1
|
Feng S, Gui J, Qin B, Ye J, Zhao Q, Guo A, Sang M, Sun X. Resveratrol Inhibits VDAC1-Mediated Mitochondrial Dysfunction to Mitigate Pathological Progression in Parkinson's Disease Model. Mol Neurobiol 2025; 62:6636-6654. [PMID: 38819635 DOI: 10.1007/s12035-024-04234-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/13/2024] [Indexed: 06/01/2024]
Abstract
An increase in α-synuclein (α-syn) levels and mutations in proteins associated with mitochondria contribute to the development of familial Parkinson's disease (PD); however, the involvement of α-syn and mitochondria in idiopathic PD remains incompletely understood. The voltage-dependent anion channel I (VDAC1) protein, which serves as a crucial regulator of mitochondrial function and a gatekeeper, plays a pivotal role in governing cellular destiny through the control of ion and respiratory metabolite flux. The ability of resveratrol (RES), which is a potent phytoalexin with antioxidant and anti-inflammatory properties, to regulate VDAC1 in PD is unknown. The objective of this study was to evaluate the role of VDAC1 in the pathological process of PD and to explore the mechanism by which resveratrol protects dopaminergic neurons by regulating VDAC1 to maintain the mitochondrial permeability transition pore (mPTP) and calcium ion balance. The effects of RES on the motor and cognitive abilities of A53T mice were evaluated by using small animal behavioral tests. Various techniques, including immunofluorescence staining, transmission electron microscopy, enzyme-linked immunoadsorption, quantitative polymerase chain reaction (PCR), and Western blotting, among others, were employed to assess the therapeutic impact of RES on neuropathy associated with PD and its potential in regulating mitochondrial VDAC1. The findings showed that RES significantly improved motor and cognitive dysfunction and restored mitochondrial function, thus reducing oxidative stress levels in A53T mice. A significant positive correlation was observed between the protein expression level of VDAC1 and mitochondrial α-syn expression, as well as disease progression, whereas no such correlation was found in VDAC2 and VDAC3. Administration of RES resulted in a significant decrease in the protein expression of VDAC1 and in the protein expression of α-syn both in vivo and in vitro. In addition, we found that RES prevents excessive opening of the mPTP in dopaminergic neurons. This may prevent the abnormal aggregation of α-syn in mitochondria and the release of mitochondrial apoptosis signals. Furthermore, the activation of VDAC1 reversed the resveratrol-induced decrease in the accumulation of α-syn in the mitochondria. These findings highlight the potential of VDAC1 as a therapeutic target for PD and identify the mechanism by which resveratrol alleviates PD-related pathology by modulating mitochondrial VDAC1.
Collapse
Affiliation(s)
- Shenglan Feng
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Jianjun Gui
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Bingqing Qin
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Junjie Ye
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
- Department of Clinical Laboratory, Wuhan Asia Heart Hospital, Wuhan, 430022, Hubei, China
| | - Qiang Zhao
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Ai Guo
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Ming Sang
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China.
| | - Xiaodong Sun
- Research Center for Translational Medicine, Hubei Provincial Clinical Research Center for Parkinsons Disease at Xiangyang No.1 Peoples Hospital, Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan, 442000, China.
| |
Collapse
|
2
|
Myrup Holst C, Esperon-Abril I, Bryske Juhl F, Jakobsgaard JE, Kristiansen JB, Vissing K, Stevnsner T. Effect of prolonged voluntary wheel running on oxidative stress and defence mechanisms in cortex and hippocampus of healthy female rats. Exp Physiol 2025. [PMID: 40448974 DOI: 10.1113/ep092815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Accepted: 05/16/2025] [Indexed: 06/02/2025]
Abstract
Physical exercise promotes brain health and cognitive function possibly through mechanisms that include strengthened resistance to oxidative stress. However, limited research has explored the cumulative effects of regular voluntary exercise on both oxidative stress and defence mechanisms in hippocampus and cortex, two regions essential for cognitive function. Especially, adaptations in the young, healthy brain are insufficiently understood. This study investigates the impact of 8 weeks of voluntary wheel running on oxidative damage and counteracting defence mechanisms in the cortex and hippocampus of young, healthy female rats. To this end, we assessed oxidative damage to proteins and DNA, antioxidant defence, and DNA repair mechanisms, focusing on the base excision repair pathway. Our findings show that 8 weeks of voluntary exercise does not significantly modify oxidative damage or antioxidant defences in either cortical or hippocampal brain regions. Instead, the voluntary wheel running intervention led to a reduction in the levels of DNA polymerase β and mitochondrial apurinic/apyrimidinic endonuclease 1, key enzymes involved in base excision repair. Moreover, mitochondrial DNA copy number increased in the cortex, but decreased in the hippocampus, suggesting distinct regional adaptations. Collectively, these results indicate that the healthy young brain maintains redox homeostasis despite reduced DNA repair capacity. By analysing a comprehensive array of biomarkers in two brain regions, this study addresses gaps in our current knowledge on prolonged training and brain health and provides valuable insights into how regular exercise produces region-specific and shared responses in the healthy state.
Collapse
Affiliation(s)
- Camilla Myrup Holst
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Iria Esperon-Abril
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Frederik Bryske Juhl
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Jonas B Kristiansen
- Exercise Biology, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Kristian Vissing
- Exercise Biology, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Tinna Stevnsner
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| |
Collapse
|
3
|
Li D, Xie M, Zeng H, Yu J, Xu R, Wang Z, Huang Y, Yang Y, Sun Y. UPR mt alleviates bone cancer pain through the restoration of mitochondrial function. Exp Cell Res 2025; 448:114568. [PMID: 40273969 DOI: 10.1016/j.yexcr.2025.114568] [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: 01/31/2025] [Revised: 04/18/2025] [Accepted: 04/20/2025] [Indexed: 04/26/2025]
Abstract
The mitochondrial unfolded protein response (UPRmt) is an intracellular retrograde signaling process that facilitates the restoration of mitochondrial homeostasis. Mitochondria are essential for neuronal signaling, and their dysfunction has been implicated as a significant mechanism in the development of chronic pain. Nevertheless, little is known about the exact function of UPRmt in bone cancer pain (BCP). This research intended to explore the connection between UPRmt and the progression of BCP. In BCP group, the ultrastructure of spinal cord mitochondria was disrupted, accompanied by a decline in ATP levels and a decrease in Mitochondrial membrane potential (MMP). Concurrently, mRNA and protein levels of UPRmt marker proteins (Atf5, Hsp60, LonP1, and ClpP) were upregulated, with the expression of Atf5, a key transcription factor of UPRmt, notably enhanced in spinal dorsal horn neurons. Nicotinamide riboside (NR)-mediated pharmacological augmentation of the UPRmt significantly alleviated BCP-induced nociceptive hypersensitivity, as demonstrated by elevated mechanical withdrawal thresholds and diminished spontaneous flinching behavior. Concomitant mitochondrial functional recovery was evidenced by restoration of MMP and normalization of ATP level. Notably, genetic knockdown of activating transcription factor 5 (Atf5) abolished both NR-induced UPRmt activation and the consequent protection against rotenone-mediated mitochondrial dysfunction. These findings establish UPRmt potentiation as an effective strategy for ameliorating mitochondrial dysfunction and attenuating BCP-associated nociception, proposing this pathway as a novel therapeutic target for clinical pain management.
Collapse
Affiliation(s)
- Dan Li
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Mingming Xie
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Haohao Zeng
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiacheng Yu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Rui Xu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhen Wang
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yulin Huang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Yan Yang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Yu'e Sun
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
4
|
Mosharov EV, Rosenberg AM, Monzel AS, Osto CA, Stiles L, Rosoklija GB, Dwork AJ, Bindra S, Junker A, Zhang Y, Fujita M, Mariani MB, Bakalian M, Sulzer D, De Jager PL, Menon V, Shirihai OS, Mann JJ, Underwood MD, Boldrini M, Thiebaut de Schotten M, Picard M. A human brain map of mitochondrial respiratory capacity and diversity. Nature 2025; 641:749-758. [PMID: 40140564 DOI: 10.1038/s41586-025-08740-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 02/04/2025] [Indexed: 03/28/2025]
Abstract
Mitochondrial oxidative phosphorylation (OXPHOS) powers brain activity1,2, and mitochondrial defects are linked to neurodegenerative and neuropsychiatric disorders3,4. To understand the basis of brain activity and behaviour, there is a need to define the molecular energetic landscape of the brain5-10. Here, to bridge the scale gap between cognitive neuroscience and cell biology, we developed a physical voxelization approach to partition a frozen human coronal hemisphere section into 703 voxels comparable to neuroimaging resolution (3 × 3 × 3 mm). In each cortical and subcortical brain voxel, we profiled mitochondrial phenotypes, including OXPHOS enzyme activities, mitochondrial DNA and volume density, and mitochondria-specific respiratory capacity. We show that the human brain contains diverse mitochondrial phenotypes driven by both topology and cell types. Compared with white matter, grey matter contains >50% more mitochondria. Moreover, the mitochondria in grey matter are biochemically optimized for energy transformation, particularly among recently evolved cortical brain regions. Scaling these data to the whole brain, we created a backwards linear regression model that integrates several neuroimaging modalities11 to generate a brain-wide map of mitochondrial distribution and specialization. This model predicted mitochondrial characteristics in an independent brain region of the same donor brain. This approach and the resulting MitoBrainMap of mitochondrial phenotypes provide a foundation for exploring the molecular energetic landscape that enables normal brain function. This resource also relates to neuroimaging data and defines the subcellular basis for regionalized brain processes relevant to neuropsychiatric and neurodegenerative disorders. All data are available at http://humanmitobrainmap.bcblab.com .
Collapse
Affiliation(s)
- Eugene V Mosharov
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Ayelet M Rosenberg
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Anna S Monzel
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Corey A Osto
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Linsey Stiles
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Gorazd B Rosoklija
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Andrew J Dwork
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Snehal Bindra
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Alex Junker
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ya Zhang
- Center for Translational and Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Masashi Fujita
- Center for Translational and Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Madeline B Mariani
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Mihran Bakalian
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - David Sulzer
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Departments of Neurology and Pharmacology, Columbia University Irving Medical Center, New York, NY, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Vilas Menon
- Center for Translational and Computational Neuroimmunology, Neuroimmunology Division, Department of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Orian S Shirihai
- Department of Medicine, Endocrinology, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - J John Mann
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Mark D Underwood
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Maura Boldrini
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Division of Molecular Imaging and Neuropathology, Columbia University Irving Medical Center, New York, NY, USA
| | - Michel Thiebaut de Schotten
- Brain Connectivity and Behaviour Laboratory, Paris, France.
- Groupe d'Imagerie Neurofonctionnelle, Institut des Maladies Neurodégénératives-UMR 5293, CNRS, CEA University of Bordeaux, Bordeaux, France.
| | - Martin Picard
- Department of Psychiatry, Divisions of Molecular Therapeutics and Behavioral Medicine, Columbia University Irving Medical Center, New York, NY, USA.
- New York State Psychiatric Institute, New York, NY, USA.
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, Columbia University Irving Medical Center, New York, NY, USA.
- Robert N. Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA.
| |
Collapse
|
5
|
Cai J, Chen Y, She Y, He X, Feng H, Sun H, Yin M, Gao J, Sheng C, Li Q, Xiao M. Aerobic exercise improves astrocyte mitochondrial quality and transfer to neurons in a mouse model of Alzheimer's disease. Brain Pathol 2025; 35:e13316. [PMID: 39462160 PMCID: PMC11961210 DOI: 10.1111/bpa.13316] [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: 04/30/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024] Open
Abstract
Mitochondrial dysfunction is a well-established hallmark of Alzheimer's disease (AD). Despite recent documentation of transcellular mitochondrial transfer, its role in the pathogenesis of AD remains unclear. In this study, we report an impairment of mitochondrial quality within the astrocytes and neurons of adult 5 × FAD mice. Following treatment with mitochondria isolated from aged astrocytes induced by exposure to amyloid protein or extended cultivation, cultured neurons exhibited an excessive generation of reactive oxygen species and underwent neurite atrophy. Notably, aerobic exercise enhanced mitochondrial quality by upregulating CD38 within hippocampal astrocytes of 5 × FAD mice. Conversely, the knockdown of CD38 diminished astrocytic-neuronal mitochondrial transfer, thereby abolishing the ameliorative effects of aerobic exercise on neuronal oxidative stress, β-amyloid plaque deposition, and cognitive dysfunction in 5 × FAD mice. These findings unveil an unexpected mechanism through which aerobic exercise facilitates the transference of healthy mitochondria from astrocytes to neurons, thus countering the AD-like progression.
