduced in brains from humans with neurodegenerative diseases (NDDs). Declines of ETC function in NDDs may result from combinations of nitrative stress (NS)-oxidative stress (OS) damage; mitochondrial and/or nuclear genomic mutations of ETC/OXPHOS genes; epigenetic modifications of ETC/OXPHOS genes; or defects in importation or assembly of ETC/OXPHOS proteins or complexes, respectively; or alterations in mitochondrial dynamics (fusion, fission, mitophagy). Substantial free energy is gained by direct O2-mediated oxidation of NADH. Traditional ETC mechanisms require separation between O2 and electrons flowing from NADH/FADH2 through the ETC. Quantum tunneling of electrons and much larger protons may facilitate this separation. Neuronal death may be viewed as a local increase in entropy requiring constant energy input to avoid. The ATP requirement of the brain may partially be used for avoidance of local entropy increase. Mitochondrial therapeutics seeks to correct deficiencies in ETC and OXPHOS.
Collapse
Affiliation(s)
| | - Isaac G. Onyango
- International Clinical Research Center, St. Anne’s University Hospital, CZ-65691 Brno, Czech Republic;
| |
Collapse
|
36
|
Kuk MU, Lee YH, Kim JW, Hwang SY, Park JT, Park SC. Potential Treatment of Lysosomal Storage Disease through Modulation of the Mitochondrial-Lysosomal Axis. Cells 2021; 10:cells10020420. [PMID: 33671306 PMCID: PMC7921977 DOI: 10.3390/cells10020420] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/13/2021] [Accepted: 02/14/2021] [Indexed: 12/28/2022] Open
Abstract
Lysosomal storage disease (LSD) is an inherited metabolic disorder caused by enzyme deficiency in lysosomes. Some treatments for LSD can slow progression, but there are no effective treatments to restore the pathological phenotype to normal levels. Lysosomes and mitochondria interact with each other, and this crosstalk plays a role in the maintenance of cellular homeostasis. Deficiency of lysosome enzymes in LSD impairs the turnover of mitochondrial defects, leading to deterioration of the mitochondrial respiratory chain (MRC). Cells with MRC impairment are associated with reduced lysosomal calcium homeostasis, resulting in impaired autophagic and endolysosomal function. This malicious feedback loop between lysosomes and mitochondria exacerbates LSD. In this review, we assess the interactions between mitochondria and lysosomes and propose the mitochondrial-lysosomal axis as a research target to treat LSD. The importance of the mitochondrial-lysosomal axis has been systematically characterized in several studies, suggesting that proper regulation of this axis represents an important investigative guide for the development of therapeutics for LSD. Therefore, studying the mitochondrial-lysosomal axis will not only add knowledge of the essential physiological processes of LSD, but also provide new strategies for treatment of LSD.
Collapse
Affiliation(s)
- Myeong Uk Kuk
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (M.U.K.); (Y.H.L.); (J.W.K.); (S.Y.H.)
| | - Yun Haeng Lee
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (M.U.K.); (Y.H.L.); (J.W.K.); (S.Y.H.)
| | - Jae Won Kim
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (M.U.K.); (Y.H.L.); (J.W.K.); (S.Y.H.)
| | - Su Young Hwang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (M.U.K.); (Y.H.L.); (J.W.K.); (S.Y.H.)
| | - Joon Tae Park
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea; (M.U.K.); (Y.H.L.); (J.W.K.); (S.Y.H.)
- Correspondence: (J.T.P.); ; (S.C.P.); Tel.: +82-32-835-8841 (J.T.P.); +82-10-5495-9200 (S.C.P.)
| | - Sang Chul Park
- The Future Life & Society Research Center, Chonnam National University, Gwangju 61186, Korea
- Correspondence: (J.T.P.); ; (S.C.P.); Tel.: +82-32-835-8841 (J.T.P.); +82-10-5495-9200 (S.C.P.)
