1
|
Pahal S, Mainali N, Balasubramaniam M, Shmookler Reis RJ, Ayyadevara S. Mitochondria in aging and age-associated diseases. Mitochondrion 2025; 82:102022. [PMID: 40023438 DOI: 10.1016/j.mito.2025.102022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
Mitochondria, essential for cellular energy, are crucial in neurodegenerative disorders (NDDs) and their age-related progression. This review highlights mitochondrial dynamics, mitovesicles, homeostasis, and organelle communication. We examine mitochondrial impacts from aging and NDDs, focusing on protein aggregation and dysfunction. Prospective therapeutic approaches include enhancing mitophagy, improving respiratory chain function, maintaining calcium and lipid balance, using microRNAs, and mitochondrial transfer to protect function. These strategies underscore the crucial role of mitochondrial health in neuronal survival and cognitive functions, offering new therapeutic opportunities.
Collapse
Affiliation(s)
- Sonu Pahal
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock AR 72205, U.S.A
| | - Nirjal Mainali
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock AR 72205, U.S.A
| | | | - Robert J Shmookler Reis
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock AR 72205, U.S.A; Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205, U.S.A; Central Arkansas Veterans Healthcare Service, Little Rock AR 72205, U.S.A.
| | - Srinivas Ayyadevara
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock AR 72205, U.S.A; Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205, U.S.A; Central Arkansas Veterans Healthcare Service, Little Rock AR 72205, U.S.A.
| |
Collapse
|
2
|
Zhang X, Buckley C, Lee MD, Chalmers S, Wilson C, McCarron JG. Mitochondria regulate inositol triphosphate-mediated Ca 2+ release triggered by voltage-dependent Ca 2+ entry in resistance arteries. J Physiol 2025; 603:2959-2978. [PMID: 40320995 DOI: 10.1113/jp288022] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/19/2025] [Indexed: 06/02/2025] Open
Abstract
An increase in cytoplasmic Ca2+ concentration activates multiple cellular activities, including cell division, metabolism, growth, contraction and death. In smooth muscle Ca2+ entry via voltage-dependent Ca2+ channels leads to a relatively uniform increase in cytoplasmic Ca2+ levels that facilitates co-ordinated contraction throughout the cell. However certain functions triggered by voltage-dependent Ca2+ channels require periodic, pulsatile Ca2+ changes. The mechanism by which Ca2+ entry through voltage-dependent channels supports both co-ordinated contraction and distinct cellular responses driven by pulsatile Ca2+ changes is unclear. Here in intact resistance arteries we show that Ca2+ entry via voltage-dependent Ca2+ channels evokes Ca2+ release via inositol triphosphate receptors (IP3Rs), generating repetitive Ca2+ oscillations and waves. We also show that mitochondria play a vital role in regulating Ca2+ signals evoked by voltage-dependent Ca2+ entry by selectively modulating Ca2+ release via IP3Rs. Depolarizing the mitochondrial membrane inhibits Ca2+ release from internal stores, reducing the overall signal-generated Ca2+ influx without altering the signal resulting from voltage-dependent Ca2+ entry. Notably neither Ca2+ entry via voltage-dependent Ca2+ channels nor Ca2+ release via IP3Rs alters mitochondrial location or mitochondrial membrane potential in intact smooth muscle cells. Collectively these results demonstrate that activation of voltage-dependent Ca2+ channels drives Ca2+ entry, which subsequently triggers Ca2+ release from the internal store in smooth muscle cells. Mitochondria selectively regulate this process by modulating IP3R-mediated amplification of Ca2+ signals, ensuring that different cellular responses are precisely controlled. KEY POINTS: In smooth muscle Ca2⁺ entry via voltage-dependent channels produces a uniform Ca2⁺ increase, enabling co-ordinated contraction in each cell. Certain functions, however, require large, pulsatile Ca2⁺ changes rather than a uniform increase. Using advanced imaging in intact arteries, we discovered that voltage-dependent Ca2⁺ entry triggers internal store Ca2⁺ release via IP₃ receptors, generating repetitive Ca2⁺ oscillations and waves. Mitochondria selectively modulate these signals by regulating only IP₃ receptor-mediated release; neither mitochondrial location nor membrane potential is altered by either type of Ca2+ signal. These findings demonstrate how voltage-dependent Ca2⁺ entry supports both co-ordinated contraction and pulsatile Ca2⁺-driven biological responses.
Collapse
Affiliation(s)
- Xun Zhang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Charlotte Buckley
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Matthew D Lee
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Susan Chalmers
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Calum Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - John G McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
3
|
Maddineni P, Kodati B, Kaipa BR, Kesavan K, Cameron Millar J, Yacoub S, Kasetti RB, Clark AF, Zode GS. Genetic and pharmacological correction of impaired mitophagy in retinal ganglion cells rescues glaucomatous neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.638142. [PMID: 39990391 PMCID: PMC11844533 DOI: 10.1101/2025.02.13.638142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Progressive loss of retinal ganglion cells (RGCs) and degeneration of optic nerve axons are the pathological hallmarks of glaucoma. Ocular hypertension (OHT) and mitochondrial dysfunction are linked to neurodegeneration and vision loss in glaucoma. However, the exact mechanism of mitochondrial dysfunction leading to glaucomatous neurodegeneration is poorly understood. Using multiple mouse models of OHT and human eyes from normal and glaucoma donors, we show that OHT induces impaired mitophagy in RGCs, resulting in the accumulation of dysfunctional mitochondria and contributing to glaucomatous neurodegeneration. Using mitophagy reporter mice, we show that impaired mitophagy precedes glaucomatous neurodegeneration. Notably, the pharmacological rescue of impaired mitophagy via Torin-2 or genetic upregulation of RGC-specific Parkin expression restores the structural and functional integrity of RGCs and their axons in mouse models of glaucoma and ex-vivo human retinal-explant cultures. Our study indicates that impaired mitophagy contributes to mitochondrial dysfunction and oxidative stress, leading to glaucomatous neurodegeneration. Enhancing mitophagy in RGCs represents a promising therapeutic strategy to prevent glaucomatous neurodegeneration.
Collapse
|
4
|
Wei Y, Du X, Guo H, Han J, Liu M. Mitochondrial dysfunction and Alzheimer's disease: pathogenesis of mitochondrial transfer. Front Aging Neurosci 2024; 16:1517965. [PMID: 39741520 PMCID: PMC11685155 DOI: 10.3389/fnagi.2024.1517965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025] Open
Abstract
In recent years, mitochondrial transfer has emerged as a universal phenomenon intertwined with various systemic physiological and pathological processes. Alzheimer's disease (AD) is a multifactorial disease, with mitochondrial dysfunction at its core. Although numerous studies have found evidence of mitochondrial transfer in AD models, the precise mechanisms remain unclear. Recent studies have revealed the dynamic transfer of mitochondria in Alzheimer's disease, not only between nerve cells and glial cells, but also between nerve cells and glial cells. In this review, we explore the pathways and mechanisms of mitochondrial transfer in Alzheimer's disease and how these transfer activities contribute to disease progression.
Collapse
Affiliation(s)
- Yun Wei
- *Correspondence: Yun Wei, ; Meixia Liu,
| | | | | | | | - Meixia Liu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
5
|
Dayal AA, Parfenteva OI, Wang H, Gebreselase BA, Gyoeva FK, Alieva IB, Minin AA. Vimentin Intermediate Filaments Maintain Membrane Potential of Mitochondria in Growing Neurites. BIOLOGY 2024; 13:995. [PMID: 39765662 PMCID: PMC11726714 DOI: 10.3390/biology13120995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 01/15/2025]
Abstract
Neural precursor cells contain two types of intermediate filaments (IFs): neurofilaments consisting of three IV type proteins and vimentin belonging to the type III IF proteins that disappear at the later stages of differentiation. The involvement of vimentin in neurogenesis was demonstrated earlier; however, the role of its temporary expression in neurons is not clear. We showed that the vimentin IFs that interacted with mitochondria maintained their membrane potential at the appropriate level, and thus, ensured their proper function. We examined the dependence of the mitochondrial membrane potential on the expression of vimentin in a CAD catecholaminergic neuronal cell line that was actively dividing in full culture media but stopped growing and started developing neurites when the serum was removed. Using the CRISPR Cas9 system to knock out the vimentin gene in these cells, we investigated the impact of this on the mitochondrial membrane potential. Our data show that the deletion of the vimentin IFs led to a decrease in the level of the mitochondrial potential. When the vimentin network in these cells was reconstituted by transfection with a plasmid that encoded human protein, the level of the potential was restored. Interestingly, mutated vimentin with a disrupted mitochondria-binding site had no such effect. Our data point to vimentin as a possible target in some neurological pathologies.
Collapse
Affiliation(s)
- Alexander A. Dayal
- Institute of Protein Research, Russian Academy of Sciences, 119334 Moscow, Russia (I.B.A.)
| | - Olga I. Parfenteva
- Institute of Protein Research, Russian Academy of Sciences, 119334 Moscow, Russia (I.B.A.)
| | - Huiying Wang
- Institute of Protein Research, Russian Academy of Sciences, 119334 Moscow, Russia (I.B.A.)
| | - Blen Amare Gebreselase
- Institute of Protein Research, Russian Academy of Sciences, 119334 Moscow, Russia (I.B.A.)
| | - Fatima K. Gyoeva
- Institute of Protein Research, Russian Academy of Sciences, 119334 Moscow, Russia (I.B.A.)
| | - Irina B. Alieva
- Institute of Protein Research, Russian Academy of Sciences, 119334 Moscow, Russia (I.B.A.)
- Belozersky Institute of Physical and Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Alexander A. Minin
- Institute of Protein Research, Russian Academy of Sciences, 119334 Moscow, Russia (I.B.A.)
| |
Collapse
|
6
|
Martínez P, Silva M, Abarzúa S, Tevy MF, Jaimovich E, Constantine-Paton M, Bustos FJ, van Zundert B. Skeletal myotubes expressing ALS mutant SOD1 induce pathogenic changes, impair mitochondrial axonal transport, and trigger motoneuron death. Mol Med 2024; 30:185. [PMID: 39455931 PMCID: PMC11505737 DOI: 10.1186/s10020-024-00942-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the loss of motoneurons (MNs), and despite progress, there is no effective treatment. A large body of evidence shows that astrocytes expressing ALS-linked mutant proteins cause non-cell autonomous toxicity of MNs. Although MNs innervate muscle fibers and ALS is characterized by the early disruption of the neuromuscular junction (NMJ) and axon degeneration, there are controversies about whether muscle contributes to non-cell-autonomous toxicity to MNs. In this study, we generated primary skeletal myotubes from myoblasts derived from ALS mice expressing human mutant SOD1G93A (termed hereafter mutSOD1). Characterization revealed that mutSOD1 skeletal myotubes display intrinsic phenotypic and functional differences compared to control myotubes generated from non-transgenic (NTg) littermates. Next, we analyzed whether ALS myotubes exert non-cell-autonomous toxicity to MNs. We report that conditioned media from mutSOD1 myotubes (mutSOD1-MCM), but not from control myotubes (NTg-MCM), induced robust death of primary MNs in mixed spinal cord cultures and compartmentalized microfluidic chambers. Our study further revealed that applying mutSOD1-MCM to the MN axonal side in microfluidic devices rapidly reduces mitochondrial axonal transport while increasing Ca2 + transients and reactive oxygen species (i.e., H2O2). These results indicate that soluble factor(s) released by mutSOD1 myotubes cause MN axonopathy that leads to lethal pathogenic changes.
Collapse
Affiliation(s)
- Pablo Martínez
- Institute of Biomedical Sciences (ICB), Faculty of Medicine & Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Mónica Silva
- Center for Exercise, Metabolism and Cancer, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Sebastián Abarzúa
- Institute of Biomedical Sciences (ICB), Faculty of Medicine & Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | | | - Enrique Jaimovich
- Center for Exercise, Metabolism and Cancer, Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Martha Constantine-Paton
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Fernando J Bustos
- Institute of Biomedical Sciences (ICB), Faculty of Medicine & Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile.
| | - Brigitte van Zundert
- Institute of Biomedical Sciences (ICB), Faculty of Medicine & Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Millennium Nucleus of Neuroepigenetics and Plasticity (EpiNeuro), Santiago, Chile.