Collapse
Affiliation(s)
- Jiachen Cai
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Nanjing Brain Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Yan Chen
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Nanjing Brain Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Yuzhu She
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Nanjing Brain Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Xiaoxin He
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Nanjing Brain Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Hu Feng
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
| | - Huaiqing Sun
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Department of NeurologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Mengmei Yin
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Department of NeurologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Junying Gao
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Department of AnatomyNanjing Medical UniversityNanjingChina
| | - Chengyu Sheng
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
| | - Qian Li
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Nanjing Brain Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| | - Ming Xiao
- Jiangsu Key Laboratory of NeurodegenerationNanjing Medical UniversityNanjingChina
- Nanjing Brain Hospital Affiliated to Nanjing Medical UniversityNanjingChina
| |
Collapse
|
6
|
Lee-Glover LP, Picard M, Shutt TE. Mitochondria - the CEO of the cell. J Cell Sci 2025; 138:jcs263403. [PMID: 40310473 PMCID: PMC12070065 DOI: 10.1242/jcs.263403] [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: 05/02/2025] Open
Abstract
As we have learned more about mitochondria over the past decades, including about their essential cellular roles and how altered mitochondrial biology results in disease, it has become apparent that they are not just powerplants pumping out ATP at the whim of the cell. Rather, mitochondria are dynamic information and energy processors that play crucial roles in directing dozens of cellular processes and behaviors. They provide instructions to enact programs that regulate various cellular operations, such as complex metabolic networks, signaling and innate immunity, and even control cell fate, dictating when cells should divide, differentiate or die. To help current and future generations of cell biologists incorporate the dynamic, multifaceted nature of mitochondria and assimilate modern discoveries into their scientific framework, mitochondria need a 21st century 'rebranding'. In this Opinion article, we argue that mitochondria should be considered as the 'Chief Executive Organelle' - the CEO - of the cell.
Collapse
Affiliation(s)
- Laurie P. Lee-Glover
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral Medicine, Columbia University Irving Medical Center, New York, 10032, USA
- Department of Neurology, H. Houston Merritt Center for Neuromuscular and Mitochondrial Disorders, Columbia University Translational Neuroscience Initiative, Columbia University Irving Medical Center, New York, 10032, USA
- New York State Psychiatric Institute, New York, 10032, USA
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, 10032, USA
| | - Timothy E. Shutt
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Alberta, T2N 4N1, Canada
| |
Collapse
|
7
|
Xu X, Duan Z, Zhou X, Zhao R, Xu J, Zhang Z, Lv M, Wan Q, Cui Y. SFXN1 Reduction Alleviates Cerebral Ischemia-Reperfusion Injury by Promoting Neuronal Survival and Reducing Neuroinflammation. CNS Neurosci Ther 2025; 31:e70457. [PMID: 40420406 PMCID: PMC12106369 DOI: 10.1111/cns.70457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/16/2025] [Accepted: 05/06/2025] [Indexed: 05/28/2025] Open
Abstract
AIM Sideroflexin 1 (SFXN1) is an important inner mitochondrial membrane protein that regulates many physiological and pathological events. However, the role of SFXN1 in cerebral ischemia-reperfusion (I/R)-induced neuronal death remains unclear. METHODS We employed in vivo injury models of transient middle cerebral artery occlusion (tMCAO) and in vitro models of lipopolysaccharide (LPS) stimulation and oxygen-glucose deprivation/reperfusion (OGD/R) to investigate the regulatory effects of SFXN1 on neuroinflammation and brain injury. Western blotting, immunofluorescence, and real-time quantitative PCR were utilized to assess SFXN1 expression, proinflammatory signaling pathways activation, and cytokine levels in vitro. Cerebral infarction was evaluated using 2,3,5-triphenyltetrazolium chloride (TTC) staining and Nissl staining. RESULTS SFXN1 expression was upregulated following cerebral I/R injury. Both neurons and microglia exhibited increased SFXN1 expression after oxygen-glucose deprivation/reoxygenation (OGD/R) treatment. SFXN1 knockdown reduced OGD/R-induced neuronal death and alleviated cerebral I/R injury. Additionally, conditioned medium from SFXN1-knockdown microglia reduced neurotoxicity in vitro. Mechanistically, SFXN1 induced mitochondrial dysfunction and neuronal death after OGD/R in an iron-independent manner. Furthermore, SFXN1 promoted the production of proinflammatory cytokines by promoting NF-κB activation, partially through iron transport in microglia after OGD/R. CONCLUSION This study reveals the novel role of SFXN1 in exacerbating cerebral I/R injury by reducing neuronal survival through the modulation of mitochondrial function and promotion of microglia-mediated neuroinflammation via NF-κB activation.
Collapse
Affiliation(s)
- Xiangyu Xu
- Institute of Neuroregeneration and NeurorehabilitationQingdao Medical College, Qingdao UniversityQingdaoShandongChina
| | - Zhongying Duan
- Institute of Neuroregeneration and NeurorehabilitationQingdao Medical College, Qingdao UniversityQingdaoShandongChina
- School of Basic MedicineQingdao Medical College, Qingdao UniversityQingdaoShandongChina
| | - Xin Zhou
- Institute of Neuroregeneration and NeurorehabilitationQingdao Medical College, Qingdao UniversityQingdaoShandongChina
- School of Basic MedicineQingdao Medical College, Qingdao UniversityQingdaoShandongChina
| | - Rui Zhao
- Department of Interventional RadiologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Jing Xu
- Shandong First Medical University & Shandong Academy of Medical SciencesJinanShandongChina
| | - Zhaolong Zhang
- Department of Interventional RadiologyThe Affiliated Hospital of Qingdao UniversityQingdaoShandongChina
| | - Mengfei Lv
- Institute of Neuroregeneration and NeurorehabilitationQingdao Medical College, Qingdao UniversityQingdaoShandongChina
| | - Qi Wan
- Institute of Neuroregeneration and NeurorehabilitationQingdao Medical College, Qingdao UniversityQingdaoShandongChina
- Faculty of Life and HealthShenzhen University of Advanced ScienceShenzhenChina
| | - Yu Cui
- Institute of Neuroregeneration and NeurorehabilitationQingdao Medical College, Qingdao UniversityQingdaoShandongChina
- School of Basic MedicineQingdao Medical College, Qingdao UniversityQingdaoShandongChina
| |
Collapse
|
8
|
Begeman A, Smolka JA, Shami A, Waingankar TP, Lewis SC. Spatial analysis of mitochondrial gene expression reveals dynamic translation hubs and remodeling in stress. SCIENCE ADVANCES 2025; 11:eads6830. [PMID: 40249810 PMCID: PMC12007585 DOI: 10.1126/sciadv.ads6830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 03/14/2025] [Indexed: 04/20/2025]
Abstract
Protein- and RNA-rich bodies contribute to the spatial organization of gene expression in the cell and are also sites of quality control critical to cell fitness. In most eukaryotes, mitochondria harbor their own genome, and all steps of mitochondrial gene expression co-occur within a single compartment-the matrix. Here, we report that processed mitochondrial RNAs are consolidated into micrometer-scale translation hubs distal to mitochondrial DNA transcription and RNA processing sites in human cells. We find that, during stress, mitochondrial messenger and ribosomal RNA are sequestered in mesoscale bodies containing mitoribosome components, concurrent with suppression of active translation. Stress bodies are triggered by proteotoxic stress downstream of double-stranded RNA accumulation in cells lacking unwinding activity of the highly conserved helicase SUPV3L1/SUV3. We propose that the spatial organization of nascent polypeptide synthesis into discrete domains serves to throttle the flow of genetic information to support recovery of mitochondrial quality control.
Collapse
Affiliation(s)
- Adam Begeman
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - John A. Smolka
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Ahmad Shami
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | | | - Samantha C. Lewis
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Innovative Genomics Institute, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, Berkeley, CA, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA
| |
Collapse
|
9
|
Chen J, Chen J, Yu C, Xia K, Yang B, Wang R, Li Y, Shi K, Zhang Y, Xu H, Zhang X, Wang J, Chen Q, Liang C. Metabolic reprogramming: a new option for the treatment of spinal cord injury. Neural Regen Res 2025; 20:1042-1057. [PMID: 38989936 PMCID: PMC11438339 DOI: 10.4103/nrr.nrr-d-23-01604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 02/27/2024] [Indexed: 07/12/2024] Open
Abstract
Spinal cord injuries impose a notably economic burden on society, mainly because of the severe after-effects they cause. Despite the ongoing development of various therapies for spinal cord injuries, their effectiveness remains unsatisfactory. However, a deeper understanding of metabolism has opened up a new therapeutic opportunity in the form of metabolic reprogramming. In this review, we explore the metabolic changes that occur during spinal cord injuries, their consequences, and the therapeutic tools available for metabolic reprogramming. Normal spinal cord metabolism is characterized by independent cellular metabolism and intercellular metabolic coupling. However, spinal cord injury results in metabolic disorders that include disturbances in glucose metabolism, lipid metabolism, and mitochondrial dysfunction. These metabolic disturbances lead to corresponding pathological changes, including the failure of axonal regeneration, the accumulation of scarring, and the activation of microglia. To rescue spinal cord injury at the metabolic level, potential metabolic reprogramming approaches have emerged, including replenishing metabolic substrates, reconstituting metabolic couplings, and targeting mitochondrial therapies to alter cell fate. The available evidence suggests that metabolic reprogramming holds great promise as a next-generation approach for the treatment of spinal cord injury. To further advance the metabolic treatment of the spinal cord injury, future efforts should focus on a deeper understanding of neurometabolism, the development of more advanced metabolomics technologies, and the design of highly effective metabolic interventions.
Collapse
Affiliation(s)
- Jiangjie Chen
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Jinyang Chen
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Chao Yu
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Kaishun Xia
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Biao Yang
- Qiandongnan Prefecture People's Hospital, Kaili, Guizhou Province, China
| | - Ronghao Wang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Yi Li
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Kesi Shi
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Yuang Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Haibin Xu
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Xuesong Zhang
- Department of Orthopedics, Fourth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jingkai Wang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Qixin Chen
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| | - Chengzhen Liang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Orthopedics Research Institute of Zhejiang University, Zhejiang University, Hangzhou, Zhejiang Province, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang Province, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang Province, China
| |
Collapse
|
10
|
Fan H, Yuan M, Wang S, Yang X, Shu L, Pu Y, Zou Q, Zhang X, Wang C, Cai Z. Dietary salt promotes cognitive impairment through repression of SIRT3/PINK1-mediated mitophagy and fission. Mol Cell Biochem 2025; 480:2345-2360. [PMID: 38997506 DOI: 10.1007/s11010-024-05069-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: 03/14/2024] [Accepted: 07/04/2024] [Indexed: 07/14/2024]
Abstract
Dietary salt is increasingly recognized as an independent risk factor for cognitive impairment. However, the exact mechanisms are not yet fully understood. Mitochondria, which play a crucial role in energy metabolism, are implicated in cognitive function through processes such as mitochondrial dynamics and mitophagy. While mitochondrial dysfunction is acknowledged as a significant determinant of cognitive function, the specific relationship between salt-induced cognitive impairment and mitochondrial health has yet to be fully elucidated. Here, we explored the underlying mechanism of cognitive impairment of mice and N2a cells treated with high-salt focusing on the mitochondrial homeostasis with western blotting, immunofluorescence, electron microscopy, RNA sequencing, and more. We further explored the potential role of SIRT3 in salt-induced mitochondrial dysfunction and synaptic alteration through plasmid transfection and siRNA. High salt diet significantly inhibited mitochondrial fission and blocked mitophagy, leading to dysfunctional mitochondria and impaired synaptic plasticity. Our findings demonstrated that SIRT3 not only promote mitochondrial fission by modulating phosphorylated DRP1, but also rescue mitophagy through promoting PINK1/Parkin-dependent pathway. Overall, our data for the first time indicate that mitochondrial homeostasis imbalance is a driver of impaired synaptic plasticity in a cognitive impairment phenotype that is exacerbated by a long-term high-salt diet, and highlight the protective role of SIRT3 in this process.