| |
Collapse
|
37
|
Davis OB, Shin HR, Lim CY, Wu EY, Kukurugya M, Maher CF, Perera RM, Ordonez MP, Zoncu R. NPC1-mTORC1 Signaling Couples Cholesterol Sensing to Organelle Homeostasis and Is a Targetable Pathway in Niemann-Pick Type C. Dev Cell 2020; 56:260-276.e7. [PMID: 33308480 DOI: 10.1016/j.devcel.2020.11.016] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/05/2020] [Accepted: 11/17/2020] [Indexed: 01/22/2023]
Abstract
Lysosomes promote cellular homeostasis through macromolecular hydrolysis within their lumen and metabolic signaling by the mTORC1 kinase on their limiting membranes. Both hydrolytic and signaling functions require precise regulation of lysosomal cholesterol content. In Niemann-Pick type C (NPC), loss of the cholesterol exporter, NPC1, causes cholesterol accumulation within lysosomes, leading to mTORC1 hyperactivation, disrupted mitochondrial function, and neurodegeneration. The compositional and functional alterations in NPC lysosomes and nature of aberrant cholesterol-mTORC1 signaling contribution to organelle pathogenesis are not understood. Through proteomic profiling of NPC lysosomes, we find pronounced proteolytic impairment compounded with hydrolase depletion, enhanced membrane damage, and defective mitophagy. Genetic and pharmacologic mTORC1 inhibition restores lysosomal proteolysis without correcting cholesterol storage, implicating aberrant mTORC1 as a pathogenic driver downstream of cholesterol accumulation. Consistently, mTORC1 inhibition ameliorates mitochondrial dysfunction in a neuronal model of NPC. Thus, cholesterol-mTORC1 signaling controls organelle homeostasis and is a targetable pathway in NPC.
Collapse
Affiliation(s)
- Oliver B Davis
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hijai R Shin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chun-Yan Lim
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Emma Y Wu
- Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Department of Pediatric Gastroenterology, Hepatology, and Nutrition, University of California, San Diego, La Jolla, CA 92037, USA
| | - Matthew Kukurugya
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Claire F Maher
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA
| | - Rushika M Perera
- Department of Anatomy and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - M Paulina Ordonez
- Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Department of Pediatric Gastroenterology, Hepatology, and Nutrition, University of California, San Diego, La Jolla, CA 92037, USA.
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
38
|
Cavinato M, Madreiter-Sokolowski CT, Büttner S, Schosserer M, Zwerschke W, Wedel S, Grillari J, Graier WF, Jansen-Dürr P. Targeting cellular senescence based on interorganelle communication, multilevel proteostasis, and metabolic control. FEBS J 2020; 288:3834-3854. [PMID: 33200494 PMCID: PMC7611050 DOI: 10.1111/febs.15631] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/02/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023]
Abstract
Cellular senescence, a stable cell division arrest caused by severe damage and stress, is a hallmark of aging in vertebrates including humans. With progressing age, senescent cells accumulate in a variety of mammalian tissues, where they contribute to tissue aging, identifying cellular senescence as a major target to delay or prevent aging. There is an increasing demand for the discovery of new classes of small molecules that would either avoid or postpone cellular senescence by selectively eliminating senescent cells from the body (i.e., ‘senolytics’) or inactivating/switching damage‐inducing properties of senescent cells (i.e., ‘senostatics/senomorphics’), such as the senescence‐associated secretory phenotype. Whereas compounds with senolytic or senostatic activity have already been described, their efficacy and specificity has not been fully established for clinical use yet. Here, we review mechanisms of senescence that are related to mitochondria and their interorganelle communication, and the involvement of proteostasis networks and metabolic control in the senescent phenotype. These cellular functions are associated with cellular senescence in in vitro and in vivo models but have not been fully exploited for the search of new compounds to counteract senescence yet. Therefore, we explore possibilities to target these mechanisms as new opportunities to selectively eliminate and/or disable senescent cells with the aim of tissue rejuvenation. We assume that this research will provide new compounds from the chemical space which act as mimetics of caloric restriction, modulators of calcium signaling and mitochondrial physiology, or as proteostasis optimizers, bearing the potential to counteract cellular senescence, thereby allowing healthy aging.