- Department of Neurology, University of Massachusetts Chan Medical School (UMMS), Worcester, MA, USA.
| |
Collapse
|
7
|
Sleigh JN, Mattedi F, Richter S, Annuario E, Ng K, Steinmark IE, Ivanova I, Darabán IL, Joshi PP, Rhymes ER, Awale S, Yahioglu G, Mitchell JC, Suhling K, Schiavo G, Vagnoni A. Age-specific and compartment-dependent changes in mitochondrial homeostasis and cytoplasmic viscosity in mouse peripheral neurons. Aging Cell 2024; 23:e14250. [PMID: 38881280 PMCID: PMC11464114 DOI: 10.1111/acel.14250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/26/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
Mitochondria are dynamic bioenergetic hubs that become compromised with age. In neurons, declining mitochondrial axonal transport has been associated with reduced cellular health. However, it is still unclear to what extent the decline of mitochondrial transport and function observed during ageing are coupled, and if somal and axonal mitochondria display compartment-specific features that make them more susceptible to the ageing process. It is also not known whether the biophysical state of the cytoplasm, thought to affect many cellular functions, changes with age to impact mitochondrial trafficking and homeostasis. Focusing on the mouse peripheral nervous system, we show that age-dependent decline in mitochondrial trafficking is accompanied by reduction of mitochondrial membrane potential and intramitochondrial viscosity, but not calcium buffering, in both somal and axonal mitochondria. Intriguingly, we observe a specific increase in cytoplasmic viscosity in the neuronal cell body, where mitochondria are most polarised, which correlates with decreased cytoplasmic diffusiveness. Increasing cytoplasmic crowding in the somatic compartment of DRG neurons grown in microfluidic chambers reduces mitochondrial axonal trafficking, suggesting a mechanistic link between the regulation of cytoplasmic viscosity and mitochondrial dynamics. Our work provides a reference for studying the relationship between neuronal mitochondrial homeostasis and the viscoelasticity of the cytoplasm in a compartment-dependent manner during ageing.
Collapse
Affiliation(s)
- James N. Sleigh
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease CentreUCL Queen Square Institute of Neurology, University College LondonLondonUK
- UK Dementia Research Institute, University College LondonLondonUK
| | - Francesca Mattedi
- Department of Basic and Clinical NeurosciencesMaurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUK
- Present address:
Department of Neuromuscular DiseasesUCL Queen Square Institute of Neurology, University College LondonLondonUK
| | - Sandy Richter
- Department of Basic and Clinical NeurosciencesMaurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUK
- Present address:
Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Emily Annuario
- Department of Basic and Clinical NeurosciencesMaurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUK
| | - Kristal Ng
- Department of Basic and Clinical NeurosciencesMaurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUK
| | | | | | - István L. Darabán
- Department of Basic and Clinical NeurosciencesMaurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUK
| | - Parth P. Joshi
- Department of Basic and Clinical NeurosciencesMaurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUK
- Present address:
Sunderland Medical School, University of SunderlandSunderlandUK
| | - Elena R. Rhymes
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease CentreUCL Queen Square Institute of Neurology, University College LondonLondonUK
- UK Dementia Research Institute, University College LondonLondonUK
| | - Shirwa Awale
- Department of PhysicsKing's College LondonLondonUK
| | - Gokhan Yahioglu
- Antikor Biopharma Ltd, Stevenage Bioscience CatalystStevenageUK
| | - Jacqueline C. Mitchell
- Department of Basic and Clinical NeurosciencesMaurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUK
| | | | - Giampietro Schiavo
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease CentreUCL Queen Square Institute of Neurology, University College LondonLondonUK
- UK Dementia Research Institute, University College LondonLondonUK
| | - Alessio Vagnoni
- Department of Basic and Clinical NeurosciencesMaurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College LondonLondonUK
- MIA‐PortugalMultidisciplinary Institute of Ageing, University of CoimbraCoimbraPortugal
| |
Collapse
|
8
|
Wai T. Is mitochondrial morphology important for cellular physiology? Trends Endocrinol Metab 2024; 35:854-871. [PMID: 38866638 DOI: 10.1016/j.tem.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024]
Abstract
Mitochondria are double membrane-bound organelles the network morphology of which in cells is shaped by opposing events of fusion and fission executed by dynamin-like GTPases. Mutations in these genes can perturb the form and functions of mitochondria in cell and animal models of mitochondrial diseases. An expanding array of chemical, mechanical, and genetic stressors can converge on mitochondrial-shaping proteins and disrupt mitochondrial morphology. In recent years, studies aimed at disentangling the multiple roles of mitochondrial-shaping proteins beyond fission or fusion have provided insights into the homeostatic relevance of mitochondrial morphology. Here, I review the pleiotropy of mitochondrial fusion and fission proteins with the aim of understanding whether mitochondrial morphology is important for cell and tissue physiology.
Collapse
Affiliation(s)
- Timothy Wai
- Institut Pasteur, Mitochondrial Biology, CNRS UMR 3691, Université Paris Cité, Paris, France.
| |
Collapse
|
9
|
Nheu D, Petratos S. How does Nogo-A signalling influence mitochondrial function during multiple sclerosis pathogenesis? Neurosci Biobehav Rev 2024; 163:105767. [PMID: 38885889 DOI: 10.1016/j.neubiorev.2024.105767] [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: 03/17/2024] [Revised: 05/30/2024] [Accepted: 06/08/2024] [Indexed: 06/20/2024]
Abstract
Multiple sclerosis (MS) is a severe neurological disorder that involves inflammation in the brain, spinal cord and optic nerve with key disabling neuropathological outcomes being axonal damage and demyelination. When degeneration of the axo-glial union occurs, a consequence of inflammatory damage to central nervous system (CNS) myelin, dystrophy and death can lead to large membranous structures from dead oligodendrocytes and degenerative myelin deposited in the extracellular milieu. For the first time, this review covers mitochondrial mechanisms that may be operative during MS-related neurodegenerative changes directly activated during accumulating extracellular deposits of myelin associated inhibitory factors (MAIFs), that include the potent inhibitor of neurite outgrowth, Nogo-A. Axonal damage may occur when Nogo-A binds to and signals through its cognate receptor, NgR1, a multimeric complex, to initially stall axonal transport and limit the delivery of important growth-dependent cargo and subcellular organelles such as mitochondria for metabolic efficiency at sites of axo-glial disintegration as a consequence of inflammation. Metabolic efficiency in axons fails during active demyelination and progressive neurodegeneration, preceded by stalled transport of functional mitochondria to fuel axo-glial integrity.
Collapse
Affiliation(s)
- Danica Nheu
- Department of Neuroscience, School of Translational Medicine, Monash University, Prahran, VIC 3004, Australia
| | - Steven Petratos
- Department of Neuroscience, School of Translational Medicine, Monash University, Prahran, VIC 3004, Australia.
| |
Collapse
|
10
|
López-Ayllón BD, Marin S, Fernández MF, García-García T, Fernández-Rodríguez R, de Lucas-Rius A, Redondo N, Mendoza-García L, Foguet C, Grigas J, Calvet A, Villalba JM, Gómez MJR, Megías D, Mandracchia B, Luque D, Lozano JJ, Calvo C, Herrán UM, Thomson TM, Garrido JJ, Cascante M, Montoya M. Metabolic and mitochondria alterations induced by SARS-CoV-2 accessory proteins ORF3a, ORF9b, ORF9c and ORF10. J Med Virol 2024; 96:e29752. [PMID: 38949191 DOI: 10.1002/jmv.29752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 06/07/2024] [Accepted: 06/08/2024] [Indexed: 07/02/2024]
Abstract
Antiviral signaling, immune response and cell metabolism are dysregulated by SARS-CoV-2, the causative agent of COVID-19. Here, we show that SARS-CoV-2 accessory proteins ORF3a, ORF9b, ORF9c and ORF10 induce a significant mitochondrial and metabolic reprogramming in A549 lung epithelial cells. While ORF9b, ORF9c and ORF10 induced largely overlapping transcriptomes, ORF3a induced a distinct transcriptome, including the downregulation of numerous genes with critical roles in mitochondrial function and morphology. On the other hand, all four ORFs altered mitochondrial dynamics and function, but only ORF3a and ORF9c induced a marked alteration in mitochondrial cristae structure. Genome-Scale Metabolic Models identified both metabolic flux reprogramming features both shared across all accessory proteins and specific for each accessory protein. Notably, a downregulated amino acid metabolism was observed in ORF9b, ORF9c and ORF10, while an upregulated lipid metabolism was distinctly induced by ORF3a. These findings reveal metabolic dependencies and vulnerabilities prompted by SARS-CoV-2 accessory proteins that may be exploited to identify new targets for intervention.
Collapse
Affiliation(s)
- Blanca D López-Ayllón
- Viral Immunology Lab, Molecular Biomedicine Department, BICS Unit. Margarita Salas Center for Biological Research (CIB-CSIC), Madrid, Spain
| | - Silvia Marin
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona (UB), Barcelona, Spain
- CIBER of Hepatic and Digestive Diseases (CIBEREHD), Institute of Health Carlos III (ISCIII), Madrid, Spain
- Institute of Biomedicine of University of Barcelona (IBUB), University of Barcelona (UB), Barcelona, Spain
| | - Marco Fariñas Fernández
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona (UB), Barcelona, Spain
- Department of Biomedical Laboratory Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Tránsito García-García
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonoses and Emergent Diseases ENZOEM, Department of Genetics, University of Córdoba, Córdoba, Spain
- Maimónides Biomedical Research, Institute of Córdoba (IMIBIC), Córdoba, Spain
| | - Raúl Fernández-Rodríguez
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonoses and Emergent Diseases ENZOEM, Department of Genetics, University of Córdoba, Córdoba, Spain
- Maimónides Biomedical Research, Institute of Córdoba (IMIBIC), Córdoba, Spain
| | - Ana de Lucas-Rius
- Viral Immunology Lab, Molecular Biomedicine Department, BICS Unit. Margarita Salas Center for Biological Research (CIB-CSIC), Madrid, Spain
| | - Natalia Redondo
- Unit of Infectious Diseases, University Hospital '12 de Octubre', Institute for Health Research Hospital '12 de Octubre' (imas12), Madrid, Spain
- Centre for Biomedical Research Network on Infectious Diseases (CIBERINFEC), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Laura Mendoza-García
- Viral Immunology Lab, Molecular Biomedicine Department, BICS Unit. Margarita Salas Center for Biological Research (CIB-CSIC), Madrid, Spain
| | - Carles Foguet
- British Heart Foundation Cardiovascular Epidemiology Unit and Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Juozas Grigas
- Laboratory of Immunology, Department of Anatomy and Physiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Institute of Microbiology and Virology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Alba Calvet
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona (UB), Barcelona, Spain
- Institute of Biomedicine of University of Barcelona (IBUB), University of Barcelona (UB), Barcelona, Spain
| | - José Manuel Villalba
- Department of Cell Biology, Physiology and Immunology, Agrifood Campus of International Excellence, University of Córdoba, Córdoba, Spain
| | - María Josefa Rodríguez Gómez
- Scientific-Technical Central Units, Instituto de Salud Carlos III (ISCIII), Majadahonda, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Diego Megías
- Scientific-Technical Central Units, Instituto de Salud Carlos III (ISCIII), Majadahonda, Spain
| | - Biagio Mandracchia
- Scientific-Technical Central Units, Instituto de Salud Carlos III (ISCIII), Majadahonda, Spain
- ETSI Telecommunication, University of Valladolid, Valladolid, Spain
| | - Daniel Luque
- Scientific-Technical Central Units, Instituto de Salud Carlos III (ISCIII), Majadahonda, Spain
- Electron Microscope Unit, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Juan José Lozano
- CIBER of Hepatic and Digestive Diseases (CIBEREHD), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Cristina Calvo
- Barcelona Institute for Molecular Biology (IBMB-CSIC), Barcelona, Spain
| | - Unai Merino Herrán
- Viral Immunology Lab, Molecular Biomedicine Department, BICS Unit. Margarita Salas Center for Biological Research (CIB-CSIC), Madrid, Spain
| | - Timothy M Thomson
- CIBER of Hepatic and Digestive Diseases (CIBEREHD), Institute of Health Carlos III (ISCIII), Madrid, Spain
- Barcelona Institute for Molecular Biology (IBMB-CSIC), Barcelona, Spain
- Translational Research and Computational Biology Laboratory, Faculty of Science and Engineering, Peruvian University Cayetano Heredia, Lima, Perú
| | - Juan J Garrido
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonoses and Emergent Diseases ENZOEM, Department of Genetics, University of Córdoba, Córdoba, Spain
- Maimónides Biomedical Research, Institute of Córdoba (IMIBIC), Córdoba, Spain
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona (UB), Barcelona, Spain
- CIBER of Hepatic and Digestive Diseases (CIBEREHD), Institute of Health Carlos III (ISCIII), Madrid, Spain
- Institute of Biomedicine of University of Barcelona (IBUB), University of Barcelona (UB), Barcelona, Spain
| | - María Montoya
- Viral Immunology Lab, Molecular Biomedicine Department, BICS Unit. Margarita Salas Center for Biological Research (CIB-CSIC), Madrid, Spain
| |
Collapse
|
11
|
Martínez P, Silva M, Abarzúa S, Tevy MF, Jaimovich E, Constantine-Paton M, Bustos FJ, van Zundert B. Skeletal myotubes expressing ALS mutant SOD1 induce pathogenic changes, impair mitochondrial axonal transport, and trigger motoneuron death. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595817. [PMID: 38826246 PMCID: PMC11142234 DOI: 10.1101/2024.05.24.595817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the loss of motoneurons (MNs), and despite progress, there is no effective treatment. A large body of evidence shows that astrocytes expressing ALS-linked mutant proteins cause non-cell autonomous toxicity of MNs. Although MNs innervate muscle fibers and ALS is characterized by the early disruption of the neuromuscular junction (NMJ) and axon degeneration, there are controversies about whether muscle contributes to non-cell-autonomous toxicity to MNs. In this study, we generated primary skeletal myotubes from myoblasts derived from ALS mice expressing human mutant SOD1 G93A (termed hereafter mutSOD1). Characterization revealed that mutSOD1 skeletal myotubes display intrinsic phenotypic and functional differences compared to control myotubes generated from non-transgenic (NTg) littermates. Next, we analyzed whether ALS myotubes exert non-cell-autonomous toxicity to MNs. We report that conditioned media from mutSOD1 myotubes (mutSOD1-MCM), but not from control myotubes (NTg-MCM), induced robust death of primary MNs in mixed spinal cord cultures and compartmentalized microfluidic chambers. Our study further revealed that applying mutSOD1-MCM to the MN axonal side in microfluidic devices rapidly reduces mitochondrial axonal transport while increasing Ca2+ transients and reactive oxygen species (i.e., H 2 O 2 ). These results indicate that soluble factor(s) released by mutSOD1 myotubes cause MN axonopathy that leads to lethal pathogenic changes.