Collapse
Affiliation(s)
- Haixia Fan
- Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
- First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Minghao Yuan
- Chongqing Medical University, Chongqing, 400016, China
| | - Shenyuan Wang
- Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
| | - Xu Yang
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
| | - Liu Shu
- Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
| | - Yinshuang Pu
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
| | - Qian Zou
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
| | - Xiaogang Zhang
- Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
| | - Chuanling Wang
- Chongqing Medical University, Chongqing, 400016, China
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China
| | - Zhiyou Cai
- Chongqing Medical University, Chongqing, 400016, China.
- Department of Neurology, Chongqing General Hospital, Chongqing, 400013, China.
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, China.
| |
Collapse
|
11
|
Xie M, Li D, Zeng H, Huang Y, Xu R, Wang Z, Yu J, Sun Y. BAM8-22 targets spinal MrgC receptors to modulate UPR mt activity in the mechanism of bone cancer pain. Front Pharmacol 2025; 16:1575733. [PMID: 40230701 PMCID: PMC11994654 DOI: 10.3389/fphar.2025.1575733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
Background Bone cancer pain (BCP) significantly impacts patients' overall quality of life. Cellular energy metabolism homeostasis is critically dependent on mitochondrial integrity, and emerging evidence suggests that mitochondrial dysfunction in chronic BCP exacerbates pain progression by disrupting nociceptive signaling pathways. Notably, G protein-coupled receptors (GPCRs), a major class of membrane receptors, modulate mitochondrial function through diverse molecular mechanisms. In this study, we investigated the role of Mas-related G protein-coupled receptor C (MrgC) in BCP pathogenesis and its regulatory effects on mitochondrial function. Methods Male C3H/HeN mice were utilized to establish a BCP model. Transmission electron microscopy and flow cytometry were employed to assess changes in mitochondrial ultrastructure, as well as levels of mtROS, ATP, and MMP in mice experiencing BCP. Following intrathecal injection of BAM8-22, we analyzed the effects of activated MrgC on mitochondrial unfolded protein response (UPRmt)-related molecules (ATF5, HSP60, LONP1, CLPP) and pain-related behaviors in BCP mice. The regulatory mechanism of MrgC on UPRmt was further explored in N2a and 293T cells. Results Mice with bone cancer pain showed improved mRNA and protein levels of UPRmt-related molecules, increased MMP and ATP, decreased mitochondrial ROS levels in the spinal cord after receiving an intrathecal injection of BAM8-22. Additionally, the paw withdrawal mechanical threshold in BCP mice increased, while the number of spontaneous foot lifts decreased. In complementary cellular studies, transfection-mediated overexpression of MrgC in N2a cells enhanced UPRmt biomarker expression, whereas RNA interference-mediated MrgC knockdown produced the opposite effect. Conclusion By activating spinal MrgC to mediate UPRmt activity and protect mitochondrial function, BAM8-22 contributes to the molecular development of BCP. This discovery suggests a new therapeutic target for BCP and offers a possible research avenue.
Collapse
Affiliation(s)
- Mingming Xie
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Dan Li
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Haohao Zeng
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yulin Huang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Rui Xu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhen Wang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Jiacheng Yu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yu’e Sun
- Department of Anesthesiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
12
|
Ray MW, Chen W, Duan C, Bravo G, Krueger K, Rosario EM, Jacob AA, Lackner LL. The Volume of Mitochondria Inherited Impacts mtDNA Homeostasis in Budding Yeast. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645216. [PMID: 40196573 PMCID: PMC11974859 DOI: 10.1101/2025.03.25.645216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Most eukaryotic cells maintain mitochondria in well-distributed, reticular networks. The size of the mitochondrial network and copy number of its genome scale with cell size. However, while the size scaling features of mitochondria and their genome are interrelated, the fitness consequences of this interdependence are not well understood. We exploit the asymmetric cell division of budding yeast to test the hypothesis that mitochondrial scaling with cell size impacts mitochondrial DNA (mtDNA) function. We find that the volume of mitochondria inherited by daughter cells affects the ability of cells to maintain functional mtDNA; daughter cells that inherit a significantly reduced volume of mitochondria have an increased frequency of losing respiratory competence. In cells with such mitochondrial inheritance defects, mtDNA integrity can be maintained by upregulating mtDNA copy number. Collectively, these data support a bet-hedging model whereby the faithful inheritance of an adequate volume of mitochondria ensures enough mtDNA copies are transmitted to daughter cells to counteract pre-existing and/or inevitable mtDNA mutations.
Collapse
Affiliation(s)
- Michael W. Ray
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - WeiTing Chen
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Chengzhe Duan
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Guadalupe Bravo
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Kyle Krueger
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Erica M. Rosario
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Alexis A. Jacob
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Laura L. Lackner
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| |
Collapse
|
13
|
Marlar-Pavey M, Tapias-Gomez D, Mettlen M, Friedman JR. Compositionally unique mitochondria in filopodia support cellular migration. Curr Biol 2025; 35:1227-1241.e6. [PMID: 39978347 PMCID: PMC11945552 DOI: 10.1016/j.cub.2025.01.062] [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: 06/28/2024] [Revised: 01/07/2025] [Accepted: 01/28/2025] [Indexed: 02/22/2025]
Abstract
Local metabolic demand within cells varies widely, and the extent to which individual mitochondria can be specialized to meet these functional needs is unclear. We examined the subcellular distribution of the mitochondrial contact site and cristae organizing system (MICOS) complex, a spatial and functional organizer of mitochondria, and discovered that it dynamically enriches at the tip of a minor population of mitochondria in the cell periphery. Based on their appearance, we term these mitochondria "METEORs". METEORs have a unique composition, and MICOS enrichment sites are depleted of mtDNA and matrix proteins and contain high levels of the Ca2+ uniporter MCU, suggesting a functional specialization. METEORs are also enriched for the myosin MYO19, which promotes their trafficking to a small subset of filopodia. We identify a positive correlation between the length of filopodia and the presence of METEORs and show that elimination of mitochondria from filopodia impairs cellular motility. Our data reveal a novel type of mitochondrial heterogeneity and suggest compositionally specialized mitochondria support cell migration.
Collapse
Affiliation(s)
- Madeleine Marlar-Pavey
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daniel Tapias-Gomez
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Marcel Mettlen
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jonathan R Friedman
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
14
|
Kageyama Y, Okura S, Sukigara A, Matsunaga A, Maekubo K, Oue T, Ishihara K, Deguchi Y, Inoue K. The Association Among Bipolar Disorder, Mitochondrial Dysfunction, and Reactive Oxygen Species. Biomolecules 2025; 15:383. [PMID: 40149919 PMCID: PMC11940798 DOI: 10.3390/biom15030383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Mitochondria, often known as the cell's powerhouses, are primarily responsible for generating energy through aerobic oxidative phosphorylation. However, their functions extend far beyond just energy production. Mitochondria play crucial roles in maintaining calcium balance, regulating apoptosis (programmed cell death), supporting cellular signaling, influencing cell metabolism, and synthesizing reactive oxygen species (ROS). Recent research has highlighted a strong link between bipolar disorder (BD) and mitochondrial dysfunction. Mitochondrial dysfunction contributes to oxidative stress, particularly through the generation of ROS, which are implicated in the pathophysiology of BD. Oxidative stress arises when there is an imbalance between the production of ROS and the cell's ability to neutralize them. In neurons, excessive ROS can damage various cellular components, including proteins in neuronal membranes and intracellular enzymes. Such damage may interfere with neurotransmitter reuptake and the function of critical enzymes, potentially affecting brain regions involved in mood regulation and emotional control, which are key aspects of BD. In this review, we will explore how various types of mitochondrial dysfunction contribute to the production of ROS. These include disruptions in energy metabolism, impaired ROS management, and defects in mitochondrial quality control mechanisms such as mitophagy (the process by which damaged mitochondria are selectively degraded). We will also examine how abnormalities in calcium signaling, which is crucial for synaptic plasticity, can lead to mitochondrial dysfunction. Additionally, we will discuss the specific mitochondrial dysfunctions observed in BD, highlighting how these defects may contribute to the disorder's pathophysiology. Finally, we will identify potential therapeutic targets to improve mitochondrial function, which could pave the way for new treatments to manage or mitigate symptoms of BD.
Collapse
|
15
|
Luo XD, Tang S, Luo XY, Quzhen L, Xia RH, Wang XW. Mitochondrial regulation of obesity by POMC neurons. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167682. [PMID: 39837429 DOI: 10.1016/j.bbadis.2025.167682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
Pro-opiomelanocortin (POMC) neurons, nestled in the hypothalamus, play a pivotal role in the intricate coordination of energy homeostasis and metabolic pathways. These neurons' mitochondria, often hailed as the cell's powerhouses, are crucial for maintaining cellular energy equilibrium and metabolic functionality. Recent research has illuminated the complex interplay between mitochondrial dynamics and POMC neuronal activity, underscoring their critical involvement in the pathogenesis of a spectrum of metabolic disorders, notably obesity and diabetes. This comprehensive review delves into the molecular mechanisms that underlie how mitochondrial function within POMC neurons modulates metabolic regulation. We dissect the impact of mitochondrial dynamics, encompassing fusion, fission, mitophagy, and biogenesis, on the regulation of POMC neuronal activity. Furthermore, we scrutinize the role of mitochondrial dysfunction in POMC neurons in the etiology of obesity, identifying key therapeutic targets within these pathways. We offer an in-depth perspective on the indispensable role of POMC neuronal mitochondria in metabolic regulation and chart future research directions to bridge the existing knowledge gaps in this field.
Collapse
Affiliation(s)
- Xing-Dan Luo
- Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, China
| | - Si Tang
- Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, China
| | - Xiang-Yun Luo
- Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, China
| | - Luosang Quzhen
- The Central Hospital of Qusong County, Shannan, Tibet Autonomous Region 856300, China
| | - Ruo-Han Xia
- Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, China.
| | - Xian-Wang Wang
- Department of Biochemistry and Molecular Biology, Center for Molecular Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China; Shannan Maternal and Child Health Hospital, Shannan, Xizang 856100, China.
| |
Collapse
|
16
|
Jiang Y, Wang H, Boergens KM, Rzepka N, Wang F, Hua Y. Efficient cell-wide mapping of mitochondria in electron microscopic volumes using webKnossos. CELL REPORTS METHODS 2025; 5:100989. [PMID: 39999790 PMCID: PMC11955265 DOI: 10.1016/j.crmeth.2025.100989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/07/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025]
Abstract
Recent technical advances in volume electron microscopy (vEM) and artificial-intelligence-assisted image processing have facilitated high-throughput quantifications of cellular structures, such as mitochondria, that are ubiquitous and morphologically diversified. A still often-overlooked computational challenge is to assign a cell identity to numerous mitochondrial instances, for which both mitochondrial and cell membrane contouring used to be required. Here, we present a vEM reconstruction procedure (called mito-SegEM) that utilizes virtual-path-based annotation to assign automatically segmented mitochondrial instances at the cellular scale, therefore bypassing the requirement of membrane contouring. The embedded toolset in webKnossos (an open-source online annotation platform) is optimized for fast annotation, visualization, and proofreading of cellular organelle networks. We demonstrate the broad applications of mito-SegEM on volumetric datasets from various tissues, including the brain, intestine, and testis, to achieve an accurate and efficient reconstruction of mitochondria in a use-dependent fashion.