Collapse
Affiliation(s)
- Maria Cavinato
- Institute for Biomedical Aging Research, Leopold-Franzens Universität Innsbruck, Austria.,Center for Molecular Biosciences Innsbruck (CMBI), Leopold-Franzens Universität Innsbruck, Austria
| | - Corina T Madreiter-Sokolowski
- Department of Health Sciences and Technology, Institute of Translational Medicine, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland.,Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Sabrina Büttner
- Institute of Molecular Biosciences, University of Graz, Austria.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Sweden
| | - Markus Schosserer
- Christian Doppler Laboratory for Skin Multimodal Analytical Imaging of Aging and Senescence, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Medical University of Vienna, Austria
| | - Werner Zwerschke
- Institute for Biomedical Aging Research, Leopold-Franzens Universität Innsbruck, Austria.,Center for Molecular Biosciences Innsbruck (CMBI), Leopold-Franzens Universität Innsbruck, Austria
| | - Sophia Wedel
- Institute for Biomedical Aging Research, Leopold-Franzens Universität Innsbruck, Austria.,Center for Molecular Biosciences Innsbruck (CMBI), Leopold-Franzens Universität Innsbruck, Austria
| | - Johannes Grillari
- Christian Doppler Laboratory for Skin Multimodal Analytical Imaging of Aging and Senescence, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Medical University of Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Wolfgang F Graier
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Austria.,BioTechMed Graz, Austria
| | - Pidder Jansen-Dürr
- Institute for Biomedical Aging Research, Leopold-Franzens Universität Innsbruck, Austria.,Center for Molecular Biosciences Innsbruck (CMBI), Leopold-Franzens Universität Innsbruck, Austria
| |
Collapse
|
39
|
Turton N, Rutherford T, Thijssen D, Hargreaves IP. Putative adjunct therapies to target mitochondrial dysfunction and oxidative stress in phenylketonuria, lysosomal storage disorders and peroxisomal disorders. Expert Opin Orphan Drugs 2020. [DOI: 10.1080/21678707.2020.1850254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Affiliation(s)
- Nadia Turton
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Tricia Rutherford
- Department of research and development, Vitaflo International Ltd, Liverpool, UK
| | - Dick Thijssen
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Iain P Hargreaves
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
40
|
Celecoxib promotes survival and upregulates the expression of neuroprotective marker genes in two different in vitro models of Parkinson's disease. Neuropharmacology 2020; 194:108378. [PMID: 33160981 DOI: 10.1016/j.neuropharm.2020.108378] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 10/12/2020] [Accepted: 10/28/2020] [Indexed: 12/24/2022]
Abstract
Parkinson's disease (PD) is the second most common age-related neurodegenerative disorder after Alzheimer's disease. Increasing evidence highlights the role of age-related chronic inflammation, oxidative stress and mitochondrial dysfunction in the pathogenesis of PD. A combination of these factors impairs the crosstalk between mitochondria and lysosomes, resulting in compromised cell homeostasis. Apolipoprotein D (APOD), an ancient and highly conserved anti-inflammatory and antioxidant lipocalin, and the transcription factor EB (TFEB), a master regulator of mitophagy, autophagy and lysosomal biogenesis, play key roles in these processes. Both APOD and TFEB have attracted attention as therapeutic targets for PD. The aim of this study was to investigate if the selective cyclooxygenase-2 inhibitor celecoxib (CXB) exerts a direct neuroprotective effect in 6-hydroxydopamine (6-OHDA) and paraquat (PQ) PD models. We found that CXB rescued SH-SY5Y cells challenged by 6-OHDA- and PQ-induced toxicity. Furthermore, treatment with CXB led to a marked and sustained upregulation of APOD and the two microphthalmia transcription factors TFEB and MITF. In sum, this study highlights the clinically approved drug CXB as a promising neuroprotective therapeutic tool in PD research that has the potential to increase the survival rate of dopaminergic neurons that are still alive at the time of diagnosis.