Collapse
|
12
|
Alberti C, Rizzo F, Anastasia A, Comi G, Corti S, Abati E. Charcot-Marie-tooth disease type 2A: An update on pathogenesis and therapeutic perspectives. Neurobiol Dis 2024; 193:106467. [PMID: 38452947 DOI: 10.1016/j.nbd.2024.106467] [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: 01/03/2024] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024] Open
Abstract
Mutations in the gene encoding MFN2 have been identified as associated with Charcot-Marie-Tooth disease type 2A (CMT2A), a neurological disorder characterized by a broad clinical phenotype involving the entire nervous system. MFN2, a dynamin-like GTPase protein located on the outer mitochondrial membrane, is well-known for its involvement in mitochondrial fusion. Numerous studies have demonstrated its participation in a network crucial for various other mitochondrial functions, including mitophagy, axonal transport, and its controversial role in endoplasmic reticulum (ER)-mitochondria contacts. Considerable progress has been made in the last three decades in elucidating the disease pathogenesis, aided by the generation of animal and cellular models that have been instrumental in studying disease physiology. A review of the literature reveals that, up to now, no definitive pharmacological treatment for any CMT2A variant has been established; nonetheless, recent years have witnessed substantial progress. Many treatment approaches, especially concerning molecular therapy, such as histone deacetylase inhibitors, peptide therapy to increase mitochondrial fusion, the new therapeutic strategies based on MF1/MF2 balance, and SARM1 inhibitors, are currently in preclinical testing. The literature on gene silencing and gene replacement therapies is still limited, except for a recent study by Rizzo et al.(Rizzo et al., 2023), which recently first achieved encouraging results in in vitro and in vivo models of the disease. The near-future goal for these promising therapies is to progress to the stage of clinical translation.
Collapse
Affiliation(s)
- Claudia Alberti
- Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy
| | - Federica Rizzo
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alessia Anastasia
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giacomo Comi
- Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy; Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy; Neuromuscular and Rare Diseases Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elena Abati
- Department of Pathophysiology and Transplantation (DEPT), University of Milan, Milan, Italy; Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
13
|
Bartal G, Yitzhaky A, Segev A, Hertzberg L. Multiple genes encoding mitochondrial ribosomes are downregulated in brain and blood samples of individuals with schizophrenia. World J Biol Psychiatry 2023; 24:829-837. [PMID: 37158323 DOI: 10.1080/15622975.2023.2211653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/04/2023] [Indexed: 05/10/2023]
Abstract
OBJECTIVES Schizophrenia is a chronic, debilitating mental disorder whose pathophysiology is complex and not fully understood. Numerous studies suggest mitochondrial dysfunction may contribute to the development of schizophrenia. While mitochondrial ribosomes (mitoribosomes) are essential for proper mitochondrial functioning, their gene expression levels have not been studied yet in schizophrenia. METHODS We performed a systematic meta-analysis of the expression of 81 mitoribosomes subunits encoding genes, integrating ten brain samples datasets of patients with schizophrenia compared to healthy controls (overall 422 samples, 211 schizophrenia, and 211 controls). We also performed a meta-analysis of their expression in blood, integrating two blood sample datasets (overall 90 samples, 53 schizophrenia, and 37 controls). RESULTS Multiple mitoribosomes subunits were significantly downregulated in brain samples (18 genes) and in blood samples (11 genes) of individuals with schizophrenia, where two showed significant downregulation in both brain and blood, MRPL4 and MRPS7. CONCLUSIONS Our results support the accumulating evidence of impaired mitochondrial activity in schizophrenia. While further research is needed to validate mitoribosomes' role as biomarkers, this direction has the potential to promote patients' stratification and personalised treatment for schizophrenia.
Collapse
Affiliation(s)
- Gideon Bartal
- The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Assif Yitzhaky
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot, Israel
| | - Aviv Segev
- The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Shalvata Mental Health Center, Hod Hasharon, Israel
| | - Libi Hertzberg
- Department of Physics of Complex Systems, Weizmann Institute of Science, Rehovot, Israel
- The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Shalvata Mental Health Center, Hod Hasharon, Israel
| |
Collapse
|
14
|
Keefe AJ, Gabrych DR, Zhu Y, Vocadlo DJ, Silverman MA. Axonal Transport of Lysosomes Is Unaffected in Glucocerebrosidase-Inhibited iPSC-Derived Forebrain Neurons. eNeuro 2023; 10:ENEURO.0079-23.2023. [PMID: 37816595 PMCID: PMC10576257 DOI: 10.1523/eneuro.0079-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/12/2023] Open
Abstract
Lysosomes are acidic organelles that traffic throughout neurons delivering catabolic enzymes to distal regions of the cell and maintaining degradative demands. Loss of function mutations in the gene GBA encoding the lysosomal enzyme glucocerebrosidase (GCase) cause the lysosomal storage disorder Gaucher's disease (GD) and are the most common genetic risk factor for synucleinopathies like Parkinson's disease (PD) and dementia with Lewy bodies (DLB). GCase degrades the membrane lipid glucosylceramide (GlcCer) and mutations in GBA, or inhibiting its activity, results in the accumulation of GlcCer and disturbs the composition of the lysosomal membrane. The lysosomal membrane serves as the platform to which intracellular trafficking complexes are recruited and activated. Here, we investigated whether lysosomal trafficking in axons was altered by inhibition of GCase with the pharmacological agent Conduritol B Epoxide (CBE). Using live cell imaging in human male induced pluripotent human stem cell (iPSC)-derived forebrain neurons, we demonstrated that lysosomal transport was similar in both control and CBE-treated neurons. Furthermore, we tested whether lysosomal rupture, a process implicated in various neurodegenerative disorders, was affected by inhibition of GCase. Using L-leucyl-L-leucine methyl ester (LLoME) to induce lysosomal membrane damage and immunocytochemical staining for markers of lysosomal rupture, we found no difference in susceptibility to rupture between control and CBE-treated neurons. These results suggest the loss of GCase activity does not contribute to neurodegenerative disease by disrupting either lysosomal transport or rupture.
Collapse
Affiliation(s)
- A J Keefe
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - D R Gabrych
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Y Zhu
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - D J Vocadlo
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - M A Silverman
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| |
Collapse
|
15
|
Escrig-Larena JI, Delgado-Pulido S, Mittelbrunn M. Mitochondria during T cell aging. Semin Immunol 2023; 69:101808. [PMID: 37473558 DOI: 10.1016/j.smim.2023.101808] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 07/22/2023]
Abstract
Mitochondrial dysfunction is a hallmark of aging that contributes to inflammaging. It is characterized by alterations of the mitochondrial DNA, reduced respiratory capacity, decreased mitochondrial membrane potential and increased reactive oxygen species production. These primary alterations disrupt other interconnected and important mitochondrial-related processes such as metabolism, mitochondrial dynamics and biogenesis, mitophagy, calcium homeostasis or apoptosis. In this review, we gather the current knowledge about the different mitochondrial processes which are altered during aging, with special focus on their contribution to age-associated T cell dysfunction and inflammaging.
Collapse
Affiliation(s)
- Jose Ignacio Escrig-Larena
- Consejo Superior de Investigaciones Científicas (CSIC), Centro de Biología Molcular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Sandra Delgado-Pulido
- Departamento de Biología Molecular, Facultad de Ciencias (UAM), Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - María Mittelbrunn
- Consejo Superior de Investigaciones Científicas (CSIC), Centro de Biología Molcular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid (UAM), Madrid, Spain.
| |
Collapse
|
16
|
Jeong Y, Davis CHO, Muscarella AM, Deshpande V, Kim KY, Ellisman MH, Marsh-Armstrong N. Glaucoma-associated Optineurin mutations increase transmitophagy in a vertebrate optic nerve. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.26.542507. [PMID: 37398269 PMCID: PMC10312487 DOI: 10.1101/2023.05.26.542507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
We previously described a process referred to as transmitophagy where mitochondria shed by retinal ganglion cell (RGC) axons are transferred to and degraded by surrounding astrocytes in the optic nerve head of mice. Since the mitophagy receptor Optineurin (OPTN) is one of few large-effect glaucoma genes and axonal damage occurs at the optic nerve head in glaucoma, here we explored whether OPTN mutations perturb transmitophagy. Live-imaging of Xenopus laevis optic nerves revealed that diverse human mutant but not wildtype OPTN increase stationary mitochondria and mitophagy machinery and their colocalization within, and in the case of the glaucoma-associated OPTN mutations also outside of, RGC axons. These extra-axonal mitochondria are degraded by astrocytes. Our studies support the view that in RGC axons under baseline conditions there are low levels of mitophagy, but that glaucoma-associated perturbations in OPTN result in increased axonal mitophagy involving the shedding and astrocytic degradation of the mitochondria.
Collapse
Affiliation(s)
- Yaeram Jeong
- Department of Ophthalmology and Vision Science, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | | | - Aaron M. Muscarella
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA 92093, USA
| | - Viraj Deshpande
- Department of Ophthalmology and Vision Science, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA 92093, USA
| | - Mark H. Ellisman
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, Department of Neurosciences, University of California San Diego School of Medicine, La Jolla, CA 92093, USA
| | - Nicholas Marsh-Armstrong
- Department of Ophthalmology and Vision Science, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Lead contact
| |
Collapse
|
17
|
Cronin SJF, Yu W, Hale A, Licht-Mayer S, Crabtree MJ, Korecka JA, Tretiakov EO, Sealey-Cardona M, Somlyay M, Onji M, An M, Fox JD, Turnes BL, Gomez-Diaz C, da Luz Scheffer D, Cikes D, Nagy V, Weidinger A, Wolf A, Reither H, Chabloz A, Kavirayani A, Rao S, Andrews N, Latremoliere A, Costigan M, Douglas G, Freitas FC, Pifl C, Walz R, Konrat R, Mahad DJ, Koslov AV, Latini A, Isacson O, Harkany T, Hallett PJ, Bagby S, Woolf CJ, Channon KM, Je HS, Penninger JM. Crucial neuroprotective roles of the metabolite BH4 in dopaminergic neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.08.539795. [PMID: 37214873 PMCID: PMC10197517 DOI: 10.1101/2023.05.08.539795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Dopa-responsive dystonia (DRD) and Parkinson's disease (PD) are movement disorders caused by the dysfunction of nigrostriatal dopaminergic neurons. Identifying druggable pathways and biomarkers for guiding therapies is crucial due to the debilitating nature of these disorders. Recent genetic studies have identified variants of GTP cyclohydrolase-1 (GCH1), the rate-limiting enzyme in tetrahydrobiopterin (BH4) synthesis, as causative for these movement disorders. Here, we show that genetic and pharmacological inhibition of BH4 synthesis in mice and human midbrain-like organoids accurately recapitulates motor, behavioral and biochemical characteristics of these human diseases, with severity of the phenotype correlating with extent of BH4 deficiency. We also show that BH4 deficiency increases sensitivities to several PD-related stressors in mice and PD human cells, resulting in worse behavioral and physiological outcomes. Conversely, genetic and pharmacological augmentation of BH4 protects mice from genetically- and chemically induced PD-related stressors. Importantly, increasing BH4 levels also protects primary cells from PD-affected individuals and human midbrain-like organoids (hMLOs) from these stressors. Mechanistically, BH4 not only serves as an essential cofactor for dopamine synthesis, but also independently regulates tyrosine hydroxylase levels, protects against ferroptosis, scavenges mitochondrial ROS, maintains neuronal excitability and promotes mitochondrial ATP production, thereby enhancing mitochondrial fitness and cellular respiration in multiple preclinical PD animal models, human dopaminergic midbrain-like organoids and primary cells from PD-affected individuals. Our findings pinpoint the BH4 pathway as a key metabolic program at the intersection of multiple protective mechanisms for the health and function of midbrain dopaminergic neurons, identifying it as a potential therapeutic target for PD.