Collapse
Affiliation(s)
- Yi Jiang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125 Shanghai, China.
| | - Haoyu Wang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125 Shanghai, China
| | - Kevin M Boergens
- Department of Physics, University of Illinois Chicago, Chicago, IL 60607, USA
| | | | - Fangfang Wang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125 Shanghai, China
| | - Yunfeng Hua
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 200125 Shanghai, China.
| |
Collapse
|
17
|
Picard M, Monzel A, Devine J, Kapri D, Enriquez J, Trumpff C. A Quantitative Approach to Mapping Mitochondrial Specialization and Plasticity. RESEARCH SQUARE 2025:rs.3.rs-5961609. [PMID: 39989954 PMCID: PMC11844627 DOI: 10.21203/rs.3.rs-5961609/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Mitochondria are a diverse family of organelles that specialize to accomplish complimentary functions 1-3. All mitochondria share general features, but not all mitochondria are created equal 4.Here we develop a quantitative pipeline to define the degree of molecular specialization among different mitochondrial phenotypes - or mitotypes. By distilling hundreds of validated mitochondrial genes/proteins into 149 biologically interpretable MitoPathway scores (MitoCarta 3.0 5) the simple mitotyping pipeline allows investigators to quantify and interpret mitochondrial diversity and plasticity from transcriptomics or proteomics data across a variety of natural and experimental contexts. We show that mouse and human multi-organ mitotypes segregate along two main axes of mitochondrial specialization, contrasting anabolic (liver) and catabolic (brain) tissues. In cultured primary human fibroblasts exhibiting robust time-dependent and treatment-induced metabolic plasticity 6-8, we demonstrate how the mitotype of a given cell type recalibrates i) over time in parallel with hallmarks of aging, and ii) in response to genetic, pharmacological, and metabolic perturbations. Investigators can now use MitotypeExplorer.org and the associated code to visualize, quantify and interpret the multivariate space of mitochondrial biology.
Collapse
|
18
|
Huang R, Huang X, Yang H, Wu H, Liu F, Saw PE, Cao M. ATF3 Knockdown Exacerbates Astrocyte Activation by Inhibiting Phosphorylation of Drp1 in Ischemic Stroke. Biologics 2025; 19:15-29. [PMID: 39963286 PMCID: PMC11831017 DOI: 10.2147/btt.s486597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 01/29/2025] [Indexed: 02/20/2025]
Abstract
Introduction ATF3, a stress-induced transcription factor, has been implicated in the injury processes of various cell types, including neurons. It is recognized as a common marker for neuronal damage following neurotrauma. However, its role in other types of glial cells, particularly astrocytes, in response to ischemic injury remains unclear. Mitochondrial dysfunction is a key factor in the pathogenesis of ischemic stroke, and impaired mitochondrial function in astrocytes is associated with astrocyte activation. This study aimed to explore the relationship between mitochondrial damage and ischemic stroke and to investigate how ATF3 regulates mitochondrial dysfunction and astrocyte activation in the context of ischemic injury. Methods In a transient middle cerebral artery occlusion (tMCAO) mouse model, we knocked down ATF3 and assessed infarct size, motor deficits, astrocyte activation, and mitochondrial damage. In vitro, we used oxygen-glucose deprivation and reoxygenation (OGD-R) to simulate ischemia and evaluated the impact of ATF3 knockdown on astrocyte activation and mitochondrial function. Results ATF3 knockdown exacerbated infarct size, motor deficits, and astrocyte activation in vivo, with increased mitochondrial damage. In vitro, ATF3 depletion worsened mitochondrial dysfunction and astrocyte activation. ATF3 interacted with Drp1 via Akt2, inhibiting mitochondrial fission and protecting astrocytes. Conclusion ATF3 regulates mitochondrial fission and protects astrocytes in ischemic stroke, highlighting its potential as a therapeutic target for stroke recovery.
Collapse
Affiliation(s)
- Rong Huang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
| | - Xiaoyan Huang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
| | - Hongmei Yang
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
| | - Haixuan Wu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
| | - Fan Liu
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
| | - Phei Er Saw
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
| | - Minghui Cao
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People’s Republic of China
- Department of Anesthesiology, Shenshan Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Shanwei, 516600, People’s Republic of China
| |
Collapse
|
19
|
Monzel AS, Devine J, Kapri D, Enriquez JA, Trumpff C, Picard M. A Quantitative Approach to Mapping Mitochondrial Specialization and Plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.635951. [PMID: 39975232 PMCID: PMC11838522 DOI: 10.1101/2025.02.03.635951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Mitochondria are a diverse family of organelles that specialize to accomplish complimentary functions1-3. All mitochondria share general features, but not all mitochondria are created equal4. Here we develop a quantitative pipeline to define the degree of molecular specialization among different mitochondrial phenotypes - or mitotypes. By distilling hundreds of validated mitochondrial genes/proteins into 149 biologically interpretable MitoPathway scores (MitoCarta 3.05) the simple mitotyping pipeline allows investigators to quantify and interpret mitochondrial diversity and plasticity from transcriptomics or proteomics data across a variety of natural and experimental contexts. We show that mouse and human multi-organ mitotypes segregate along two main axes of mitochondrial specialization, contrasting anabolic (liver) and catabolic (brain) tissues. In cultured primary human fibroblasts exhibiting robust time-dependent and treatment-induced metabolic plasticity6-8, we demonstrate how the mitotype of a given cell type recalibrates i) over time in parallel with hallmarks of aging, and ii) in response to genetic, pharmacological, and metabolic perturbations. Investigators can now use MitotypeExplorer.org and the associated code to visualize, quantify and interpret the multivariate space of mitochondrial biology.
Collapse
Affiliation(s)
- Anna S. Monzel
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Jack Devine
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Darshana Kapri
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Jose Antonio Enriquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Caroline Trumpff
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Martin Picard
- Division of Behavioral Medicine, Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- Department of Neurology, H. Houston Merritt Center, Neuromuscular Medicine Division, Columbia University Medical Center, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, NY, USA
| |
Collapse
|
20
|
Pannoni KE, Fischer QS, Tarannum R, Cawley ML, Alsalman MM, Acosta N, Ezigbo C, Gil DV, Campbell LA, Farris S. MCU expression in hippocampal CA2 neurons modulates dendritic mitochondrial morphology and synaptic plasticity. Sci Rep 2025; 15:4540. [PMID: 39915602 PMCID: PMC11802895 DOI: 10.1038/s41598-025-85958-4] [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: 11/01/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025] Open
Abstract
Neuronal mitochondria are diverse across cell types and subcellular compartments in order to meet unique energy demands. While mitochondria are essential for synaptic transmission and synaptic plasticity, the mechanisms regulating mitochondria to support normal synapse function are incompletely understood. The mitochondrial calcium uniporter (MCU) is proposed to couple neuronal activity to mitochondrial ATP production, which would allow neurons to rapidly adapt to changing energy demands. MCU is uniquely enriched in hippocampal CA2 distal dendrites compared to proximal dendrites, however, the functional significance of this layer-specific enrichment is not clear. Synapses onto CA2 distal dendrites readily express plasticity, unlike the plasticity-resistant synapses onto CA2 proximal dendrites, but the mechanisms underlying these different plasticity profiles are unknown. Using a CA2-specific MCU knockout (cKO) mouse, we found that MCU deletion impairs plasticity at distal dendrite synapses. However, mitochondria were more fragmented and spine head area was diminished throughout the dendritic layers of MCU cKO mice versus control mice. Fragmented mitochondria might have functional changes, such as altered ATP production, that could explain the structural and functional deficits at cKO synapses. Differences in MCU expression across cell types and circuits might be a general mechanism to tune mitochondrial function to meet distinct synaptic demands.
Collapse
Affiliation(s)
- Katy E Pannoni
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Quentin S Fischer
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Renesa Tarannum
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, USA
| | - Mikel L Cawley
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Tech, Blacksburg, VA, USA
| | - Mayd M Alsalman
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Nicole Acosta
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Chisom Ezigbo
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Daniela V Gil
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Logan A Campbell
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Shannon Farris
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA.
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA.
- Virginia Tech Carilion School of Medicine, Roanoke, VA, USA.
| |
Collapse
|
21
|
Riou A, Broeglin A, Grimm A. Mitochondrial transplantation in brain disorders: Achievements, methods, and challenges. Neurosci Biobehav Rev 2025; 169:105971. [PMID: 39638101 DOI: 10.1016/j.neubiorev.2024.105971] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/08/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Mitochondrial transplantation is a new treatment strategy aimed at repairing cellular damage by introducing healthy mitochondria into injured cells. The approach shows promise in protecting brain function in various neurological disorders such as traumatic brain injury/ischemia, neurodegenerative diseases, cognitive disorders, and cancer. These conditions are often characterized by mitochondrial dysfunction, leading to impaired energy production and neuronal death. The review highlights promising preclinical studies where mitochondrial transplantation has been shown to restore mitochondrial function, reduce inflammation, and improve cognitive and motor functions in several animal models. It also addresses significant challenges that must be overcome before this therapy can be clinically applied. Current efforts to overcome these challenges, including advancements in isolation techniques, cryopreservation methods, finding an appropriate mitochondria source, and potential delivery routes, are discussed. Considering the rising incidence of neurological disorders and the limited effectiveness of current treatments, this review offers a comprehensive overview of the current state of mitochondrial transplantation research and critically assesses the remaining obstacles. It provides valuable insights that could steer future studies and potentially lead to more effective treatments for various brain disorders.
Collapse
Affiliation(s)
- Aurélien Riou
- Research Cluster Molecular and Cognitive Neurosciences, Department of Biomedicine, University of Basel, Basel 4055, Switzerland
| | - Aline Broeglin
- Research Cluster Molecular and Cognitive Neurosciences, Department of Biomedicine, University of Basel, Basel 4055, Switzerland
| | - Amandine Grimm
- Research Cluster Molecular and Cognitive Neurosciences, Department of Biomedicine, University of Basel, Basel 4055, Switzerland; Neurobiology Lab for Brain Aging and Mental Health, University Psychiatric Clinics Basel, Basel 4002, Switzerland.
| |
Collapse
|
22
|
Wen P, Sun Z, Gou F, Wang J, Fan Q, Zhao D, Yang L. Oxidative stress and mitochondrial impairment: Key drivers in neurodegenerative disorders. Ageing Res Rev 2025; 104:102667. [PMID: 39848408 DOI: 10.1016/j.arr.2025.102667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/25/2025]
Abstract
Mitochondrial dysfunction and oxidative stress are critical factors in the pathogenesis of neurodegenerative diseases. The complex interplay between these factors exacerbates neuronal damage and accelerates disease progression. In neurodegenerative diseases, mitochondrial dysfunction impairs ATP production and promotes the generation of reactive oxygen species (ROS). The accumulation of ROS further damages mitochondrial DNA, proteins, and lipids, creating a vicious cycle of oxidative stress and mitochondrial impairment. This review aims to elucidate the mechanisms by which mitochondrial dysfunction and oxidative stress lead to neurodegeneration, and to highlight potential therapeutic targets to mitigate their harmful effects.
Collapse
Affiliation(s)
- Pei Wen
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhixin Sun
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Fengting Gou
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jingjing Wang
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Qing Fan
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Deming Zhao
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lifeng Yang
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China.
| |
Collapse
|
23
|
Gupta S, Kishore A, Rishi V, Aggarwal A. Mitochondria and its epigenetic dynamics: Insight into synaptic regulation and synaptopathies. Funct Integr Genomics 2025; 25:26. [PMID: 39849126 DOI: 10.1007/s10142-025-01530-3] [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: 10/09/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/25/2025]
Abstract
Mitochondria, the cellular powerhouses, are pivotal to neuronal function and health, particularly through their role in regulating synaptic structure and function. Spine reprogramming, which underlies synapse development, depends heavily on mitochondrial dynamics-such as biogenesis, fission, fusion, and mitophagy as well as functions including ATP production, calcium (Ca2+) regulation, and retrograde signaling. Mitochondria supply the energy necessary for assisting synapse development and plasticity, while also regulating intracellular Ca2+ homeostasis to prevent excitotoxicity and support synaptic neurotransmission. Additionally, the dynamic processes of mitochondria ensure mitochondrial quality and adaptability, which are essential for maintaining effective synaptic activity. Emerging evidence highlights the significant role of epigenetic modifications in regulating mitochondrial dynamics and function. Epigenetic changes influence gene expression, which in turn affects mitochondrial activity, ensuring coordinated responses necessary for synapse development. Furthermore, metabolic changes within mitochondria can impact the epigenetic machinery, thereby modulating gene expression patterns that support synaptic integrity. Altered epigenetic regulation affecting mitochondrial dynamics and functions is linked to several neurological disorders, including Amyotrophic Lateral Sclerosis, Huntington's, Alzheimer's, and Parkinson's diseases, emphasizing its crucial function. The review delves into the molecular machinery involved in mitochondrial dynamics, ATP and Ca2+ regulation, highlighting the role of key proteins that facilitate the processes. Additionally, it also shed light on the emerging epigenetic factors influencing these regulations. It provides a thorough summary on the current understanding of the role of mitochondria in synapse development and emphasizes the importance of both molecular and epigenetic mechanisms in maintaining synaptic integrity.