Collapse
|
41
|
Jung SH, Lee W, Park SH, Lee KY, Choi YJ, Choi S, Kang D, Kim S, Chang TS, Hong SS, Lee BH. Diclofenac impairs autophagic flux via oxidative stress and lysosomal dysfunction: Implications for hepatotoxicity. Redox Biol 2020; 37:101751. [PMID: 33080439 PMCID: PMC7575798 DOI: 10.1016/j.redox.2020.101751] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/24/2020] [Accepted: 10/02/2020] [Indexed: 12/17/2022] Open
Abstract
Treatment with nonsteroidal anti-inflammatory drugs (NSAIDs) is associated with various side effects, including cardiovascular and hepatic disorders. Studies suggest that mitochondrial damage and oxidative stress are important mediators of toxicity, yet the underlying mechanisms are poorly understood. In this study, we identified that some NSAIDs, including diclofenac, inhibit autophagic flux in hepatocytes. Further detailed studies demonstrated that diclofenac induced a reactive oxygen species (ROS)-dependent increase in lysosomal pH, attenuated cathepsin activity and blocked autophagosome-lysosome fusion. The reactivation of lysosomal function by treatment with clioquinol or transfection with the transcription factor EB restored lysosomal pH and thus autophagic flux. The production of mitochondrial ROS is critical for this process since scavenging ROS reversed lysosomal dysfunction and activated autophagic flux. The compromised lysosomal activity induced by diclofenac also inhibited the fusion with and degradation of mitochondria by mitophagy. Diclofenac-induced cell death and hepatotoxicity were effectively protected by rapamycin. Thus, we demonstrated that diclofenac induces the intracellular ROS production and lysosomal dysfunction that lead to the suppression of autophagy. Impaired autophagy fails to maintain mitochondrial integrity and aggravates the cellular ROS burden, which leads to diclofenac-induced hepatotoxicity.
Collapse
Affiliation(s)
- Seung-Hwan Jung
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Wonseok Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Seung-Hyun Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Kang-Yo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - You-Jin Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Soohee Choi
- Department of Life Science, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Republic of Korea
| | - Dongmin Kang
- Department of Life Science, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Republic of Korea
| | - Sinri Kim
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, Republic of Korea
| | - Tong-Shin Chang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine, Inha University, Sinheung-dong, Jung-gu, Incheon, 400-712, Republic of Korea
| | - Byung-Hoon Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea.
| |
Collapse
|
42
|
Field BC, Gordillo R, Scherer PE. The Role of Ceramides in Diabetes and Cardiovascular Disease Regulation of Ceramides by Adipokines. Front Endocrinol (Lausanne) 2020; 11:569250. [PMID: 33133017 PMCID: PMC7564167 DOI: 10.3389/fendo.2020.569250] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022] Open
Abstract
Metabolic dysfunction is intertwined with the pathophysiology of both diabetes and cardiovascular disease. Recently, one particular lipid class has been shown to influence the development and sustainment of these diseases: ceramides. As a subtype of sphingolipids, these species are particularly central to many sphingolipid pathways. Increased levels of ceramides are known to correlate with impaired cardiovascular and metabolic health. Furthermore, the interaction between ceramides and adipokines, most notably adiponectin and leptin, appears to play a role in the pathophysiology of these conditions. Adiponectin appears to counteract the detrimental effects of elevated ceramides, largely through activation of the ceramidase activity of its receptors. Elevated ceramides appear to worsen leptin resistance, which is an important phenomenon in the pathophysiology of obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Bianca C. Field
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ruth Gordillo
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Philipp E. Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
43
|
De Lira MN, Raman SJ, Schulze A, Schneider-Schaulies S, Avota E. Neutral Sphingomyelinase-2 (NSM 2) Controls T Cell Metabolic Homeostasis and Reprogramming During Activation. Front Mol Biosci 2020; 7:217. [PMID: 33088808 PMCID: PMC7498697 DOI: 10.3389/fmolb.2020.00217] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/04/2020] [Indexed: 12/15/2022] Open
Abstract
Neutral sphingomyelinase-2 (NSM2) is a member of a superfamily of enzymes responsible for conversion of sphingomyelin into phosphocholine and ceramide at the cytosolic leaflet of the plasma membrane. Upon specific ablation of NSM2, T cells proved to be hyper-responsive to CD3/CD28 co-stimulation, indicating that the enzyme acts to dampen early overshooting activation of these cells. It remained unclear whether hyper-reactivity of NSM2-deficient T cells is supported by a deregulated metabolic activity in these cells. Here, we demonstrate that ablation of NSM2 activity affects metabolism of the quiescent CD4+ T cells which accumulate ATP in mitochondria and increase basal glycolytic activity. This supports enhanced production of total ATP and metabolic switch early after TCR/CD28 stimulation. Most interestingly, increased metabolic activity in resting NSM2-deficient T cells does not support sustained response upon stimulation. While elevated under steady-state conditions in NSM2-deficient CD4+ T cells, the mTORC1 pathway regulating mitochondria size, oxidative phosphorylation, and ATP production is impaired after 24 h of stimulation. Taken together, the absence of NSM2 promotes a hyperactive metabolic state in unstimulated CD4+ T cells yet fails to support sustained T cell responses upon antigenic stimulation.