Collapse
Affiliation(s)
- Shane J F Cronin
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Weonjin Yu
- Signature Program in Neuroscience and Behavioural Disorders, Duke-National University of Singapore (NUS) Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Ashley Hale
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Simon Licht-Mayer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Mark J Crabtree
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Joanna A Korecka
- Neurodegeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA
| | - Evgenii O Tretiakov
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Marco Sealey-Cardona
- Department of Structural and Computational Biology, Max Perutz Labs, Vienna Biocenter Campus 5, 1030, Vienna, Austria
| | - Mate Somlyay
- Department of Structural and Computational Biology, Max Perutz Labs, Vienna Biocenter Campus 5, 1030, Vienna, Austria
| | - Masahiro Onji
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Meilin An
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Jesse D Fox
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Bruna Lenfers Turnes
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Carlos Gomez-Diaz
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Débora da Luz Scheffer
- LABOX, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC 88037-100, Brazil
| | - Domagoj Cikes
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Vanja Nagy
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases (LBI-RUD); Department of Neurology, Medical University of Vienna (MUW), 1090 Vienna, Austria
| | - Adelheid Weidinger
- Ludwig Boltzmann Institute for Traumatology. The Research Center in Cooperation with AUVA, Donaueschingen Str. 13, 1200 Vienna, Austria
| | - Alexandra Wolf
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Harald Reither
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Antoine Chabloz
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Anoop Kavirayani
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
| | - Shuan Rao
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Nick Andrews
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Alban Latremoliere
- Neurosurgery Department, Neurosurgery Pain Research Institute, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Michael Costigan
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Gillian Douglas
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | | | - Christian Pifl
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Roger Walz
- Center for Applied Neurocience, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil; Neurology Division, Internal Medicine Department, University Hospital of UFSC, Florianópolis, Brazil
| | - Robert Konrat
- Department of Structural and Computational Biology, Max Perutz Labs, Vienna Biocenter Campus 5, 1030, Vienna, Austria
| | - Don J Mahad
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Andrey V Koslov
- Ludwig Boltzmann Institute for Traumatology. The Research Center in Cooperation with AUVA, Donaueschingen Str. 13, 1200 Vienna, Austria
| | - Alexandra Latini
- LABOX, Departamento de Bioquímica, Universidade Federal de Santa Catarina, Florianópolis, SC 88037-100, Brazil
| | - Ole Isacson
- Neurodegeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of Neuroscience, Biomedicum 7D, Karolinska Institute, Solna, Sweden
| | - Penelope J Hallett
- Neurodegeneration Research Institute, Harvard Medical School/McLean Hospital, Belmont, MA, 02478, USA
| | - Stefan Bagby
- Department of Biology and Biochemistry and the Milner Centre for Evolution, University of Bath, Bath, UK
| | - Clifford J Woolf
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA and Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Keith M Channon
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Hyunsoo Shawn Je
- Signature Program in Neuroscience and Behavioural Disorders, Duke-National University of Singapore (NUS) Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| |
Collapse
|
18
|
Banarase TA, Sammeta SS, Wankhede NL, Mangrulkar SV, Rahangdale SR, Aglawe MM, Taksande BG, Upaganlawar AB, Umekar MJ, Kale MB. Mitophagy regulation in aging and neurodegenerative disease. Biophys Rev 2023; 15:239-255. [PMID: 37124925 PMCID: PMC10133433 DOI: 10.1007/s12551-023-01057-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 03/24/2023] [Indexed: 04/07/2023] Open
Abstract
Mitochondria are the primary cellular energy generators, supplying the majority of adenosine triphosphate through oxidative phosphorylation, which is necessary for neuron function and survival. Mitophagy is the metabolic process of eliminating dysfunctional or redundant mitochondria. It is a type of autophagy and it is crucial for maintaining mitochondrial and neuronal health. Impaired mitophagy leads to an accumulation of damaged mitochondria and proteins leading to the dysregulation of mitochondrial quality control processes. Recent research shows the vital role of mitophagy in neurons and the pathogenesis of major neurodegenerative diseases. Mitophagy also plays a major role in the process of aging. This review describes the alterations that are being caused in the mitophagy process at the molecular level in aging and in neurodegenerative diseases, particularly Alzheimer's, Parkinson's, and Huntington's diseases and amyotrophic lateral sclerosis, also looks at how mitophagy can be exploited as a therapeutic target for these diseases.
Collapse
Affiliation(s)
- Trupti A. Banarase
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Shivkumar S. Sammeta
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Nitu L. Wankhede
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Shubhada V. Mangrulkar
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Sandip R. Rahangdale
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Manish M. Aglawe
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Brijesh G. Taksande
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Aman B. Upaganlawar
- SNJB’s Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra India 423101
| | - Milind J. Umekar
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Mayur B. Kale
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| |
Collapse
|
19
|
Kikuchi S, Kohno T, Kojima T, Tatsumi H, Ohsaki Y, Ninomiya T. Oxygen-Glucose Deprivation Decreases the Motility and Length of Axonal Mitochondria in Cultured Dorsal Root Ganglion Cells of Rats. Cell Mol Neurobiol 2023; 43:1267-1280. [PMID: 35771293 PMCID: PMC11414435 DOI: 10.1007/s10571-022-01247-y] [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: 04/08/2022] [Accepted: 06/20/2022] [Indexed: 11/03/2022]
Abstract
Controlling axonal mitochondria is important for maintaining normal function of the neural network. Oxygen-glucose deprivation (OGD), a model used for mimicking ischemia, eventually induces neuronal cell death similar to axonal degeneration. Axonal mitochondria are disrupted during OGD-induced neural degeneration; however, the mechanism underlying mitochondrial dysfunction has not been completely understood. We focused on the dynamics of mitochondria in axons exposed to OGD; we observed that the number of motile mitochondria significantly reduced in 1 h following OGD exposure. In our observation, the decreased length of stationary mitochondria was affected by the following factors: first, the halt of motile mitochondria; second, the fission of longer stationary mitochondria; and third, a transformation from tubular to spherical shape in OGD-exposed axons. Motile mitochondria reduction preceded stationary mitochondria fragmentation in OGD exposure; these conditions induced the decrease of stationary mitochondria in three different ways. Our results suggest that mitochondrial morphological changes precede the axonal degeneration while ischemia-induced neurodegeneration.
Collapse
Affiliation(s)
- Shin Kikuchi
- Department of Anatomy 1, Sapporo Medical University School of Medicine, West 17, South 1, Chuo-ku, Sapporo, Hokkaido, 060-8556, Japan.
| | - Takayuki Kohno
- Department of Cell Sciences, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, West 17, South 1, Chuo-ku, Sapporo, Hokkaido, 060-8556, Japan
| | - Takashi Kojima
- Department of Cell Sciences, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, West 17, South 1, Chuo-ku, Sapporo, Hokkaido, 060-8556, Japan
| | - Haruyuki Tatsumi
- Department of Anatomy 1, Sapporo Medical University School of Medicine, West 17, South 1, Chuo-ku, Sapporo, Hokkaido, 060-8556, Japan
| | - Yuki Ohsaki
- Department of Anatomy 1, Sapporo Medical University School of Medicine, West 17, South 1, Chuo-ku, Sapporo, Hokkaido, 060-8556, Japan
| | - Takafumi Ninomiya
- Department of Anatomy 1, Sapporo Medical University School of Medicine, West 17, South 1, Chuo-ku, Sapporo, Hokkaido, 060-8556, Japan
| |
Collapse
|
20
|
Gautam M, Genç B, Helmold B, Ahrens A, Kuka J, Makrecka-Kuka M, Günay A, Koçak N, Aguilar-Wickings IR, Keefe D, Zheng G, Swaminathan S, Redmon M, Zariwala HA, Özdinler PH. SBT-272 improves TDP-43 pathology in ALS upper motor neurons by modulating mitochondrial integrity, motility, and function. Neurobiol Dis 2023; 178:106022. [PMID: 36716828 DOI: 10.1016/j.nbd.2023.106022] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/09/2023] [Accepted: 01/25/2023] [Indexed: 01/28/2023] Open
Abstract
Mitochondrial defects are one of the common underlying causes of neuronal vulnerability in neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS), and TDP-43 pathology is the most commonly observed proteinopathy. Disrupted inner mitochondrial membrane (IMM) reported in the upper motor neurons (UMNs) of ALS patients with TDP-43 pathology is recapitulated in the UMNs of well-characterized hTDP-43 mouse model of ALS. The construct validity, such as shared and common cellular pathology in mice and human, offers a unique opportunity to test treatment strategies that may translate to patients. SBT-272 is a well-tolerated brain-penetrant small molecule that stabilizes cardiolipin, a phospholipid found in IMM, thereby restoring mitochondrial structure and respiratory function. We investigated whether SBT-272 can improve IMM structure and health in UMNs diseased with TDP-43 pathology in our well-characterized UMN reporter line for ALS. We found that SBT-272 significantly improved mitochondrial structural integrity and restored mitochondrial motility and function. This led to improved health of diseased UMNs in vitro. In comparison to edaravone and AMX0035, SBT-272 appeared more effective in restoring health of diseased UMNs. Chronic treatment of SBT-272 for sixty days starting at an early symptomatic stage of the disease in vivo led to a significant reduction in astrogliosis, microgliosis, and TDP-43 pathology in the ALS motor cortex. Our results underscore the therapeutic potential of SBT-272, especially within the context of TDP-43 pathology and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Mukesh Gautam
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA
| | - Barış Genç
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA
| | - Benjamin Helmold
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA
| | - Angela Ahrens
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA
| | - Janis Kuka
- Latvian Institute of Organic Synthesis (LIOS), Aizkraukles Street 21, LV-2006 Riga, Latvia
| | - Marina Makrecka-Kuka
- Latvian Institute of Organic Synthesis (LIOS), Aizkraukles Street 21, LV-2006 Riga, Latvia
| | - Aksu Günay
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA
| | - Nuran Koçak
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA
| | - Izaak R Aguilar-Wickings
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA
| | - Dennis Keefe
- Stealth BioTherapeutics, 140 Kendrick St Building C, Needham, MA 02494, USA
| | - Guozhu Zheng
- Stealth BioTherapeutics, 140 Kendrick St Building C, Needham, MA 02494, USA
| | - Suchitra Swaminathan
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, 420 E Superior St, Chicago, IL 60611, USA.; Robert H. Lurie Comprehensive Cancer Research Center, Feinberg School of Medicine, Northwestern University, 675 N St Clair Fl 21 Ste 100, Chicago, IL 60611, USA
| | - Martin Redmon
- Stealth BioTherapeutics, 140 Kendrick St Building C, Needham, MA 02494, USA
| | - Hatim A Zariwala
- Stealth BioTherapeutics, 140 Kendrick St Building C, Needham, MA 02494, USA
| | - P Hande Özdinler
- Department of Neurology, Feinberg School of Medicine, Northwestern University, 303 E Chicago Ave, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Research Center, Feinberg School of Medicine, Northwestern University, 675 N St Clair Fl 21 Ste 100, Chicago, IL 60611, USA; Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Center for Developmental Therapeutics, Northwestern University, 2205 Tech Dr, Evanston, IL 60208, USA..
| |
Collapse
|
21
|
Yokota S, Shah SH, Huie EL, Wen RR, Luo Z, Goldberg JL. Kif5a Regulates Mitochondrial Transport in Developing Retinal Ganglion Cells In Vitro. Invest Ophthalmol Vis Sci 2023; 64:4. [PMID: 36862119 PMCID: PMC9983700 DOI: 10.1167/iovs.64.3.4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
Purpose Axon transport of organelles and neurotrophic factors is necessary for maintaining cellular function and survival of retinal ganglion cells (RGCs). However, it is not clear how trafficking of mitochondria, essential for RGC growth and maturation, changes during RGC development. The purpose of this study was to understand the dynamics and regulation of mitochondrial transport during RGC maturation using acutely purified RGCs as a model system. Methods Primary RGCs were immunopanned from rats of either sex during three stages of development. MitoTracker dye and live-cell imaging were used to quantify mitochondrial motility. Analysis of single-cell RNA sequencing was used to identify Kinesin family member 5A (Kif5a) as a relevant motor candidate for mitochondrial transport. Kif5a expression was manipulated with either short hairpin RNA (shRNA) or exogenous expression adeno-associated virus viral vectors. Results Anterograde and retrograde mitochondrial trafficking and motility decreased through RGC development. Similarly, the expression of Kif5a, a motor protein that transports mitochondria, also decreased during development. Kif5a knockdown decreased anterograde mitochondrial transport, while Kif5a expression increased general mitochondrial motility and anterograde mitochondrial transport. Conclusions Our results suggested that Kif5a directly regulates mitochondrial axonal transport in developing RGCs. Future work exploring the role of Kif5a in vivo in RGCs is indicated.