Collapse
Affiliation(s)
- Shiwangi Gupta
- National Agri-Food and Biomanufacturing Institute, Sector-81, SAS Nagar, Knowledge City, Punjab, India
- Department of Biotechnology, Sector-25, Panjab University, BMS block I, Chandigarh, India
| | - Abhinoy Kishore
- Indian Institute of Science, Bengaluru, India
- Chandigarh Group of Colleges, Landran, Punjab, India
| | - Vikas Rishi
- National Agri-Food and Biomanufacturing Institute, Sector-81, SAS Nagar, Knowledge City, Punjab, India
| | - Aanchal Aggarwal
- National Agri-Food and Biomanufacturing Institute, Sector-81, SAS Nagar, Knowledge City, Punjab, India.
| |
Collapse
|
24
|
Andoh M, Shinoda N, Taira Y, Araki T, Kasahara Y, Takeuchi H, Miura M, Ikegaya Y, Koyama R. Nonapoptotic caspase-3 guides C1q-dependent synaptic phagocytosis by microglia. Nat Commun 2025; 16:918. [PMID: 39843445 PMCID: PMC11754728 DOI: 10.1038/s41467-025-56342-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
Caspases are known to mediate neuronal apoptosis during brain development. However, here we show that nonapoptotic activation of caspase-3 at presynapses drives microglial synaptic phagocytosis. Real-time observation and spatiotemporal manipulation of synaptic caspase-3 in the newly established, mouse-derived culture system demonstrate that increased neuronal activity triggers localized presynaptic caspase-3 activation, facilitating synaptic tagging by complements. High-resolution live imaging reveals that caspase-3 activation promotes synapse-selective complement-dependent microglial phagocytosis without axonal shearing. Furthermore, activity-dependent caspase-3 activation at inhibitory presynapses induces microglial phagocytosis in mice and increases seizure susceptibility. This increased susceptibility is reversed by genetic depletion of microglial complement receptors. Thus, localized, nonapoptotic caspase activity guides complement-dependent microglial synaptic phagocytosis and remodels neuronal circuits.
Collapse
Affiliation(s)
- Megumi Andoh
- Department of Translational Neurobiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-8502, Japan
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Natsuki Shinoda
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yusuke Taira
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Tasuku Araki
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yuka Kasahara
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Haruki Takeuchi
- Laboratory of Molecular Neurobiology, Department of Biophysics and Biochemistry, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0032, Japan
| | - Masayuki Miura
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
- Institute for AI and Beyond, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0032, Japan
- Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita City, Osaka, 565-0871, Japan
| | - Ryuta Koyama
- Department of Translational Neurobiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, 187-8502, Japan.
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan.
- Institute for AI and Beyond, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0032, Japan.
| |
Collapse
|
25
|
Cheng W, Yin C, Yu S, Chen X, Hong N, Jin W. scMMO-atlas: a single cell multimodal omics atlas and portal for exploring fine cell heterogeneity and cell dynamics. Nucleic Acids Res 2025; 53:D1186-D1194. [PMID: 39315707 PMCID: PMC11701702 DOI: 10.1093/nar/gkae821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/27/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024] Open
Abstract
Single-cell multimodal sequencing parallelly captures multiple modalities of the same cell, providing unparalleled insights into cell heterogeneity and cell dynamics. For example, joint profiling of chromatin accessibility and transcriptome from the same single cell (scATAC + RNA) identified new cell subsets within the well-defined clusters. However, lack of single-cell multimodal omics (scMMO) database has led to data fragmentation, seriously hindering access, utilization and mining of scMMO data. Here, we constructed a scMMO atlas by collecting and integrating various scMMO data, then constructed scMMO database and portal called scMMO-atlas (https://www.biosino.org/scMMO-atlas/). scMMO-atlas includes scATAC + RNA (ISSAAS-seq, SNARE-seq, paired-seq, sci-CAR, scCARE-seq, 10X Multiome and so on), scRNA + protein, scATAC + protein and scTri-modal omics data, with 3 168 824 cells from 27 cell tissues/organs. scMMO-atlas offered an interactive portal for visualization and featured analysis for each modality and the integrated data. Integrated analysis of scATAC + RNA data of mouse cerebral cortex in scMMO-atlas identified more cell subsets compared with unimodal omics data. Among these new cell subsets, there is an early astrocyte subset highly expressed Grm3, called Astro-Grm3. Furthermore, we identified Ex-L6-Tle4-Nrf1, a progenitor of Ex-L6-Tle4, indicating the statistical power provided by the big data in scMMO-atlas. In summary, scMMO-atlas offers cell atlas, database and portal to facilitate data utilization and biological insight.
Collapse
Affiliation(s)
- Wenwen Cheng
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Changhui Yin
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shiya Yu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xi Chen
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
| | - Ni Hong
- School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055 Guangdong, China
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenfei Jin
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
26
|
Stawikowska A, Dziembowska M, Kuzniewska B. Metabolic Phenotyping of Synaptic Mitochondria Using MitoPlates™ and Synaptoneurosomes. Methods Mol Biol 2025; 2878:67-74. [PMID: 39546257 DOI: 10.1007/978-1-0716-4264-1_4] [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] [Indexed: 11/17/2024]
Abstract
Mitochondrial functional assays using MitoPlates™ S-1 allow us to characterize mitochondria in terms of substrate metabolism. MitoPlates™ are 96-well microplates pre-coated with a diverse set of substrates. The electron flow from NADH and FADH2 producing mitochondrial substrates is measured based on the reduction of redox dye, that acts as a terminal electron acceptor. Here, we describe the application of MitoPlates™ to characterize the metabolism of synaptic mitochondria enclosed in isolated pre- and postsynaptic terminals (synaptoneurosomes).
Collapse
Affiliation(s)
- Aleksandra Stawikowska
- Department of Animal Physiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Magdalena Dziembowska
- Department of Animal Physiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Bozena Kuzniewska
- Department of Animal Physiology, Faculty of Biology, University of Warsaw, Warsaw, Poland.
| |
Collapse
|
27
|
Rizzollo F, Agostinis P. Mitochondria-Lysosome Contact Sites: Emerging Players in Cellular Homeostasis and Disease. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2025; 8:25152564251329250. [PMID: 40109887 PMCID: PMC11920999 DOI: 10.1177/25152564251329250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/22/2025]
Abstract
Mitochondria and lysosomes regulate a multitude of biological processes that are essential for the maintenance of nutrient and metabolic homeostasis and overall cell viability. Recent evidence reveals that these pivotal organelles, similarly to others previously studied, communicate through specialized membrane contact sites (MCSs), hereafter referred to as mitochondria-lysosome contacts (or MLCs), which promote their dynamic interaction without involving membrane fusion. Signal integration through MLCs is implicated in key processes, including mitochondrial fission and dynamics, and the exchange of calcium, cholesterol, and amino acids. Impairments in the formation and function of MLCs are increasingly associated with age-related diseases, specifically neurodegenerative disorders and lysosomal storage diseases. However, MLCs may play roles in other pathological contexts where lysosomes and mitochondria are crucial. In this review, we introduce the methodologies used to study MLCs and discuss known molecular players and key factors involved in their regulation in mammalian cells. We also argue other potential regulatory mechanisms depending on the acidic lysosomal pH and their impact on MLC's function. Finally, we explore the emerging implications of dysfunctional mitochondria-lysosome interactions in disease, highlighting their potential as therapeutic targets in cancer.
Collapse
Affiliation(s)
- Francesca Rizzollo
- Cell Death Research and Therapy Laboratory, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research and Therapy Laboratory, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
28
|
Yan HH, He JJ, Fu C, Chen JH, Tang AH. ATAD1 Regulates Neuronal Development and Synapse Formation Through Tuning Mitochondrial Function. Int J Mol Sci 2024; 26:44. [PMID: 39795902 PMCID: PMC11719905 DOI: 10.3390/ijms26010044] [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: 11/04/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
Mitochondrial function is essential for synaptic function. ATAD1, an AAA+ protease involved in mitochondrial quality control, governs fission-fusion dynamics within the organelle. However, the distribution and functional role of ATAD1 in neurons remain poorly understood. In this study, we demonstrate that ATAD1 is primarily localized to mitochondria in dendrites and, to a lesser extent, in spines in cultured hippocampal neurons. We found that ATAD1 deficiency disrupts the mitochondrial fission-fusion balance, resulting in mitochondrial fragmentation. This deficiency also impairs dendritic branching, hinders dendritic spine maturation, and reduces glutamatergic synaptic transmission in hippocampal neuron. To further investigate the underlying mechanism, we employed an ATP hydrolysis-deficient mutant of ATAD1 to rescue the neuronal deficits associated with ATAD1 loss. We discovered that the synaptic deficits are independent of the mitochondrial morphology changes but rely on its ATP hydrolysis. Furthermore, we show that ATAD1 loss leads to impaired mitochondrial function, including decreased ATP production, impaired membrane potential, and elevated oxidative stress. In conclusion, our results provide evidence that ATAD1 is crucial for maintaining mitochondrial function and regulating neurodevelopment and synaptic function.
Collapse
Affiliation(s)
- Hao-Hao Yan
- Hefei National Laboratory for Physical Sciences at the Microscale, MOE Key Laboratory for Membrane-Less Organelles & Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.-H.Y.); (J.-J.H.); (C.F.)
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- Neurodegenerative Disorder Research Center and Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China
| | - Jia-Jia He
- Hefei National Laboratory for Physical Sciences at the Microscale, MOE Key Laboratory for Membrane-Less Organelles & Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.-H.Y.); (J.-J.H.); (C.F.)
| | - Chuanhai Fu
- Hefei National Laboratory for Physical Sciences at the Microscale, MOE Key Laboratory for Membrane-Less Organelles & Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.-H.Y.); (J.-J.H.); (C.F.)
| | - Jia-Hui Chen
- Department of Anatomy, School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Ai-Hui Tang
- Hefei National Laboratory for Physical Sciences at the Microscale, MOE Key Laboratory for Membrane-Less Organelles & Cellular Dynamics, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; (H.-H.Y.); (J.-J.H.); (C.F.)
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230088, China
- Neurodegenerative Disorder Research Center and Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
29
|
Comyn T, Preat T, Pavlowsky A, Plaçais PY. Mitochondrial plasticity: An emergent concept in neuronal plasticity and memory. Neurobiol Dis 2024; 203:106740. [PMID: 39557174 DOI: 10.1016/j.nbd.2024.106740] [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: 10/04/2024] [Revised: 11/14/2024] [Accepted: 11/14/2024] [Indexed: 11/20/2024] Open
Abstract
Mitochondria are classically viewed as 'on demand' energy suppliers to neurons in support of their activity. In order to adapt to a wide range of demands, mitochondria need to be highly dynamic and capable of adjusting their metabolic activity, shape, and localization. Although these plastic properties give them a central support role in basal neuronal physiology, recent lines of evidence point toward a role for mitochondria in the regulation of high-order cognitive functions such as memory formation. In this review, we discuss the interplay between mitochondrial function and neural plasticity in sustaining memory formation at the molecular and cellular levels. First, we explore the global significance of mitochondria in memory formation. Then, we will detail the memory-relevant cellular and molecular mechanisms of mitochondrial plasticity. Finally, we focus on those mitochondrial functions, including but not limited to ATP production, that give mitochondria their pivotal role in memory formation. Altogether, this review highlights the central role of mitochondrial structural and functional plasticity in supporting and regulating neuronal plasticity and memory.