Collapse
Affiliation(s)
| | | | - Almut Schulze
- Division of Tumor Metabolism and Microenvironment, German Cancer Research Center, Heidelberg, Germany
| | | | - Elita Avota
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
44
|
Stepien KM, Roncaroli F, Turton N, Hendriksz CJ, Roberts M, Heaton RA, Hargreaves I. Mechanisms of Mitochondrial Dysfunction in Lysosomal Storage Disorders: A Review. J Clin Med 2020; 9:jcm9082596. [PMID: 32796538 PMCID: PMC7463786 DOI: 10.3390/jcm9082596] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial dysfunction is emerging as an important contributory factor to the pathophysiology of lysosomal storage disorders (LSDs). The cause of mitochondrial dysfunction in LSDs appears to be multifactorial, although impaired mitophagy and oxidative stress appear to be common inhibitory mechanisms shared amongst these heterogeneous disorders. Once impaired, dysfunctional mitochondria may impact upon the function of the lysosome by the generation of reactive oxygen species as well as depriving the lysosome of ATP which is required by the V-ATPase proton pump to maintain the acidity of the lumen. Given the reported evidence of mitochondrial dysfunction in LSDs together with the important symbiotic relationship between these two organelles, therapeutic strategies targeting both lysosome and mitochondrial dysfunction may be an important consideration in the treatment of LSDs. In this review we examine the putative mechanisms that may be responsible for mitochondrial dysfunction in reported LSDs which will be supplemented with morphological and clinical information.
Collapse
Affiliation(s)
- Karolina M. Stepien
- Adult Inherited Metabolic Diseases, Salford Royal NHS Foundation Trust, Salford M6 8HD, UK
- Correspondence:
| | - Federico Roncaroli
- Division of Neuroscience and Experimental Psychology, School of Biology, Medicine and Health, University of Manchester and Manchester Centre for Clinical Neuroscience, Salford Royal NHS Foundation Trust, Salford M6 8HD, UK;
| | - Nadia Turton
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, UK; (N.T.); (R.A.H.); (I.H.)
| | - Christian J. Hendriksz
- Paediatrics and Child Health, Steve Biko Academic Unit, University of Pretoria, 0002 Pretoria, South Africa;
| | - Mark Roberts
- Neurology Department, Salford Royal NHS Foundation Trust, Salford M6 8HD, UK;
| | - Robert A. Heaton
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, UK; (N.T.); (R.A.H.); (I.H.)
| | - Iain Hargreaves
- School of Pharmacy, Liverpool John Moore University, Byrom Street, Liverpool L3 3AF, UK; (N.T.); (R.A.H.); (I.H.)
| |
Collapse
|
45
|
Brindisi M, Fiorillo M, Frattaruolo L, Sotgia F, Lisanti MP, Cappello AR. Cholesterol and Mevalonate: Two Metabolites Involved in Breast Cancer Progression and Drug Resistance through the ERRα Pathway. Cells 2020; 9:E1819. [PMID: 32751976 PMCID: PMC7465765 DOI: 10.3390/cells9081819] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the second greatest cause of cancer-related death in women. Resistance to endocrine treatments or chemotherapy is a limiting drawback. In this context, this work aims to evaluate the effects of cholesterol and mevalonate during tumor progression and their contribution in the onset of resistance to clinical treatments in use today. In this study, we demonstrated that cholesterol and mevalonate treatments were able to activate the estrogen-related receptor alpha (ERRα) pathway, increasing the expression levels of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), ERbB2/human epithelial receptor (HER2), tumor protein D52 (TPD52), and NOTCH2 proteins in breast cancer cells. The activation of this pathway is shown to be responsible for intense metabolic switching, higher proliferation rates, sustained motility, the propagation of cancer stem-like cells (CSCs), and lipid droplet formation. All of these events are related to greater tumor propagation, aggressiveness, and drug resistance. Furthermore, the activation and expression of proteins induced by the treatment with cholesterol or mevalonate are consistent with those obtained from the MCF-7/TAMr cell line, which is largely used as a breast cancer model of acquired endocrine therapy resistance. Altogether, our data indicate that cholesterol and mevalonate are two metabolites implicated in breast cancer progression, aggressiveness, and drug resistance, through the activation of the ERRα pathway. Our findings enable us to identify the ERRα receptor as a poor prognostic marker in patients with breast carcinoma, suggesting the correlation between cholesterol/mevalonate and ERRα as a new possible target in breast cancer treatment.