Collapse
Affiliation(s)
- Satoshi Yokota
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, California, United States.,Kobe City Eye Hospital, Kobe, Hyogo, Japan
| | - Sahil H Shah
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, California, United States
| | - Emma Lee Huie
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, California, United States
| | - Runxia Rain Wen
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, California, United States
| | - Ziming Luo
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, California, United States
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, California, United States
| |
Collapse
|
22
|
Optic Nerve Injury Enhanced Mitochondrial Fission and Increased Mitochondrial Density without Altering the Uniform Mitochondrial Distribution in the Unmyelinated Axons of Retinal Ganglion Cells in a Mouse Model. Int J Mol Sci 2023; 24:ijms24054356. [PMID: 36901786 PMCID: PMC10002508 DOI: 10.3390/ijms24054356] [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: 01/27/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Glaucomatous optic neuropathy (GON), a major cause of blindness, is characterized by the loss of retinal ganglion cells (RGCs) and the degeneration of their axons. Mitochondria are deeply involved in maintaining the health of RGCs and their axons. Therefore, lots of attempts have been made to develop diagnostic tools and therapies targeting mitochondria. Recently, we reported that mitochondria are uniformly distributed in the unmyelinated axons of RGCs, possibly owing to the ATP gradient. Thus, using transgenic mice expressing yellow fluorescent protein targeting mitochondria exclusively in RGCs within the retina, we assessed the alteration of mitochondrial distributions induced by optic nerve crush (ONC) via in vitro flat-mount retinal sections and in vivo fundus images captured with a confocal scanning ophthalmoscope. We observed that the mitochondrial distribution in the unmyelinated axons of survived RGCs after ONC remained uniform, although their density increased. Furthermore, via in vitro analysis, we discovered that the mitochondrial size is attenuated following ONC. These results suggest that ONC induces mitochondrial fission without disrupting the uniform mitochondrial distribution, possibly preventing axonal degeneration and apoptosis. The in vivo visualization system of axonal mitochondria in RGCs may be applicable in the detection of the progression of GON in animal studies and potentially in humans.
Collapse
|
23
|
Whitehall JC, Smith ALM, Greaves LC. Mitochondrial DNA Mutations and Ageing. Subcell Biochem 2023; 102:77-98. [PMID: 36600130 DOI: 10.1007/978-3-031-21410-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Mitochondria are subcellular organelles present in most eukaryotic cells which play a significant role in numerous aspects of cell biology. These include carbohydrate and fatty acid metabolism to generate cellular energy through oxidative phosphorylation, apoptosis, cell signalling, haem biosynthesis and reactive oxygen species production. Mitochondrial dysfunction is a feature of many human ageing tissues, and since the discovery that mitochondrial DNA mutations were a major underlying cause of changes in oxidative phosphorylation capacity, it has been proposed that they have a role in human ageing. However, there is still much debate on whether mitochondrial DNA mutations play a causal role in ageing or are simply a consequence of the ageing process. This chapter describes the structure of mammalian mitochondria, and the unique features of mitochondrial genetics, and reviews the current evidence surrounding the role of mitochondrial DNA mutations in the ageing process. It then focusses on more recent discoveries regarding the role of mitochondrial dysfunction in stem cell ageing and age-related inflammation.
Collapse
Affiliation(s)
- Julia C Whitehall
- Wellcome Centre for Mitochondrial Research, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Anna L M Smith
- Wellcome Centre for Mitochondrial Research, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK
| | - Laura C Greaves
- Wellcome Centre for Mitochondrial Research, Biosciences Institute, Newcastle University, Newcastle Upon Tyne, UK.
| |
Collapse
|
24
|
Hypoxia-Inducible Factor 1 and Mitochondria: An Intimate Connection. Biomolecules 2022; 13:biom13010050. [PMID: 36671435 PMCID: PMC9855368 DOI: 10.3390/biom13010050] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
The general objective of the review is to explain the interaction between HIF-1 and mitochondria. On the one hand, this review describes the effects of HIF-1 on mitochondrial structure, including quantity, distribution, and morphology, as well as on mitochondrial metabolism and respiratory function. On the other hand, various factors, including mitochondrial activation of enzymes, the respiratory chain, complex and decoupling proteins, affect the stability and activity of HIF-1. It is possible to develop future molecular therapeutic interventions by understanding the interrelationships between HIF-1 and mitochondria.
Collapse
|
25
|
Jurcău MC, Andronie-Cioara FL, Jurcău A, Marcu F, Ţiț DM, Pașcalău N, Nistor-Cseppentö DC. The Link between Oxidative Stress, Mitochondrial Dysfunction and Neuroinflammation in the Pathophysiology of Alzheimer's Disease: Therapeutic Implications and Future Perspectives. Antioxidants (Basel) 2022; 11:2167. [PMID: 36358538 PMCID: PMC9686795 DOI: 10.3390/antiox11112167] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 08/26/2023] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia, has increasing incidence, increasing mortality rates, and poses a huge burden on healthcare. None of the currently approved drugs for the treatment of AD influence disease progression. Many clinical trials aiming at inhibiting amyloid plaque formation, increasing amyloid beta clearance, or inhibiting neurofibrillary tangle pathology yielded inconclusive results or failed. Meanwhile, research has identified many interlinked vicious cascades implicating oxidative stress, mitochondrial dysfunction, and chronic neuroinflammation, and has pointed to novel therapeutic targets such as improving mitochondrial bioenergetics and quality control, diminishing oxidative stress, or modulating the neuroinflammatory pathways. Many novel molecules tested in vitro or in animal models have proven efficient, but their translation into clinic needs further research regarding appropriate doses, delivery routes, and possible side effects. Cell-based therapies and extracellular vesicle-mediated delivery of messenger RNAs and microRNAs seem also promising strategies allowing to target specific signaling pathways, but need further research regarding the most appropriate harvesting and culture methods as well as control of the possible tumorigenic side effects. The rapidly developing area of nanotechnology could improve drug delivery and also be used in early diagnosis.
Collapse
Affiliation(s)
| | - Felicia Liana Andronie-Cioara
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Anamaria Jurcău
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Florin Marcu
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Delia Mirela Ţiț
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| | - Nicoleta Pașcalău
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Delia Carmen Nistor-Cseppentö
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| |
Collapse
|
26
|
Sabui A, Biswas M, Somvanshi PR, Kandagiri P, Gorla M, Mohammed F, Tammineni P. Decreased anterograde transport coupled with sustained retrograde transport contributes to reduced axonal mitochondrial density in tauopathy neurons. Front Mol Neurosci 2022; 15:927195. [PMID: 36245925 PMCID: PMC9561864 DOI: 10.3389/fnmol.2022.927195] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Mitochondria are essential organelle required for neuronal homeostasis. Mitochondria supply ATP and buffer calcium at synaptic terminals. However, the complex structural geometry of neurons poses a unique challenge in transporting mitochondria to synaptic terminals. Kinesin motors supply mitochondria to the axonal compartments, while cytoplasmic dynein is required for retrograde transport. Despite the importance of presynaptic mitochondria, how and whether axonal mitochondrial transport and distribution are altered in tauopathy neurons remain poorly studied. In the current study, we have shown that anterograde transport of mitochondria is reduced in P301L neurons, while there is no change in the retrograde transport. Consistently, axonal mitochondrial abundance is reduced in P301L neurons. We further studied the possible role of two opposing motor proteins on mitochondrial transport and found that mitochondrial association of kinesin is decreased significantly in P301L cells. Interestingly, fitting our experimental data into mathematical equations suggested a possible rise in dynein activity to maintain retrograde flux in P301L cells. Our data indicate that decreased kinesin-mediated transport coupled with sustained retrograde transport might reduce axonal mitochondria in tauopathy neurons, thus contributing to the synaptic deficits in Alzheimer’s disease (AD) and other tauopathies.
Collapse
Affiliation(s)
- Anusruti Sabui
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Mitali Biswas
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | | | - Preethi Kandagiri
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Madhavi Gorla
- Centre for Biotechnology, Institute of Science and Technology (IST), Jawaharlal Nehru Technological University Hyderabad, Hyderabad, India
| | - Fareed Mohammed
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Prasad Tammineni
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, India
- *Correspondence: Prasad Tammineni,
| |
Collapse
|
27
|
Abstract
The maintenance of a healthy mitochondrial network and the ability to adjust organelle population in response to internal or external stimuli are essential for the function and the survival of eukaryotic cells. Over the last two decades several studies have demonstrated the paramount importance of mitophagy, a selective form of autophagy that removes damaged and/or superfluous organelles, in organismal physiology. Post-mitotic neuronal cells are particularly vulnerable to mitochondrial damage, and mitophagy impairment has emerged as a causative factor in multiple neurodegenerative pathologies, including Alzheimer's disease and Parkinson's disease among others. Although mitochondrial turnover is a multifaceted process, neurons have to tackle additional complications, arising from their pronounced bioenergetic demands and their unique architecture and cellular polarisation that render the degradation of distal organelles challenging. Mounting evidence indicates that despite the functional conservation of mitophagy pathways, the unique features of neuronal physiology have led to the adaptation of compartmentalised solutions, which serve to ensure seamless mitochondrial removal in every part of the cell. In this review, we summarise the current knowledge concerning the molecular mechanisms that mediate mitophagy compartmentalisation and discuss their implications in various human pathologies.
Collapse
|
28
|
Quintero H, Shiga Y, Belforte N, Alarcon-Martinez L, El Hajji S, Villafranca-Baughman D, Dotigny F, Di Polo A. Restoration of mitochondria axonal transport by adaptor Disc1 supplementation prevents neurodegeneration and rescues visual function. Cell Rep 2022; 40:111324. [PMID: 36103832 DOI: 10.1016/j.celrep.2022.111324] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/01/2022] [Accepted: 08/17/2022] [Indexed: 11/03/2022] Open
Abstract
Deficits in mitochondrial transport are a common feature of neurodegenerative diseases. We investigated whether loss of components of the mitochondrial transport machinery impinge directly on metabolic stress, neuronal death, and circuit dysfunction. Using multiphoton microscope live imaging, we showed that ocular hypertension, a major risk factor in glaucoma, disrupts mitochondria anterograde axonal transport leading to energy decline in vulnerable neurons. Gene- and protein-expression analysis revealed loss of the adaptor disrupted in schizophrenia 1 (Disc1) in retinal neurons subjected to high intraocular pressure. Disc1 gene delivery was sufficient to rescue anterograde transport and replenish axonal mitochondria. A genetically encoded ATP sensor combined with longitudinal live imaging showed that Disc1 supplementation increased ATP production in stressed neurons. Disc1 gene therapy promotes neuronal survival, reverses abnormal single-cell calcium dynamics, and restores visual responses. Our study demonstrates that enhancing anterograde mitochondrial transport is an effective strategy to alleviate metabolic stress and neurodegeneration.
Collapse
Affiliation(s)
- Heberto Quintero
- Department of Neuroscience, University of Montreal, PO Box 6128, Station Centre-ville, Montreal, QC H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC H2X 0A9, Canada
| | - Yukihiro Shiga
- Department of Neuroscience, University of Montreal, PO Box 6128, Station Centre-ville, Montreal, QC H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC H2X 0A9, Canada
| | - Nicolas Belforte
- Department of Neuroscience, University of Montreal, PO Box 6128, Station Centre-ville, Montreal, QC H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC H2X 0A9, Canada
| | - Luis Alarcon-Martinez
- Department of Neuroscience, University of Montreal, PO Box 6128, Station Centre-ville, Montreal, QC H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC H2X 0A9, Canada
| | - Sana El Hajji
- Department of Neuroscience, University of Montreal, PO Box 6128, Station Centre-ville, Montreal, QC H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC H2X 0A9, Canada
| | - Deborah Villafranca-Baughman
- Department of Neuroscience, University of Montreal, PO Box 6128, Station Centre-ville, Montreal, QC H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC H2X 0A9, Canada
| | - Florence Dotigny
- Department of Neuroscience, University of Montreal, PO Box 6128, Station Centre-ville, Montreal, QC H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC H2X 0A9, Canada
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, PO Box 6128, Station Centre-ville, Montreal, QC H3C 3J7, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC H2X 0A9, Canada.
| |
Collapse
|
29
|
Imbriani P, Martella G, Bonsi P, Pisani A. Oxidative stress and synaptic dysfunction in rodent models of Parkinson's disease. Neurobiol Dis 2022; 173:105851. [PMID: 36007757 DOI: 10.1016/j.nbd.2022.105851] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 08/02/2022] [Accepted: 08/20/2022] [Indexed: 11/26/2022] Open
Abstract
Parkinson's disease (PD) is a multifactorial disorder involving a complex interplay between a variety of genetic and environmental factors. In this scenario, mitochondrial impairment and oxidative stress are widely accepted as crucial neuropathogenic mechanisms, as also evidenced by the identification of PD-associated genes that are directly involved in mitochondrial function. The concept of mitochondrial dysfunction is closely linked to that of synaptic dysfunction. Indeed, compelling evidence supports the role of mitochondria in synaptic transmission and plasticity, although many aspects have not yet been fully elucidated. Here, we will provide a brief overview of the most relevant evidence obtained in different neurotoxin-based and genetic rodent models of PD, focusing on mitochondrial impairment and synaptopathy, an early central event preceding overt nigrostriatal neurodegeneration. The identification of early deficits occurring in PD pathogenesis is crucial in view of the development of potential disease-modifying therapeutic strategies.