Collapse
Affiliation(s)
- Typhaine Comyn
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, 10 rue Vauquelin, 75005 Paris, France
| | - Thomas Preat
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, 10 rue Vauquelin, 75005 Paris, France.
| | - Alice Pavlowsky
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, 10 rue Vauquelin, 75005 Paris, France.
| | - Pierre-Yves Plaçais
- Energy & Memory, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, 10 rue Vauquelin, 75005 Paris, France.
| |
Collapse
|
30
|
Thomou C, Nussbaumer M, Grammenou E, Komini C, Vlaikou AM, Papageorgiou MP, Filiou MD. Early Handling Exerts Anxiolytic Effects and Alters Brain Mitochondrial Dynamics in Adult High Anxiety Mice. Mol Neurobiol 2024; 61:10593-10612. [PMID: 38761326 PMCID: PMC11584496 DOI: 10.1007/s12035-024-04116-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 03/09/2024] [Indexed: 05/20/2024]
Abstract
Early handling (EH), the brief separation of pups from their mother during early life, has been shown to exert beneficial effects. However, the impact of EH in a high anxiety background as well as the role of brain mitochondria in shaping EH-driven responses remain elusive.Here, we used a high (HAB) vs. normal (NAB) anxiety-related behavior mouse model to study how EH affects pup and dam behavior in divergent anxiety backgrounds. We also investigated EH-induced effects at the protein and mRNA levels in adult male HAB mice in the hypothalamus, the prefrontal cortex, and the hippocampus by examining the same mitochondrial/energy pathways and mitochondrial dynamics mechanisms (fission, fusion, biogenesis, and mitophagy) in all three brain regions.EH exerts anxiolytic effects in adult HAB but not NAB male mice and does not affect HAB or NAB maternal behavior, although basal HAB vs. NAB maternal behaviors differ. In adult HAB male mice, EH does not impact oxidative phosphorylation (OXPHOS) and oxidative stress in any of the brain regions studied but leads to increased protein expression of glycolysis enzymes and a correlation of anxiety-related behavior with Krebs cycle enzymes in HAB mice in the hypothalamus. Intriguingly, EH alters mitochondrial dynamics by increasing hypothalamic DRP1, OPA1, and PGC1a protein levels. At the mRNA level, we observe altered, EH-driven mitochondrial dynamics mRNA signatures which predominantly affect the prefrontal cortex.Taken together, our results show that EH exerts anxiolytic effects in adulthood in high anxiety and modulates mitochondrial dynamics pathways in a brain region-specific manner.
Collapse
Affiliation(s)
- Christina Thomou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| | - Markus Nussbaumer
- Laboratory of Biochemistry, Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| | - Eleni Grammenou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| | - Chrysoula Komini
- Laboratory of Biochemistry, Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| | - Angeliki-Maria Vlaikou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| | - Maria P Papageorgiou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece
| | - Michaela D Filiou
- Laboratory of Biochemistry, Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece.
- Biomedical Research Institute, Foundation for Research and Technology-Hellas (FORTH), Ioannina, Greece.
- Institute of Biosciences, University of Ioannina, Ioannina, Greece.
| |
Collapse
|
31
|
Cencelli G, Pedini G, Ricci C, Rosina E, Cecchetti G, Gentile A, Aiello G, Pacini L, Garrone B, Ombrato R, Coletta I, Prati F, Milanese C, Bagni C. Early dysregulation of GSK3β impairs mitochondrial activity in Fragile X Syndrome. Neurobiol Dis 2024; 203:106726. [PMID: 39510449 DOI: 10.1016/j.nbd.2024.106726] [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: 06/07/2024] [Revised: 11/03/2024] [Accepted: 11/03/2024] [Indexed: 11/15/2024] Open
Abstract
The finely tuned regulation of mitochondria activity is essential for proper brain development. Fragile X Syndrome (FXS), the leading cause of inherited intellectual disability, is a neurodevelopmental disorder in which mitochondrial dysfunction has been increasingly implicated. This study investigates the role of Glycogen Synthase Kinase 3β (GSK3β) in FXS. Several studies have reported the dysregulation of GSK3β in FXS, and its role in mitochondrial function is also well established. However, the link between disrupted GSK3β activity and mitochondrial dysfunction in FXS remains unexplored. Utilizing Fmr1 knockout (KO) mice and human cell lines from individuals with FXS, we uncovered a developmental window where dysregulated GSK3β activity disrupts mitochondrial function. Notably, a partial inhibition of GSK3β activity in FXS fibroblasts from young individuals rescues the observed mitochondrial defects, suggesting that targeting GSK3β in the early stages may offer therapeutic benefits for this condition.
Collapse
Affiliation(s)
- Giulia Cencelli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Giorgia Pedini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Carlotta Ricci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Eleonora Rosina
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Giorgia Cecchetti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Antonietta Gentile
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Giuseppe Aiello
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Laura Pacini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; Faculty of Medicine, UniCamillus, Saint Camillus International University of Health and Medical Sciences, 00131 Rome, Italy
| | | | | | | | | | | | - Claudia Bagni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland.
| |
Collapse
|
32
|
Wischhof L, Mathew AJ, Bonaguro L, Beyer M, Ehninger D, Nicotera P, Bano D. Mitochondrial complex I inhibition enhances astrocyte responsiveness to pro-inflammatory stimuli. Sci Rep 2024; 14:27182. [PMID: 39516523 PMCID: PMC11549212 DOI: 10.1038/s41598-024-78434-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Inhibition of the mitochondrial oxidative phosphorylation (OXPHOS) system can lead to metabolic disorders and neurodegenerative diseases. In primary mitochondrial disorders, reactive astrocytes often accompany neuronal degeneration and may contribute to neurotoxic inflammatory cascades that elicit brain lesions. The influence of mitochondria to astrocyte reactivity as well as the underlying molecular mechanisms remain elusive. Here we report that mitochondrial Complex I dysfunction promotes neural progenitor cell differentiation into astrocytes that are more responsive to neuroinflammatory stimuli. We show that the SWItch/Sucrose Non-Fermentable (SWI/SNF/BAF) chromatin remodeling complex takes part in the epigenetic regulation of astrocyte responsiveness, since its pharmacological inhibition abrogates the expression of inflammatory genes. Furthermore, we demonstrate that Complex I deficient human iPSC-derived astrocytes negatively influence neuronal physiology upon cytokine stimulation. Together, our data describe the SWI/SNF/BAF complex as a sensor of altered mitochondrial OXPHOS and a downstream epigenetic regulator of astrocyte-mediated neuroinflammation.
Collapse
Affiliation(s)
- Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany
| | - Amal John Mathew
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany
| | - Lorenzo Bonaguro
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn and West German Genome Center, Bonn, Germany
- Genomics and Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Marc Beyer
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn and West German Genome Center, Bonn, Germany
| | - Dan Ehninger
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany
| | - Pierluigi Nicotera
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany
| | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany.
| |
Collapse
|
33
|
Liu X, Wang J, Shen K, Jin W. p53/HIF-1α regulates neuronal aging and autophagy in spinal cord ischemia/reperfusion injury. Mech Ageing Dev 2024; 222:112000. [PMID: 39515667 DOI: 10.1016/j.mad.2024.112000] [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: 04/22/2024] [Revised: 09/11/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024]
Abstract
OBJECTION Spinal cord injury (SCI)-induced hindlimb dysfunction affects the physical and mental health of patients. There is growing evidence suggesting that the recovery capacity of elderly SCI patients is poorer than that of young individuals. However, the specific molecular mechanisms remain unclear. METHODS RNA expression profiles of SCI samples were collected from the GEO database, and key genes involved in the progression of SCI were identified by the limma package in R software. A diagnostic model based on SCIDEG was constructed using LASSO regression analysis. Subsequently, correlation analysis was conducted to identify biological pathways influenced by the key genes. Furthermore, SCI animal models were established in different age groups to examine the expression of relevant genes and verify the molecular mechanism of p53/HIF-1α axis. RESULTS We initially identified 34 ischemia-hypoxia-related genes potentially involved in the progression of SCI. Subsequently, we constructed a diagnostic model based on SCIDEGs using LASSO regression analysis. This model highlighted 9 key genes (TP53, SFTPA1, MASP2, KRT14, IL9, HIF1A, HGFAC, FUT7, and ALPP), which demonstrated high diagnostic accuracy in both the training set (AUC=1) and the validation set (AUC=0.855). Further cross-analysis with ischemia-reperfusion-related datasets confirmed the involvement of HIF1A and TP53. We also observed significant enrichment of HIF1A in organoids composed of mature neurons, which induced neuronal damage. In subsequent spinal cord injury animal models of different age groups, we found that HIF-1α expression was downregulated in the spinal cord tissues of elderly animals. Additionally, we discovered that TP53 activation induces cellular senescence in aging neurons and suppresses HIF-1α expression and autophagy levels within these cells. CONCLUSION In summary, our study suggests that the p53/HIF-1α signaling pathway plays a critical role in regulating neuronal aging and autophagy in the pathogenesis of SCI. Importantly, HIF-1α may represent a promising therapeutic target for SCI treatment.
Collapse
Affiliation(s)
- Xingzhen Liu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 201999, China
| | - Jia Wang
- Department of Pathology, Shanghai Xin Hua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200092, China
| | - Kangping Shen
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 201999, China.
| | - Wenjie Jin
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 201999, China.
| |
Collapse
|
34
|
Feng L, Li B, Yong SS, Wen X, Tian Z. The emerging role of exercise in Alzheimer's disease: Focus on mitochondrial function. Ageing Res Rev 2024; 101:102486. [PMID: 39243893 DOI: 10.1016/j.arr.2024.102486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by memory impairment and cognitive dysfunction, which eventually leads to the disability and mortality of older adults. Although the precise mechanisms by which age promotes the development of AD remains poorly understood, mitochondrial dysfunction plays a central role in the development of AD. Currently, there is no effective treatment for this debilitating disease. It is well accepted that exercise exerts neuroprotective effects by ameliorating mitochondrial dysfunction in the neurons of AD, which involves multiple mechanisms, including mitochondrial dynamics, biogenesis, mitophagy, transport, and signal transduction. In addition, exercise promotes mitochondria communication with other organelles in AD neurons, which should receive more attentions in the future.
Collapse
Affiliation(s)
- Lili Feng
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China.
| | - Bowen Li
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China
| | - Su Sean Yong
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China
| | - Xu Wen
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China.
| | - Zhenjun Tian
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
35
|
Cicali KA, Tapia-Rojas C. Synaptic mitochondria: A crucial factor in the aged hippocampus. Ageing Res Rev 2024; 101:102524. [PMID: 39369797 DOI: 10.1016/j.arr.2024.102524] [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: 08/27/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 10/08/2024]
Abstract
Aging is a multifaceted biological process characterized by progressive molecular and cellular damage accumulation. The brain hippocampus undergoes functional deterioration with age, caused by cellular deficits, decreased synaptic communication, and neuronal death, ultimately leading to memory impairment. One of the factors contributing to this dysfunction is the loss of mitochondrial function. In neurons, mitochondria are categorized into synaptic and non-synaptic pools based on their location. Synaptic mitochondria, situated at the synapses, play a crucial role in maintaining neuronal function and synaptic plasticity, whereas non-synaptic mitochondria are distributed throughout other neuronal compartments, supporting overall cellular metabolism and energy supply. The proper function of synaptic mitochondria is essential for synaptic transmission as they provide the energy required and regulate calcium homeostasis at the communication sites between neurons. Maintaining the structure and functionality of synaptic mitochondria involves intricate processes, including mitochondrial dynamics such as fission, fusion, transport, and quality control mechanisms. These processes ensure that mitochondria remain functional, replace damaged organelles, and sustain cellular homeostasis at synapses. Notably, deficiencies in these mechanisms have been increasingly associated with aging and the onset of age-related neurodegenerative diseases. Synaptic mitochondria from the hippocampus are particularly vulnerable to age-related changes, including alterations in morphology and a decline in functionality, which significantly contribute to decreased synaptic activity during aging. This review comprehensively explores the critical roles that mitochondrial dynamics and quality control mechanisms play in preserving synaptic activity and neuronal function. It emphasizes the emerging evidence linking the deterioration of synaptic mitochondria to the aging process and the development of neurodegenerative diseases, highlighting the importance of these organelles from hippocampal neurons as potential therapeutic targets for mitigating cognitive decline and synaptic degeneration associated with aging. The novelty of this review lies in its focus on the unique vulnerability of hippocampal synaptic mitochondria to aging, underscoring their importance in maintaining brain function across the lifespan.