Collapse
Affiliation(s)
- Matteo Brindisi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy; (M.B.); (M.F.); (L.F.)
| | - Marco Fiorillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy; (M.B.); (M.F.); (L.F.)
- Translational Medicine, School of Science, Engineering and the Environment (SEE), University of Salford, Greater Manchester M5 4WT, UK
| | - Luca Frattaruolo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy; (M.B.); (M.F.); (L.F.)
| | - Federica Sotgia
- Translational Medicine, School of Science, Engineering and the Environment (SEE), University of Salford, Greater Manchester M5 4WT, UK
| | - Michael P. Lisanti
- Translational Medicine, School of Science, Engineering and the Environment (SEE), University of Salford, Greater Manchester M5 4WT, UK
| | - Anna Rita Cappello
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, 87036 Rende (CS), Italy; (M.B.); (M.F.); (L.F.)
| |
Collapse
|
46
|
Sabouny R, Shutt TE. Reciprocal Regulation of Mitochondrial Fission and Fusion. Trends Biochem Sci 2020; 45:564-577. [DOI: 10.1016/j.tibs.2020.03.009] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 03/03/2020] [Accepted: 03/16/2020] [Indexed: 12/24/2022]
|
47
|
Yañez MJ, Marín T, Balboa E, Klein AD, Alvarez AR, Zanlungo S. Finding pathogenic commonalities between Niemann-Pick type C and other lysosomal storage disorders: Opportunities for shared therapeutic interventions. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165875. [PMID: 32522631 DOI: 10.1016/j.bbadis.2020.165875] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/06/2020] [Accepted: 06/03/2020] [Indexed: 12/12/2022]
Abstract
Lysosomal storage disorders (LSDs) are diseases characterized by the accumulation of macromolecules in the late endocytic system and are caused by inherited defects in genes that encode mainly lysosomal enzymes or transmembrane lysosomal proteins. Niemann-Pick type C disease (NPCD), a LSD characterized by liver damage and progressive neurodegeneration that leads to early death, is caused by mutations in the genes encoding the NPC1 or NPC2 proteins. Both proteins are involved in the transport of cholesterol from the late endosomal compartment to the rest of the cell. Loss of function of these proteins causes primary cholesterol accumulation, and secondary accumulation of other lipids, such as sphingolipids, in lysosomes. Despite years of studying the genetic and molecular bases of NPCD and related-lysosomal disorders, the pathogenic mechanisms involved in these diseases are not fully understood. In this review we will summarize the pathogenic mechanisms described for NPCD and we will discuss their relevance for other LSDs with neurological components such as Niemann- Pick type A and Gaucher diseases. We will particularly focus on the activation of signaling pathways that may be common to these three pathologies with emphasis on how the intra-lysosomal accumulation of lipids leads to pathology, specifically to neurological impairments. We will show that although the primary lipid storage defect is different in these three LSDs, there is a similar secondary accumulation of metabolites and activation of signaling pathways that can lead to common pathogenic mechanisms. This analysis might help to delineate common pathological mechanisms and therapeutic targets for lysosomal storage diseases.