Collapse
Affiliation(s)
- Paola Imbriani
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Giuseppina Martella
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Paola Bonsi
- Laboratory of Neurophysiology and Plasticity, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Antonio Pisani
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; IRCCS Mondino Foundation, Pavia, Italy.
| |
Collapse
|
30
|
Harbauer AB, Hees JT, Wanderoy S, Segura I, Gibbs W, Cheng Y, Ordonez M, Cai Z, Cartoni R, Ashrafi G, Wang C, Perocchi F, He Z, Schwarz TL. Neuronal mitochondria transport Pink1 mRNA via synaptojanin 2 to support local mitophagy. Neuron 2022; 110:1516-1531.e9. [PMID: 35216662 PMCID: PMC9081165 DOI: 10.1016/j.neuron.2022.01.035] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 06/25/2021] [Accepted: 01/28/2022] [Indexed: 01/18/2023]
Abstract
PTEN-induced kinase 1 (PINK1) is a short-lived protein required for the removal of damaged mitochondria through Parkin translocation and mitophagy. Because the short half-life of PINK1 limits its ability to be trafficked into neurites, local translation is required for this mitophagy pathway to be active far from the soma. The Pink1 transcript is associated and cotransported with neuronal mitochondria. In concert with translation, the mitochondrial outer membrane proteins synaptojanin 2 binding protein (SYNJ2BP) and synaptojanin 2 (SYNJ2) are required for tethering Pink1 mRNA to mitochondria via an RNA-binding domain in SYNJ2. This neuron-specific adaptation for the local translation of PINK1 provides distal mitochondria with a continuous supply of PINK1 for the activation of mitophagy.
Collapse
Affiliation(s)
- Angelika B Harbauer
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; Max Planck Institute of Neurobiology, Am Klopferspitz 18, 82152 Martinsried, Germany; Institute of Neuronal Cell Biology, Technical University of Munich, Biedersteiner Straße 29, 80802 Munich, Germany; Munich Cluster of Systems Neurology, Feodor-Lynen-Straße 17, 81377 Munich, Germany.
| | - J Tabitha Hees
- Max Planck Institute of Neurobiology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Simone Wanderoy
- Max Planck Institute of Neurobiology, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Inmaculada Segura
- Max Planck Institute of Neurobiology, Am Klopferspitz 18, 82152 Martinsried, Germany; Ludwig-Maximilians-Universität München, Department of Cellular Physiology Biomedical Center Munich - BMC, Großhaderner Str. 9, 82152 Martinsried, Germany
| | - Whitney Gibbs
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Yiming Cheng
- Munich Cluster of Systems Neurology, Feodor-Lynen-Straße 17, 81377 Munich, Germany; Institute for Diabetes and Obesity, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Munich, Germany
| | - Martha Ordonez
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Zerong Cai
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Romain Cartoni
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Ghazaleh Ashrafi
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Chen Wang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Fabiana Perocchi
- Institute of Neuronal Cell Biology, Technical University of Munich, Biedersteiner Straße 29, 80802 Munich, Germany; Munich Cluster of Systems Neurology, Feodor-Lynen-Straße 17, 81377 Munich, Germany; Institute for Diabetes and Obesity, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Munich, Germany
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Thomas L Schwarz
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA; Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
31
|
Reiss AB, Ahmed S, Dayaramani C, Glass AD, Gomolin IH, Pinkhasov A, Stecker MM, Wisniewski T, De Leon J. The role of mitochondrial dysfunction in Alzheimer's disease: A potential pathway to treatment. Exp Gerontol 2022; 164:111828. [DOI: 10.1016/j.exger.2022.111828] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/15/2022] [Accepted: 04/27/2022] [Indexed: 11/17/2022]
|
32
|
Slater PG, Domínguez-Romero ME, Villarreal M, Eisner V, Larraín J. Mitochondrial function in spinal cord injury and regeneration. Cell Mol Life Sci 2022; 79:239. [PMID: 35416520 PMCID: PMC11072423 DOI: 10.1007/s00018-022-04261-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 12/21/2022]
Abstract
Many people around the world suffer from some form of paralysis caused by spinal cord injury (SCI), which has an impact on quality and life expectancy. The spinal cord is part of the central nervous system (CNS), which in mammals is unable to regenerate, and to date, there is a lack of full functional recovery therapies for SCI. These injuries start with a rapid and mechanical insult, followed by a secondary phase leading progressively to greater damage. This secondary phase can be potentially modifiable through targeted therapies. The growing literature, derived from mammalian and regenerative model studies, supports a leading role for mitochondria in every cellular response after SCI: mitochondrial dysfunction is the common event of different triggers leading to cell death, cellular metabolism regulates the immune response, mitochondrial number and localization correlate with axon regenerative capacity, while mitochondrial abundance and substrate utilization regulate neural stem progenitor cells self-renewal and differentiation. Herein, we present a comprehensive review of the cellular responses during the secondary phase of SCI, the mitochondrial contribution to each of them, as well as evidence of mitochondrial involvement in spinal cord regeneration, suggesting that a more in-depth study of mitochondrial function and regulation is needed to identify potential targets for SCI therapeutic intervention.
Collapse
Affiliation(s)
- Paula G Slater
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile.
| | - Miguel E Domínguez-Romero
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| | - Maximiliano Villarreal
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| | - Verónica Eisner
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| | - Juan Larraín
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| |
Collapse
|
33
|
Jeong YY, Han S, Jia N, Zhang M, Sheshadri P, Tammineni P, Cheung J, Nissenbaum M, Baskar SS, Kwan K, Margolis DJ, Jiang P, Kusnecov AW, Cai Q. Broad activation of the Parkin pathway induces synaptic mitochondrial deficits in early tauopathy. Brain 2022; 145:305-323. [PMID: 35022692 PMCID: PMC8967101 DOI: 10.1093/brain/awab243] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/20/2021] [Accepted: 06/17/2021] [Indexed: 01/18/2023] Open
Abstract
Mitochondrial defects are a hallmark of early pathophysiology in Alzheimer's disease, with pathologically phosphorylated tau reported to induce mitochondrial toxicity. Mitophagy constitutes a key pathway in mitochondrial quality control by which damaged mitochondria are targeted for autophagy. However, few details are known regarding the intersection of mitophagy and pathologies in tauopathy. Here, by applying biochemical and cell biological approaches including time-lapse confocal imaging in live tauopathy neurons, combined with gene rescue experiments via stereotactic injections of adeno-associated virus particles into tauopathy mouse brains, electrophysiological recordings and behavioural tests, we demonstrate for the first time that mitochondrial distribution deficits at presynaptic terminals are an early pathological feature in tauopathy brains. Furthermore, Parkin-mediated mitophagy is extensively activated in tauopathy neurons, which accelerates mitochondrial Rho GTPase 1 (Miro1) turnover and consequently halts Miro1-mediated mitochondrial anterograde movement towards synaptic terminals. As a result, mitochondrial supply at tauopathy synapses is disrupted, impairing synaptic function. Strikingly, increasing Miro1 levels restores the synaptic mitochondrial population by enhancing mitochondrial anterograde movement and thus reverses tauopathy-associated synaptic failure. In tauopathy mouse brains, overexpression of Miro1 markedly elevates synaptic distribution of mitochondria and protects against synaptic damage and neurodegeneration, thereby counteracting impairments in learning and memory as well as synaptic plasticity. Taken together, our study reveals that activation of the Parkin pathway triggers an unexpected effect-depletion of mitochondria from synaptic terminals, a characteristic feature of early tauopathy. We further provide new mechanistic insights into how parkin activation-enhanced Miro1 degradation and impaired mitochondrial anterograde transport drive tauopathy-linked synaptic pathogenesis and establish a foundation for future investigations into new therapeutic strategies to prevent synaptic deterioration in Alzheimer's disease and other tauopathies.
Collapse
Affiliation(s)
- Yu Young Jeong
- Division of Life Science, Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Sinsuk Han
- Division of Life Science, Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Nuo Jia
- Division of Life Science, Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Mingyang Zhang
- Division of Life Science, Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Preethi Sheshadri
- Division of Life Science, Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Prasad Tammineni
- Division of Life Science, Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Jasmine Cheung
- Division of Life Science, Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Marialaina Nissenbaum
- Department of Psychology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Sindhuja S Baskar
- Division of Life Science, Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Kelvin Kwan
- Division of Life Science, Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - David J Margolis
- Division of Life Science, Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Peng Jiang
- Division of Life Science, Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Alexander W. Kusnecov
- Department of Psychology, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Qian Cai
- Division of Life Science, Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
34
|
Matsumoto N, Hori I, Kajita MK, Murase T, Nakamura W, Tsuji T, Miyake S, Inatani M, Konishi Y. Intermitochondrial signaling regulates the uniform distribution of stationary mitochondria in axons. Mol Cell Neurosci 2022; 119:103704. [DOI: 10.1016/j.mcn.2022.103704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/27/2021] [Accepted: 01/31/2022] [Indexed: 10/19/2022] Open
|
35
|
Badal K, Zhao Y, Miller KE, Puthanveettil SV. Live Imaging and Quantitative Analysis of Organelle Transport in Sensory Neurons of Aplysia Californica. Methods Mol Biol 2022; 2431:23-48. [PMID: 35412270 DOI: 10.1007/978-1-0716-1990-2_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Axonal transport moves proteins, RNAs, and organelles between the soma and synapses to support synaptic function and activity-dependent changes in synaptic strength. This transport is impaired in several neurodegenerative disorders such as Alzheimer's disease. Thus, it is critical to understand the regulation and underlying mechanisms of the transport process. Aplysia californica provides a powerful experimental system for studying the interplay between synaptic activity and transport because its defined synaptic circuits can be built in-vitro. Advantages include precise pre- and postsynaptic manipulation, and high-resolution imaging of axonal transport. Here, we describe methodologies for the quantitative analysis of axonal transport in Aplysia sensory neurons.
Collapse
Affiliation(s)
- Kerriann Badal
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida, Jupiter, FL, USA
- Integrated Biology Program, Florida Atlantic University, Jupiter, FL, USA
| | - Yibo Zhao
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida, Jupiter, FL, USA
| | - Kyle E Miller
- Department of Integrative Biology, Michigan State University, East Lansing, MI, USA.
| | | |
Collapse
|
36
|
Zaninello M, Palikaras K, Sotiriou A, Tavernarakis N, Scorrano L. Sustained intracellular calcium rise mediates neuronal mitophagy in models of autosomal dominant optic atrophy. Cell Death Differ 2022; 29:167-177. [PMID: 34389813 PMCID: PMC8738763 DOI: 10.1038/s41418-021-00847-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial dysfunction and mitophagy are often hallmarks of neurodegenerative diseases such as autosomal dominant optic atrophy (ADOA) caused by mutations in the key mitochondrial dynamics protein optic atrophy 1 (Opa1). However, the second messengers linking mitochondrial dysfunction to initiation of mitophagy remain poorly characterized. Here, we show in mammalian and nematode neurons that Opa1 mutations trigger Ca2+-dependent mitophagy. Deletion or expression of mutated Opa1 in mouse retinal ganglion cells and Caenorhabditis elegans motor neurons lead to mitochondrial dysfunction, increased cytosolic Ca2+ levels, and decreased axonal mitochondrial density. Chelation of Ca2+ restores mitochondrial density in neuronal processes, neuronal function, and viability. Mechanistically, sustained Ca2+ levels activate calcineurin and AMPK, placed in the same genetic pathway regulating axonal mitochondrial density. Our data reveal that mitophagy in ADOA depends on Ca2+-calcineurin-AMPK signaling cascade.
Collapse
Affiliation(s)
- Marta Zaninello
- grid.428736.cVeneto Institute of Molecular Medicine, Padova, Italy ,grid.5608.b0000 0004 1757 3470Department of Biology, University of Padova, Padova, Italy ,grid.6190.e0000 0000 8580 3777Present Address: Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Konstantinos Palikaras
- grid.5216.00000 0001 2155 0800Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Aggeliki Sotiriou
- grid.4834.b0000 0004 0635 685XInstitute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete Greece
| | - Nektarios Tavernarakis
- grid.4834.b0000 0004 0635 685XInstitute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete Greece ,grid.8127.c0000 0004 0576 3437Department of Basic Sciences, School of Medicine, University of Crete, Heraklion, Crete Greece
| | - Luca Scorrano
- grid.428736.cVeneto Institute of Molecular Medicine, Padova, Italy ,grid.5608.b0000 0004 1757 3470Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
37
|
Corrigan RR, Piontkivska H, Casadesus G. Amylin Pharmacology in Alzheimer's Disease Pathogenesis and Treatment. Curr Neuropharmacol 2022; 20:1894-1907. [PMID: 34852745 PMCID: PMC9886804 DOI: 10.2174/1570159x19666211201093147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/12/2021] [Accepted: 11/26/2021] [Indexed: 11/22/2022] Open
Abstract
The metabolic peptide hormone amylin, in concert with other metabolic peptides like insulin and leptin, has an important role in metabolic homeostasis and has been intimately linked to Alzheimer's disease (AD). Interestingly, this pancreatic amyloid peptide is known to self-aggregate much like amyloid-beta and has been reported to be a source of pathogenesis in both Type II diabetes mellitus (T2DM) and Alzheimer's disease. The traditional "gain of toxic function" properties assigned to amyloid proteins are, however, contrasted by several reports highlighting neuroprotective effects of amylin and a recombinant analog, pramlintide, in the context of these two diseases. This suggests that pharmacological therapies aimed at modulating the amylin receptor may be therapeutically beneficial for AD development, as they already are for T2DMM. However, the nature of amylin receptor signaling is highly complex and not well studied in the context of CNS function. Therefore, to begin to address this pharmacological paradox in amylin research, the goal of this review is to summarize the current research on amylin signaling and CNS functions and critically address the paradoxical nature of this hormone's signaling in the context of AD pathogenesis.