Collapse
Affiliation(s)
- Karina A Cicali
- Laboratory of Neurobiology of Aging, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba, Santiago 8580702, Chile; Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile
| | - Cheril Tapia-Rojas
- Laboratory of Neurobiology of Aging, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba, Santiago 8580702, Chile; Facultad de Medicina y Ciencia, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile.
| |
Collapse
|
36
|
Xia K, Jin Z, Qiu Q, Zhou Y, Lu Y, Qiu T, Zhou J, Chen Z. Ligustilide alleviates oxidative stress during renal ischemia-reperfusion injury through maintaining Sirt3-dependent mitochondrial homeostasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155975. [PMID: 39216302 DOI: 10.1016/j.phymed.2024.155975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/29/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Renal ischemia-reperfusion (I/R) injury is an inevitable complication during renal transplantation and is closely related to patient prognosis. Mitochondrial damage induced oxidative stress is the core link of renal I/R injury. Ligustilide (LIG), a natural compound extracted from ligusticum chuanxiong hort and angelica sinensis, has exhibited the potential to protect mitochondrial function. However, whether LIG can ameliorate renal I/R injury requires further investigation. Delving deeper into the precise targets and mechanisms of LIG's effect on renal I/R injury is crucial. PURPOSE This study aimed to elucidate the specific mechanism of LIG's protective effect on renal I/R injury. METHODS In this study, an in vivo model of renal ischemia-reperfusion (I/R) injury was developed in mice, along with an in vitro model of hypoxia-reoxygenation (H/R) using human proximal renal tubular epithelial cells (HK-2). To assess the impact of LIG on renal injury, various methods were employed, including serum creatinine (Cr) and blood urea nitrogen (BUN) testing, hematoxylin and eosin (HE) staining, and immunohistochemistry (IHC) for kidney injury molecule-1 (KIM-1). The effects of LIG on oxidative stress were examined using fluorescent probes dihydroethidium (DHE) and dichlorodihydrofluorescein diacetate (DCFH-DA), TdT-mediated dUTP Nick-End Labeling (TUNEL) staining, and flow cytometry. Additionally, the influence of LIG on mitochondrial morphology and function was evaluated through transmission electron microscopy (TEM), Mito Tracker Red CMXRos staining, adenosine triphosphate (ATP) concentration assays, and JC-1 staining. The potential mechanism involving LIG and Sirt3 was explored by manipulating Sirt3 expression through cell transfection. RESULTS The results showed that LIG could provide protective function for mitochondria to alleviate oxidative stress induced by renal I/R. Further mechanistic studies indicated that LIG maintained mitochondrial homeostasis by targeting Sirt3. CONCLUSION Our findings demonstrated that LIG alleviated oxidative stress during renal I/R injury through maintaining Sirt3-dependent mitochondrial homeostasis. Overall, our data raised the possibility of LIG as a novel therapy for renal I/R injury.
Collapse
Affiliation(s)
- Kang Xia
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zeya Jin
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qiangmin Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yujie Zhou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yifan Lu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Jiangqiao Zhou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Zhongbao Chen
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
37
|
Teranishi M, Ito M, Huang Z, Nishiyama Y, Masuda A, Mino H, Tachibana M, Inada T, Ohno K. Extremely Low-Frequency Electromagnetic Field (ELF-EMF) Increases Mitochondrial Electron Transport Chain Activities and Ameliorates Depressive Behaviors in Mice. Int J Mol Sci 2024; 25:11315. [PMID: 39457098 PMCID: PMC11508854 DOI: 10.3390/ijms252011315] [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: 09/08/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Compromised mitochondrial electron transport chain (ETC) activities are associated with depression in humans and rodents. However, the effects of the enhancement of mitochondrial ETC activities on depression remain elusive. We recently reported that an extremely low-frequency electromagnetic field (ELF-EMF) of as low as 10 μT induced hormetic activation of mitochondrial ETC complexes in human/mouse cultured cells and mouse livers. Chronic social defeat stress (CSDS) for 10 consecutive days caused behavioral defects mimicking depression in mice, and using an ELF-EMF for two to six weeks ameliorated them. CSDS variably decreased the mitochondrial ETC proteins in the prefrontal cortex (PFC) in 10 days, which were increased by an ELF-EMF in six weeks. CSDS had no effect on the mitochondrial oxygen consumption rate in the PFC in 10 days, but using an ELF-EMF for six weeks enhanced it. CSDS inactivated SOD2 by enhancing its acetylation and increased lipid peroxidation in the PFC. In contrast, the ELF-EMF activated the Sirt3-FoxO3a-SOD2 pathway and suppressed lipid peroxidation. Furthermore, CSDS increased markers for mitophagy, which was suppressed by the ELF-EMF in six weeks. The ELF-EMF exerted beneficial hormetic effects on mitochondrial energy production, mitochondrial antioxidation, and mitochondrial dynamics in a mouse model of depression. We envisage that an ELF-EMF is a promising therapeutic option for depression.
Collapse
Affiliation(s)
- Masaki Teranishi
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
| | - Mikako Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
| | - Zhizhou Huang
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
| | - Yuki Nishiyama
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
| | - Akio Masuda
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
| | - Hiroyuki Mino
- Division of Material Science (Physics), Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan;
| | - Masako Tachibana
- Department of Psychiatry, Nagoya University Hospital, Nagoya 466-8560, Japan;
| | - Toshiya Inada
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan;
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
- Graduate School of Nutritional Sciences, Nagoya University of Arts and Sciences, Nisshin 470-0196, Japan
| |
Collapse
|
38
|
Li LG, Zhang D, Huang Q, Yan M, Chen NN, Yang Y, Xiao RC, Liu H, Han N, Qureshi AM, Hu J, Leng F, Hui YJ. Mitochondrial disruption resulting from Cepharanthine-mediated TOM inhibition triggers ferroptosis in colorectal cancer cells. J Cancer Res Clin Oncol 2024; 150:460. [PMID: 39402386 PMCID: PMC11478973 DOI: 10.1007/s00432-024-05974-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Chemotherapy for colorectal cancer (CRC) urgently needs low-toxicity and highly effective phytomedicine. Cepharanthine (Cep) shown to have multiple anti-tumor effects, including colorectal cancer, whose pivotal mechanisms are not fully understood. Herein, the present work aims to reveal the impact of Cep on the mitochondrial and anti-injury functions of CRC cells. METHODS The TOM70/20 expression was screened by bioinformatic databases. SW480 cells were utilized as the colorectal cancer cell model. The expression of TOM70/20 and the downstream molecules were measured by western blots (WB). The ferroptosis was analyzed using Transmission electron microscopy (TEM), C11-BODIPY, PGSK, and DCFH-DA probes, wherein the detection was performed by flow cytometry and laser confocal microscopy. The anti-cancer efficacy was conducted by CCK-8 and Annexin-V/PI assay. The rescue experiments were carried out using Fer-1 and TOM70 plasmid transfection. RESULTS Bioinformatic data identified TOM20 and TOM70 were highly expressed in colorectal cancer, which could be down-regulated by Cep. Further findings disclosed that Cep treatment destroyed the mitochondria and inactivated the NRF2 signaling pathway, an essential pathway for resistance to ferroptosis, thereby promoting reactive oxygen species (ROS) generation in CRC cells. As a result, prominent ferroptosis could be observed in CRC cells in response to Cep, which thereby led to the reduced cell viability of cancer cells. On the contrary, recovery of TOM70 dampened the Cep-elicited mitochondria damage, ferroptosis, and anti-cancer efficacy. CONCLUSION In summary, Cep-mediated TOM inhibition inactivates the NRF2 signaling pathway, thereby triggering ferroptosis and achieving an anti-colorectal cancer effect. The current study provides an innovative chemotherapeutic approach for colorectal cancer with phytomedicine.
Collapse
Affiliation(s)
- Liu-Gen Li
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, 442000, Hubei, China
| | - Di Zhang
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Renmin South Road No. 32, Shiyan, 442000, Hubei, China
| | - Qi Huang
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, 442000, Hubei, China
| | - Min Yan
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, 442000, Hubei, China
| | - Nan-Nan Chen
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, 442000, Hubei, China
| | - Yan Yang
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Renmin South Road No. 32, Shiyan, 442000, Hubei, China
| | - Rong-Cheng Xiao
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Renmin South Road No. 32, Shiyan, 442000, Hubei, China
| | - Hui Liu
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Renmin South Road No. 32, Shiyan, 442000, Hubei, China
| | - Ning Han
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, 442000, Hubei, China
| | - Abdul Moiz Qureshi
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Renmin South Road No. 32, Shiyan, 442000, Hubei, China
| | - Jun Hu
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, 442000, Hubei, China
| | - Fan Leng
- Shiyan Key Laboratory of Natural Medicine Nanoformulation Research, Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Renmin Road No. 30, Shiyan, 442000, Hubei, China
| | - Yuan-Jian Hui
- Department of General Surgery, Taihe Hospital, Hubei University of Medicine, Renmin South Road No. 32, Shiyan, 442000, Hubei, China.
| |
Collapse
|
39
|
Ghoochani A, Heiby JC, Rawat ES, Medoh UN, Di Fraia D, Dong W, Gastou M, Nyame K, Laqtom NN, Gomez-Ospina N, Ori A, Abu-Remaileh M. Cell-Type Resolved Protein Atlas of Brain Lysosomes Identifies SLC45A1-Associated Disease as a Lysosomal Disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618295. [PMID: 39464040 PMCID: PMC11507716 DOI: 10.1101/2024.10.14.618295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Mutations in lysosomal genes cause neurodegeneration and neurological lysosomal storage disorders (LSDs). Despite their essential role in brain homeostasis, the cell-type-specific composition and function of lysosomes remain poorly understood. Here, we report a quantitative protein atlas of the lysosome from mouse neurons, astrocytes, oligodendrocytes, and microglia. We identify dozens of novel lysosomal proteins and reveal the diversity of the lysosomal composition across brain cell types. Notably, we discovered SLC45A1, mutations in which cause a monogenic neurological disease, as a neuron-specific lysosomal protein. Loss of SLC45A1 causes lysosomal dysfunction in vitro and in vivo. Mechanistically, SLC45A1 plays a dual role in lysosomal sugar transport and stabilization of V1 subunits of the V-ATPase. SLC45A1 deficiency depletes the V1 subunits, elevates lysosomal pH, and disrupts iron homeostasis causing mitochondrial dysfunction. Altogether, our work redefines SLC45A1-associated disease as a LSD and establishes a comprehensive map to study lysosome biology at cell-type resolution in the brain and its implications for neurodegeneration.