Collapse
Affiliation(s)
- M J Yañez
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - T Marín
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - E Balboa
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A D Klein
- Centro de Genética y Genómica, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - A R Alvarez
- Laboratory of Cell Signaling, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago, Chile; CARE UC, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - S Zanlungo
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
48
|
Cagin U, Puzzo F, Gomez MJ, Moya-Nilges M, Sellier P, Abad C, Van Wittenberghe L, Daniele N, Guerchet N, Gjata B, Collaud F, Charles S, Sola MS, Boyer O, Krijnse-Locker J, Ronzitti G, Colella P, Mingozzi F. Rescue of Advanced Pompe Disease in Mice with Hepatic Expression of Secretable Acid α-Glucosidase. Mol Ther 2020; 28:2056-2072. [PMID: 32526204 PMCID: PMC7474269 DOI: 10.1016/j.ymthe.2020.05.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/15/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
Pompe disease is a neuromuscular disorder caused by disease-associated variants in the gene encoding for the lysosomal enzyme acid α-glucosidase (GAA), which converts lysosomal glycogen to glucose. We previously reported full rescue of Pompe disease in symptomatic 4-month-old Gaa knockout (Gaa−/−) mice by adeno-associated virus (AAV) vector-mediated liver gene transfer of an engineered secretable form of GAA (secGAA). Here, we showed that hepatic expression of secGAA rescues the phenotype of 4-month-old Gaa−/− mice at vector doses at which the native form of GAA has little to no therapeutic effect. Based on these results, we then treated severely affected 9-month-old Gaa−/− mice with an AAV vector expressing secGAA and followed the animals for 9 months thereafter. AAV-treated Gaa−/− mice showed complete reversal of the Pompe phenotype, with rescue of glycogen accumulation in most tissues, including the central nervous system, and normalization of muscle strength. Transcriptomic profiling of skeletal muscle showed rescue of most altered pathways, including those involved in mitochondrial defects, a finding supported by structural and biochemical analyses, which also showed restoration of lysosomal function. Together, these results provide insight into the reversibility of advanced Pompe disease in the Gaa−/− mouse model via liver gene transfer of secGAA.
Collapse
Affiliation(s)
- Umut Cagin
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Francesco Puzzo
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France; Sorbonne Université, Paris, France
| | - Manuel Jose Gomez
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | | | - Pauline Sellier
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Catalina Abad
- Université de Rouen Normandie-IRIB, 76183 Rouen, France
| | | | - Nathalie Daniele
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Nicolas Guerchet
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Bernard Gjata
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Fanny Collaud
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Severine Charles
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Marcelo Simon Sola
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Olivier Boyer
- Université de Rouen Normandie-IRIB, 76183 Rouen, France
| | | | - Giuseppe Ronzitti
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Pasqualina Colella
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France
| | - Federico Mingozzi
- INTEGRARE, Genethon, INSERM, Université d'Evry, Université Paris-Saclay, 91002 Evry, France; Sorbonne Université, Paris, France; Spark Therapeutics, Philadelphia, PA 19103, USA.
| |
Collapse
|
49
|
Bertero E, Kutschka I, Maack C, Dudek J. Cardiolipin remodeling in Barth syndrome and other hereditary cardiomyopathies. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165803. [PMID: 32348916 DOI: 10.1016/j.bbadis.2020.165803] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/19/2019] [Accepted: 04/13/2020] [Indexed: 12/18/2022]
Abstract
Mitochondria play a prominent role in cardiac energy metabolism, and their function is critically dependent on the integrity of mitochondrial membranes. Disorders characterized by mitochondrial dysfunction are commonly associated with cardiac disease. The mitochondrial phospholipid cardiolipin directly interacts with a number of essential protein complexes in the mitochondrial membranes including the respiratory chain, mitochondrial metabolite carriers, and proteins critical for mitochondrial morphology. Barth syndrome is an X-linked disorder caused by an inherited defect in the biogenesis of the mitochondrial phospholipid cardiolipin. How cardiolipin deficiency impacts on mitochondrial function and how mitochondrial dysfunction causes cardiomyopathy has been intensively studied in cellular and animal models of Barth syndrome. These findings may also have implications for the molecular mechanisms underlying other inherited disorders associated with defects in cardiolipin, such as Sengers syndrome and dilated cardiomyopathy with ataxia (DCMA).
Collapse
Affiliation(s)
- Edoardo Bertero
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, 97078 Würzburg, Germany
| | - Ilona Kutschka
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, 97078 Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, 97078 Würzburg, Germany
| | - Jan Dudek
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, 97078 Würzburg, Germany.
| |
Collapse
|
50
|
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
|