Collapse
Affiliation(s)
| | | | - Gemma Casadesus
- Address correspondence to this author at the Department of Pharmacology and Therapeutics, University of Florida, PO Box 100495. Gainesville, FL32610 USA; Tel: 352-294-5346; E-mail:
| |
Collapse
|
38
|
Smith SA, Ogawa SA, Chau L, Whelan KA, Hamilton KE, Chen J, Tan L, Chen EZ, Keilbaugh S, Fogt F, Bewtra M, Braun J, Xavier RJ, Clish CB, Slaff B, Weljie AM, Bushman FD, Lewis JD, Li H, Master SR, Bennett MJ, Nakagawa H, Wu GD. Mitochondrial dysfunction in inflammatory bowel disease alters intestinal epithelial metabolism of hepatic acylcarnitines. J Clin Invest 2021; 131:133371. [PMID: 33141762 DOI: 10.1172/jci133371] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 10/09/2020] [Indexed: 12/26/2022] Open
Abstract
As the interface between the gut microbiota and the mucosal immune system, there has been great interest in the maintenance of colonic epithelial integrity through mitochondrial oxidation of butyrate, a short-chain fatty acid produced by the gut microbiota. Herein, we showed that the intestinal epithelium could also oxidize long-chain fatty acids, and that luminally delivered acylcarnitines in bile could be consumed via apical absorption by the intestinal epithelium, resulting in mitochondrial oxidation. Finally, intestinal inflammation led to mitochondrial dysfunction in the apical domain of the surface epithelium that may reduce the consumption of fatty acids, contributing to higher concentrations of fecal acylcarnitines in murine Citrobacter rodentium-induced colitis and human inflammatory bowel disease. These results emphasized the importance of both the gut microbiota and the liver in the delivery of energy substrates for mitochondrial metabolism by the intestinal epithelium.
Collapse
Affiliation(s)
- Sarah A Smith
- Department of Medicine, Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sayaka A Ogawa
- Department of Medicine, Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lillian Chau
- Department of Medicine, Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kelly A Whelan
- Fels Institute for Cancer Research and Molecular Biology, Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Kathryn E Hamilton
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jie Chen
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lu Tan
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Eric Z Chen
- Department of Informatics, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sue Keilbaugh
- Department of Medicine, Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Franz Fogt
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Meenakshi Bewtra
- Department of Medicine, Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jonathan Braun
- Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ramnik J Xavier
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard University, Cambridge, Massachusetts, USA.,Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Clary B Clish
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard University, Cambridge, Massachusetts, USA
| | - Barry Slaff
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Aalim M Weljie
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James D Lewis
- Department of Medicine, Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hongzhe Li
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Stephen R Master
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Michael J Bennett
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hiroshi Nakagawa
- Division of Digestive and Liver Diseases, Department of Medicine, Herbert Irving Comprehensive Cancer Research Center, Columbia University Irving Medical Center, New York, New York, USA
| | - Gary D Wu
- Department of Medicine, Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
39
|
Franco JA. Under Pressure: A Microfluidic Chip for Prolonged, Anesthetic-Free Imaging of Neuronal Mitostasis in Caenorhabditis elegans. eNeuro 2021; 8:ENEURO.0304-21.2021. [PMID: 34475223 PMCID: PMC8422848 DOI: 10.1523/eneuro.0304-21.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Highlighted Research Paper: Tracking Mitochondrial Density and Positioning along a Growing Neuronal Process in Individual C. elegans Neuron Using a Long-Term Growth and Imaging Microfluidic Device by Sudip Mondal, Jyoti Dubey, Anjali Awasthi, Guruprasad Reddy Sure, Amruta Vasudevan, and Sandhya P. Koushika.
Collapse
Affiliation(s)
- Joy A Franco
- Department of Mechanical Engineering, Stanford University, Stanford, California 94305
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305
| |
Collapse
|
40
|
Otomo A, Ono S, Sato K, Mitsui S, Shimakura K, Kimura H, Hadano S. High-throughput quantitative analysis of axonal transport in cultured neurons from SOD1 H46R ALS mice by using a microfluidic device. Neurosci Res 2021; 174:46-52. [PMID: 34352295 DOI: 10.1016/j.neures.2021.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 07/15/2021] [Accepted: 07/30/2021] [Indexed: 11/15/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by selective loss of motor neurons. We have previously shown that autophagosome-like vesicular structures are progressively accumulated in the spinal axons of an ALS mouse model, overexpressing human Cu/Zn superoxide dismutase (SOD1) mutant, prior to the onset of motor symptoms. This suggests that axonal transport perturbation can be an early sign of neuronal dysfunction. However, the exact causal relationship between axonal transport deficits and neurodegeneration is not fully understood. To clarify whether axonal transport of organelles even in neurons at early developmental stages was affected by overexpression of mutant SOD1, we conducted a microfluidic device-based high-throughput quantitative analysis of the axonal transport of acidic vesicles and mitochondria in primary cultured cortical neurons established from SOD1H46R transgenic mice. Compared to wild-type (WT), a significantly increased number of motile acidic vesicles, i.e., autophagosomes and/or late-endosomes, was observed in the axons of SOD1H46R neurons. By contrast, mitochondria moving along the axons were significantly decreased in SOD1H46R compared to WT. Since such phenotypes, where the axonal transport of these organelles is differently affected by mutant SOD1 expression, emerge before axonal degeneration, axonal transport deficits could dysregulate axon homeostasis, thereby ultimately accelerating neurodegeneration.
Collapse
Affiliation(s)
- Asako Otomo
- Molecular Neuropathobiology Laboratory, Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan; Micro/Nano Technology Center, Tokai University, Hiratsuka, Kanagawa, 259-1292, Japan
| | - Suzuka Ono
- Molecular Neuropathobiology Laboratory, Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Kai Sato
- Molecular Neuropathobiology Laboratory, Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Shun Mitsui
- Molecular Neuropathobiology Laboratory, Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Kento Shimakura
- Molecular Neuropathobiology Laboratory, Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Hiroshi Kimura
- Micro/Nano Technology Center, Tokai University, Hiratsuka, Kanagawa, 259-1292, Japan; Department of Mechanical Engineering, School of Engineering, Tokai University, Hiratsuka, Kanagawa, 259-1292, Japan.
| | - Shinji Hadano
- Molecular Neuropathobiology Laboratory, Department of Molecular Life Sciences, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan; Micro/Nano Technology Center, Tokai University, Hiratsuka, Kanagawa, 259-1292, Japan; The Institute of Medical Sciences, Tokai University, Isehara, Kanagawa, 259-1193, Japan; Research Center for Brain and Nervous Diseases, Tokai University Graduate School of Medicine, Isehara, Kanagawa, 259-1193, Japan.
| |
Collapse
|
41
|
Aghanoori MR, Margulets V, Smith DR, Kirshenbaum LA, Gitler D, Fernyhough P. Sensory neurons derived from diabetic rats exhibit deficits in functional glycolysis and ATP that are ameliorated by IGF-1. Mol Metab 2021; 49:101191. [PMID: 33592336 PMCID: PMC7940986 DOI: 10.1016/j.molmet.2021.101191] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE The distal dying-back of the longest nerve fibres is a hallmark of diabetic neuropathy, and impaired provision of energy in the form of adenosine triphosphate (ATP) may contribute to this neurodegenerative process. We hypothesised that energy supplementation via glycolysis and/or mitochondrial oxidative phosphorylation is compromised in cultured dorsal root ganglion (DRG) sensory neurons from diabetic rodents, thus contributing to axonal degeneration. Functional analysis of glycolysis and mitochondrial respiration and real-time measurement of ATP levels in live cells were our specific means to test this hypothesis. METHODS DRG neuron cultures from age-matched control or streptozotocin (STZ)-induced type 1 diabetic rats were used for in vitro studies. Three plasmids containing ATP biosensors of varying affinities were transfected into neurons to study endogenous ATP levels in real time. The Seahorse XF analyser was used for glycolysis and mitochondrial respiration measurements. RESULTS Fluorescence resonance energy transfer (FRET) efficiency (YFP/CFP ratio) of the ATP biosensors AT1.03 (low affinity) and AT1.03YEMK (medium affinity) were significantly higher than that measured using the ATP-insensitive construct AT1.03R122/6K in both cell bodies and neurites of DRG neurons (p < 0.0001). The ATP level was homogenous along the axons but higher in cell bodies in cultured DRG neurons from both control and diabetic rats. Treatment with oligomycin (an ATP synthase inhibitor in mitochondria) decreased the ATP levels in cultured DRG neurons. Likewise, blockade of glycolysis using 2-deoxy-d-glucose (2-DG: a glucose analogue) reduced ATP levels (p < 0.001). Cultured DRG neurons derived from diabetic rats showed a diminishment of ATP levels (p < 0.01), glycolytic capacity, glycolytic reserve and non-glycolytic acidification. Application of insulin-like growth factor-1 (IGF-1) significantly elevated all the above parameters in DRG neurons from diabetic rats. Oligomycin pre-treatment of DRG neurons, to block oxidative phosphorylation, depleted the glycolytic reserve and lowered basal respiration in sensory neurons derived from control and diabetic rats. Depletion was much higher in sensory neurons from diabetic rats compared to control rats. In addition, an acute increase in glucose concentration, in the presence or absence of oligomycin, elevated parameters of glycolysis by 1.5- to 2-fold while having no impact on mitochondrial respiration. CONCLUSION We provide the first functional evidence for decreased glycolytic capacity in DRG neurons derived from type 1 diabetic rats. IGF-1 protected against the loss of ATP supplies in DRG cell bodies and axons in neurons derived from diabetic rats by augmenting various parameters of glycolysis and mitochondrial respiration.
Collapse
Affiliation(s)
- Mohamad-Reza Aghanoori
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada; Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| | - Vicky Margulets
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Darrell R Smith
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Lorrie A Kirshenbaum
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada; Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada; Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada
| | - Daniel Gitler
- Department of Physiology and Cell Biology, Faculty of Health Sciences, and Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Paul Fernyhough
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada; Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
42
|
Mondal S, Dubey J, Awasthi A, Sure GR, Vasudevan A, Koushika SP. Tracking Mitochondrial Density and Positioning along a Growing Neuronal Process in Individual C. elegans Neuron Using a Long-Term Growth and Imaging Microfluidic Device. eNeuro 2021; 8:ENEURO.0360-20.2021. [PMID: 34035072 PMCID: PMC8260276 DOI: 10.1523/eneuro.0360-20.2021] [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: 08/16/2020] [Revised: 04/18/2021] [Accepted: 05/06/2021] [Indexed: 12/16/2022] Open
Abstract
The long cellular architecture of neurons requires regulation in part through transport and anchoring events to distribute intracellular organelles. During development, cellular and subcellular events such as organelle additions and their recruitment at specific sites on the growing axons occur over different time scales and often show interanimal variability thus making it difficult to identify specific phenomena in population averages. To measure the variability in subcellular events such as organelle positions, we developed a microfluidic device to feed and immobilize Caenorhabditis elegans for high-resolution imaging over several days. The microfluidic device enabled long-term imaging of individual animals and allowed us to investigate organelle density using mitochondria as a testbed in a growing neuronal process in vivo Subcellular imaging of an individual neuron in multiple animals, over 36 h in our microfluidic device, shows the addition of new mitochondria along the neuronal process and an increase in the accumulation of synaptic vesicles (SVs) at synapses. Long-term imaging of individual C. elegans touch receptor neurons (TRNs) shows that the addition of new mitochondria takes place along the entire neuronal process length at a rate of ∼0.6 mitochondria/h. The threshold for the addition of a new mitochondrion occurs when the average separation between the two preexisting mitochondria exceeds 24 μm. Our assay provides a new opportunity to move beyond simple observations obtained from in vitro assays to allow the discovery of genes that regulate positioning of mitochondria in neurons.
Collapse
Affiliation(s)
- Sudip Mondal
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka 560065, India
- Department of Mechanical Engineering, The University of Texas at Austin, Austin, Texas 78712
| | - Jyoti Dubey
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka 560065, India
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore, Karnataka 560065, India
- Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Anjali Awasthi
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka 560065, India
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani, Rajasthan 333031, India
| | - Guruprasad Reddy Sure
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka 560065, India
- Sastra University, Thirumalaisamudram, Tamil Nadu 613401, India
| | - Amruta Vasudevan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| | - Sandhya P Koushika
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| |
Collapse
|
43
|
Agrawal A, Koslover EF. Optimizing mitochondrial maintenance in extended neuronal projections. PLoS Comput Biol 2021; 17:e1009073. [PMID: 34106921 PMCID: PMC8216566 DOI: 10.1371/journal.pcbi.1009073] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/21/2021] [Accepted: 05/12/2021] [Indexed: 11/18/2022] Open
Abstract
Neurons rely on localized mitochondria to fulfill spatially heterogeneous metabolic demands. Mitochondrial aging occurs on timescales shorter than the neuronal lifespan, necessitating transport of fresh material from the soma. Maintaining an optimal distribution of healthy mitochondria requires an interplay between a stationary pool localized to sites of high metabolic demand and a motile pool capable of delivering new material. Interchange between these pools can occur via transient fusion / fission events or by halting and restarting entire mitochondria. Our quantitative model of neuronal mitostasis identifies key parameters that govern steady-state mitochondrial health at discrete locations. Very infrequent exchange between stationary and motile pools optimizes this system. Exchange via transient fusion allows for robust maintenance, which can be further improved by selective recycling through mitophagy. These results provide a framework for quantifying how perturbations in organelle transport and interactions affect mitochondrial homeostasis in neurons, a key aspect underlying many neurodegenerative disorders.
Collapse
Affiliation(s)
- Anamika Agrawal
- Department of Physics, University of California San Diego, La Jolla, California, United States of America
| | - Elena F. Koslover
- Department of Physics, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
44
|
Datta S, Jaiswal M. Mitochondrial calcium at the synapse. Mitochondrion 2021; 59:135-153. [PMID: 33895346 DOI: 10.1016/j.mito.2021.04.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 03/28/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022]
Abstract
Mitochondria are dynamic organelles, which serve various purposes, including but not limited to the production of ATP and various metabolites, buffering ions, acting as a signaling hub, etc. In recent years, mitochondria are being seen as the central regulators of cellular growth, development, and death. Since neurons are highly specialized cells with a heavy metabolic demand, it is not surprising that neurons are one of the most mitochondria-rich cells in an animal. At synapses, mitochondrial function and dynamics is tightly regulated by synaptic calcium. Calcium influx during synaptic activity causes increased mitochondrial calcium influx leading to an increased ATP production as well as buffering of synaptic calcium. While increased ATP production is required during synaptic transmission, calcium buffering by mitochondria is crucial to prevent faulty neurotransmission and excitotoxicity. Interestingly, mitochondrial calcium also regulates the mobility of mitochondria within synapses causing mitochondria to halt at the synapse during synaptic transmission. In this review, we summarize the various roles of mitochondrial calcium at the synapse.
Collapse
Affiliation(s)
- Sayantan Datta
- Tata Institute of Fundamental Research, Hyderabad, India
| | - Manish Jaiswal
- Tata Institute of Fundamental Research, Hyderabad, India.
| |
Collapse
|
45
|
Abstract
Mitochondria are multifunctional organelles that not only produce energy for the cell, but are also important for cell signalling, apoptosis and many biosynthetic pathways. In most cell types, they form highly dynamic networks that are constantly remodelled through fission and fusion events, repositioned by motor-dependent transport and degraded when they become dysfunctional. Motor proteins and their tracks are key regulators of mitochondrial homeostasis, and in this Review, we discuss the diverse functions of the three classes of motor proteins associated with mitochondria - the actin-based myosins, as well as the microtubule-based kinesins and dynein. In addition, Miro and TRAK proteins act as adaptors that link kinesin-1 and dynein, as well as myosin of class XIX (MYO19), to mitochondria and coordinate microtubule- and actin-based motor activities. Here, we highlight the roles of motor proteins and motor-linked track dynamics in the transporting and docking of mitochondria, and emphasize their adaptations in specialized cells. Finally, we discuss how motor-cargo complexes mediate changes in mitochondrial morphology through fission and fusion, and how they modulate the turnover of damaged organelles via quality control pathways, such as mitophagy. Understanding the importance of motor proteins for mitochondrial homeostasis will help to elucidate the molecular basis of a number of human diseases.
Collapse
Affiliation(s)
- Antonina J Kruppa
- Cambridge Institute for Medical Research, Department of Clinical Biochemistry, University of Cambridge, Cambridge Biomedical Campus, The Keith Peters Building, Hills Road, Cambridge CB2 0XY, UK
| | - Folma Buss
- Cambridge Institute for Medical Research, Department of Clinical Biochemistry, University of Cambridge, Cambridge Biomedical Campus, The Keith Peters Building, Hills Road, Cambridge CB2 0XY, UK
| |
Collapse
|
46
|
Khan H, Beck C, Kunze A. Multi-curvature micropatterns unveil distinct calcium and mitochondrial dynamics in neuronal networks. LAB ON A CHIP 2021; 21:1164-1174. [PMID: 33543185 PMCID: PMC7990709 DOI: 10.1039/d0lc01205j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Tangential curvatures are a key geometric feature of tissue folds in the human cerebral cortex. In the brain, these smoother and firmer bends are called gyri and sulci and form distinctive curved tissue patterns imposing a mechanical stimulus on neuronal networks. This stimulus is hypothesized to be essential for proper brain cell function but lacks in most standard neuronal cell assays. A variety of soft lithographic micropatterning techniques can be used to integrate round geometries in cell assays. Most microfabricated patterns, however, focus only on a small set of defined curvatures. In contrast, curvatures in the brain span a wide physical range, leaving it unknown which precise role distinct curvatures may play on neuronal cell signaling. Here we report a hydrogel-based multi-curvature design consisting of over twenty bands of distinct parallel curvature ranges to precisely engineer neuronal networks' growth and signaling under patterns of arcs. Monitoring calcium and mitochondrial dynamics in primary rodent neurons grown over two weeks in the multi-curvature patterns, we found that static calcium signaling was locally attenuated under higher curvatures (k > 0.01 μm-1). In contrast, to randomize growth, transient calcium signaling showed higher synchronicity when neurons formed networks in confined multi-curvature patterns. Additionally, we found that mitochondria showed lower motility under high curvatures (k > 0.01 μm-1) than under lower curvatures (k < 0.01 μm-1). Our results demonstrate how sensitive neuronal cell function may be linked and controlled through specific curved geometric features. Furthermore, the hydrogel-based multi-curvature design possesses high compatibility with various surfaces, allowing a flexible integration of geometric features into next-generation neuro devices, cell assays, tissue engineering, and implants.
Collapse
Affiliation(s)
- Hammad Khan
- Department of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, USA.
| | | | | |
Collapse
|
47
|
Bury AG, Vincent AE, Turnbull DM, Actis P, Hudson G. Mitochondrial isolation: when size matters. Wellcome Open Res 2021; 5:226. [PMID: 33718619 PMCID: PMC7931255 DOI: 10.12688/wellcomeopenres.16300.2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2020] [Indexed: 12/24/2022] Open
Abstract
Mitochondrial vitality is critical to cellular function, with mitochondrial dysfunction linked to a growing number of human diseases. Tissue and cellular heterogeneity, in terms of genetics, dynamics and function means that increasingly mitochondrial research is conducted at the single cell level. Whilst there are several technologies that are currently available for single-cell analysis, each with their advantages, they cannot be easily adapted to study mitochondria with subcellular resolution. Here we review the current techniques and strategies for mitochondrial isolation, critically discussing each technology's limitations for future mitochondrial research. Finally, we highlight and discuss the recent breakthroughs in sub-cellular isolation techniques, with a particular focus on nanotechnologies that enable the isolation of mitochondria from subcellular compartments. This allows isolation of mitochondria with unprecedented spatial precision with minimal disruption to mitochondria and their immediate cellular environment.
Collapse
Affiliation(s)
- Alexander G Bury
- Wellcome Trust Centre for Mitochondrial Research, Medical School, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK.,Biosciences Institute, Medical School, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK.,Pollard Institute, School of Electronic and Electrical Engineering, University of Leeds, Leeds, LS2 9JT, UK
| | - Amy E Vincent
- Wellcome Trust Centre for Mitochondrial Research, Medical School, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK.,Translational and Clinical Research Institute, Medical School, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK
| | - Doug M Turnbull
- Wellcome Trust Centre for Mitochondrial Research, Medical School, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK.,Translational and Clinical Research Institute, Medical School, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK
| | - Paolo Actis
- Pollard Institute, School of Electronic and Electrical Engineering, University of Leeds, Leeds, LS2 9JT, UK
| | - Gavin Hudson
- Wellcome Trust Centre for Mitochondrial Research, Medical School, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK.,Biosciences Institute, Medical School, Newcastle University, Newcastle-upon-Tyne, NE1 7RU, UK
| |
Collapse
|
48
|
Di Benedetto G, Iannucci LF, Surdo NC, Zanin S, Conca F, Grisan F, Gerbino A, Lefkimmiatis K. Compartmentalized Signaling in Aging and Neurodegeneration. Cells 2021; 10:464. [PMID: 33671541 PMCID: PMC7926881 DOI: 10.3390/cells10020464] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
The cyclic AMP (cAMP) signalling cascade is necessary for cell homeostasis and plays important roles in many processes. This is particularly relevant during ageing and age-related diseases, where drastic changes, generally decreases, in cAMP levels have been associated with the progressive decline in overall cell function and, eventually, the loss of cellular integrity. The functional relevance of reduced cAMP is clearly supported by the finding that increases in cAMP levels can reverse some of the effects of ageing. Nevertheless, despite these observations, the molecular mechanisms underlying the dysregulation of cAMP signalling in ageing are not well understood. Compartmentalization is widely accepted as the modality through which cAMP achieves its functional specificity; therefore, it is important to understand whether and how this mechanism is affected during ageing and to define which is its contribution to this process. Several animal models demonstrate the importance of specific cAMP signalling components in ageing, however, how age-related changes in each of these elements affect the compartmentalization of the cAMP pathway is largely unknown. In this review, we explore the connection of single components of the cAMP signalling cascade to ageing and age-related diseases whilst elaborating the literature in the context of cAMP signalling compartmentalization.
Collapse
Affiliation(s)
- Giulietta Di Benedetto
- Neuroscience Institute, National Research Council of Italy (CNR), 35121 Padova, Italy;
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
| | - Liliana F. Iannucci
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Nicoletta C. Surdo
- Neuroscience Institute, National Research Council of Italy (CNR), 35121 Padova, Italy;
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
| | - Sofia Zanin
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Filippo Conca
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Biology, University of Padova, 35122 Padova, Italy
| | - Francesca Grisan
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Biology, University of Padova, 35122 Padova, Italy
| | - Andrea Gerbino
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70121 Bari, Italy;
| | - Konstantinos Lefkimmiatis
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
49
|
Abstract
Mitochondria are signaling hubs responsible for the generation of energy through oxidative phosphorylation, the production of key metabolites that serve the bioenergetic and biosynthetic needs of the cell, calcium (Ca2+) buffering and the initiation/execution of apoptosis. The ability of mitochondria to coordinate this myriad of functions is achieved through the exquisite regulation of fundamental dynamic properties, including remodeling of the mitochondrial network via fission and fusion, motility and mitophagy. In this Review, we summarize the current understanding of the mechanisms by which these dynamic properties of the mitochondria support mitochondrial function, review their impact on human cortical development and highlight areas in need of further research.
Collapse
Affiliation(s)
- Tierney Baum
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
50
|
Misrani A, Tabassum S, Yang L. Mitochondrial Dysfunction and Oxidative Stress in Alzheimer's Disease. Front Aging Neurosci 2021; 13:617588. [PMID: 33679375 PMCID: PMC7930231 DOI: 10.3389/fnagi.2021.617588] [Citation(s) in RCA: 322] [Impact Index Per Article: 80.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
Mitochondria play a pivotal role in bioenergetics and respiratory functions, which are essential for the numerous biochemical processes underpinning cell viability. Mitochondrial morphology changes rapidly in response to external insults and changes in metabolic status via fission and fusion processes (so-called mitochondrial dynamics) that maintain mitochondrial quality and homeostasis. Damaged mitochondria are removed by a process known as mitophagy, which involves their degradation by a specific autophagosomal pathway. Over the last few years, remarkable efforts have been made to investigate the impact on the pathogenesis of Alzheimer’s disease (AD) of various forms of mitochondrial dysfunction, such as excessive reactive oxygen species (ROS) production, mitochondrial Ca2+ dyshomeostasis, loss of ATP, and defects in mitochondrial dynamics and transport, and mitophagy. Recent research suggests that restoration of mitochondrial function by physical exercise, an antioxidant diet, or therapeutic approaches can delay the onset and slow the progression of AD. In this review, we focus on recent progress that highlights the crucial role of alterations in mitochondrial function and oxidative stress in the pathogenesis of AD, emphasizing a framework of existing and potential therapeutic approaches.
Collapse
Affiliation(s)
- Afzal Misrani
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Sidra Tabassum
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Li Yang
- School of Life Sciences, Guangzhou University, Guangzhou, China
| |
Collapse
|