Collapse
Affiliation(s)
- Ali Ghoochani
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network; Chevy Chase, MD, 20815, USA
- These authors contributed equally
| | - Julia C. Heiby
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI) e.V., Jena, Germany
- These authors contributed equally
| | - Eshaan S. Rawat
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network; Chevy Chase, MD, 20815, USA
| | - Uche N. Medoh
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network; Chevy Chase, MD, 20815, USA
- Current affiliation: Arc Institute, Palo Alto, CA 94304, USA
| | - Domenico Di Fraia
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI) e.V., Jena, Germany
- Current affiliation: Department of Biology, University of Rochester, Rochester, NY, USA
| | - Wentao Dong
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network; Chevy Chase, MD, 20815, USA
| | - Marc Gastou
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Kwamina Nyame
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network; Chevy Chase, MD, 20815, USA
| | - Nouf N. Laqtom
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network; Chevy Chase, MD, 20815, USA
| | - Natalia Gomez-Ospina
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Alessandro Ori
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI) e.V., Jena, Germany
- Co-senior authors
| | - Monther Abu-Remaileh
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA 94305, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network; Chevy Chase, MD, 20815, USA
- The Phil & Penny Knight Initiative for Brain Resilience at the Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
- Co-senior authors
- Lead author
| |
Collapse
|
40
|
Marino Y, Inferrera F, D'Amico R, Impellizzeri D, Cordaro M, Siracusa R, Gugliandolo E, Fusco R, Cuzzocrea S, Di Paola R. Role of mitochondrial dysfunction and biogenesis in fibromyalgia syndrome: Molecular mechanism in central nervous system. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167301. [PMID: 38878832 DOI: 10.1016/j.bbadis.2024.167301] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/22/2024] [Accepted: 06/07/2024] [Indexed: 08/18/2024]
Abstract
A critical role for mitochondrial dysfunction has been shown in the pathogenesis of fibromyalgia. It is a chronic pain syndrome characterized by neuroinflammation and impaired oxidative balance in the central nervous system. Boswellia serrata (BS), a natural polyphenol, is a well-known able to influence the mitochondrial metabolism. The objective of this study was to evaluate the mitochondrial dysfunction and biogenesis in fibromyalgia and their modulation by BS. To induce the model reserpine (1 mg/Kg) was subcutaneously administered for three consecutive days and BS (100 mg/Kg) was given orally for twenty-one days. BS reduced pain like behaviors in reserpine-injected rats and the astrocytes activation in the dorsal horn of the spinal cord and prefrontal cortex that are recognized as key regions associated with the neuropathic pain. Vulnerability to neuroinflammation and impaired neuronal plasticity have been described as consequences of mitochondrial dysfunction. BS administration increased PGC-1α expression in the nucleus of spinal cord and brain tissues, promoting the expression of regulatory genes for mitochondrial biogenesis (NRF-1, Tfam and UCP2) and cellular antioxidant defence mechanisms (catalase, SOD2 and Prdx 3). According with these data BS reduced lipid peroxidation and the GSSG/GSH ratio and increased SOD activity in the same tissues. Our results also showed that BS administration mitigates cytochrome-c leakage by promoting mitochondrial function and supported the movement of PGC-1α protein into the nucleus restoring the quality control of mitochondria. Additionally, BS reduced Drp1 and Fis1, preventing both mitochondrial fission and cell death, and increased the expression of Mfn2 protein, facilitating mitochondrial fusion. Overall, our results showed important mitochondrial dysfunction in central nervous system in fibromyalgia syndrome and the role of BS in restoring mitochondrial dynamics.
Collapse
Affiliation(s)
- Ylenia Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Francesca Inferrera
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Ramona D'Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, 98125 Messina, Italy.
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Enrico Gugliandolo
- Department of Veterinary Science, University of Messina, 98168 Messina, Italy.
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Rosanna Di Paola
- Department of Veterinary Science, University of Messina, 98168 Messina, Italy.
| |
Collapse
|
41
|
Zaninello M, Baptista P, Duarte FV. Mitochondrial Dynamics and mRNA Translation: A Local Synaptic Tale. BIOLOGY 2024; 13:746. [PMID: 39336173 PMCID: PMC11428642 DOI: 10.3390/biology13090746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Mitochondria are dynamic organelles that can adjust and respond to different stimuli within a cell. This plastic ability allows them to effectively coordinate several cellular functions in cells and becomes particularly relevant in highly complex cells such as neurons. An imbalance in mitochondrial dynamics can disrupt mitochondrial function, leading to abnormal cellular function and ultimately to a range of diseases, including neurodegenerative disorders. Regulation of mRNA transport and local translation inside neurons is crucial for maintaining the proteome of distal mitochondria, which is vital for energy production and synaptic function. A significant portion of the axonal transcriptome is dedicated to mRNAs for mitochondrial proteins, emphasizing the importance of local translation in sustaining mitochondrial function in areas far from the cell body. In neurons, local translation and the regulation of mRNAs encoding mitochondrial-shaping proteins could be essential for synaptic plasticity and neuronal health. The dynamics of these mRNAs, including their transport and local translation, may influence the morphology and function of mitochondria, thereby affecting the overall energy status and responsiveness of synapses. Comprehending the mitochondria-related mRNA regulation and local translation, as well as its influence on mitochondrial morphology near the synapses will help to better understand neuronal physiology and neurological diseases where mitochondrial dysfunction and impaired synaptic plasticity play a central role.
Collapse
Affiliation(s)
- Marta Zaninello
- Institute for Genetics, University of Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| | - Pedro Baptista
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Filipe V Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
42
|
Jusic A, Erpapazoglou Z, Dalgaard LT, Lakkisto P, de Gonzalo-Calvo D, Benczik B, Ágg B, Ferdinandy P, Fiedorowicz K, Schroen B, Lazou A, Devaux Y, on behalf of EU-CardioRNA COST Action CA17129, AtheroNET COST Action CA21153. Guidelines for mitochondrial RNA analysis. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102262. [PMID: 39091381 PMCID: PMC11292373 DOI: 10.1016/j.omtn.2024.102262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Mitochondria are the energy-producing organelles of mammalian cells with critical involvement in metabolism and signaling. Studying their regulation in pathological conditions may lead to the discovery of novel drugs to treat, for instance, cardiovascular or neurological diseases, which affect high-energy-consuming cells such as cardiomyocytes, hepatocytes, or neurons. Mitochondria possess both protein-coding and noncoding RNAs, such as microRNAs, long noncoding RNAs, circular RNAs, and piwi-interacting RNAs, encoded by the mitochondria or the nuclear genome. Mitochondrial RNAs are involved in anterograde-retrograde communication between the nucleus and mitochondria and play an important role in physiological and pathological conditions. Despite accumulating evidence on the presence and biogenesis of mitochondrial RNAs, their study continues to pose significant challenges. Currently, there are no standardized protocols and guidelines to conduct deep functional characterization and expression profiling of mitochondrial RNAs. To overcome major obstacles in this emerging field, the EU-CardioRNA and AtheroNET COST Action networks summarize currently available techniques and emphasize critical points that may constitute sources of variability and explain discrepancies between published results. Standardized methods and adherence to guidelines to quantify and study mitochondrial RNAs in normal and disease states will improve research outputs, their reproducibility, and translation potential to clinical application.
Collapse
Affiliation(s)
- Amela Jusic
- HAYA Therapeutics SA, Route De La Corniche 6, SuperLab Suisse - Batiment Serine, 1066 Epalinges, Switzerland
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1445 Strassen, Luxembourg
| | - Zoi Erpapazoglou
- Ιnstitute for Fundamental Biomedical Research, B.S.R.C. “Alexander Fleming”, Vari, 16672 Athens, Greece
| | - Louise Torp Dalgaard
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark
| | - Päivi Lakkisto
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
- Department of Clinical Chemistry, University of Helsinki and Helsinki University Hospital, 00014 Helsinki, Finland
| | - David de Gonzalo-Calvo
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, 25198 Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, 28029 Madrid, Spain
| | - Bettina Benczik
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Bence Ágg
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Péter Ferdinandy
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
| | | | - Blanche Schroen
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, ER 6229 Maastricht, the Netherlands
| | - Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1445 Strassen, Luxembourg
| | - on behalf of EU-CardioRNA COST Action CA17129
- HAYA Therapeutics SA, Route De La Corniche 6, SuperLab Suisse - Batiment Serine, 1066 Epalinges, Switzerland
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1445 Strassen, Luxembourg
- Ιnstitute for Fundamental Biomedical Research, B.S.R.C. “Alexander Fleming”, Vari, 16672 Athens, Greece
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
- Department of Clinical Chemistry, University of Helsinki and Helsinki University Hospital, 00014 Helsinki, Finland
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, 25198 Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, 28029 Madrid, Spain
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, 61614 Poznan, Poland
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, ER 6229 Maastricht, the Netherlands
- School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - AtheroNET COST Action CA21153
- HAYA Therapeutics SA, Route De La Corniche 6, SuperLab Suisse - Batiment Serine, 1066 Epalinges, Switzerland
- Cardiovascular Research Unit, Department of Precision Health, Luxembourg Institute of Health, 1445 Strassen, Luxembourg
- Ιnstitute for Fundamental Biomedical Research, B.S.R.C. “Alexander Fleming”, Vari, 16672 Athens, Greece
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
- Department of Clinical Chemistry, University of Helsinki and Helsinki University Hospital, 00014 Helsinki, Finland
- Translational Research in Respiratory Medicine, University Hospital Arnau de Vilanova and Santa Maria, IRBLleida, 25198 Lleida, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, 28029 Madrid, Spain
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, 61614 Poznan, Poland
- Department of Physiology, Cardiovascular Research Institute Maastricht, Maastricht University, ER 6229 Maastricht, the Netherlands
- School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
43
|
Bai H, Zhang S, Yang H, Wang J, Chen H, Li J, Li L, Yang Q, Peng B, Zhu Z, Ni S, Liu K, Lei W, Tao TH, Feng Y. Advanced nerve regeneration enabled by neural conformal electronic stimulators enhancing mitochondrial transport. Bioact Mater 2024; 39:287-301. [PMID: 38827170 PMCID: PMC11143791 DOI: 10.1016/j.bioactmat.2024.05.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/04/2024] [Accepted: 05/17/2024] [Indexed: 06/04/2024] Open
Abstract
Addressing peripheral nerve defects remains a significant challenge in regenerative neurobiology. Autografts emerged as the gold-standard management, however, are hindered by limited availability and potential neuroma formation. Numerous recent studies report the potential of wireless electronic system for nerve defects repair. Unfortunately, few has met clinical needs for inadequate electrode precision, poor nerve entrapment and insufficient bioactivity of the matrix material. Herein, we present an advanced wireless electrical nerve stimulator, based on water-responsive self-curling silk membrane with excellent bioabsorbable and biocompatible properties. We constructed a unique bilayer structure with an oriented pre-stretched inner layer and a general silk membrane as outer layer. After wetting, the simultaneous contraction of inner layer and expansion of outer layer achieved controllable super-contraction from 2D flat surface to 3D structural reconfiguration. It enables shape-adaptive wrapping to cover around nerves, overcomes the technical obstacle of preparing electrodes on the inner wall of the conduit, and prevents electrode breakage caused by material expansion in water. The use of fork capacitor-like metal interface increases the contact points between the metal and the regenerating nerve, solving the challenge of inefficient and rough electrical stimulation methods in the past. Newly developed electronic stimulator is effective in restoring 10 mm rat sciatic nerve defects comparable to autologous grafts. The underlying mechanism involves that electric stimulation enhances anterograde mitochondrial transport to match energy demands. This newly introduced device thereby demonstrated the potential as a viable and efficacious alternative to autografts for enhancing peripheral nerve repair and functional recovery.
Collapse
Affiliation(s)
- Hao Bai
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Siqi Zhang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Huiran Yang
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Jing Wang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Hongli Chen
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jia Li
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, China
| | - Lin Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen, 361005, Fujian, China
| | - Qian Yang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an, 710072, PR China
| | - Ziyi Zhu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Siyuan Ni
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Keyin Liu
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Wei Lei
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Tiger H. Tao
- State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, 200031, China
- Institute of Brain-Intelligence Technology, Zhangjiang Laboratory, Shanghai, 200031, China
| | - Yafei Feng
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
44
|
Gong QY, Wang W, Cai L, Jing Y, Yang DX, Yuan F, Tian HL, Ding J, Chen H, Xu ZM. Transplantation of astrocyte-derived mitochondria into injured astrocytes has a protective effect following stretch injury. Mitochondrion 2024; 78:101902. [PMID: 38768694 DOI: 10.1016/j.mito.2024.101902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 05/04/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
Traumatic brain injury (TBI) is a global public-health problem. Astrocytes, and their mitochondria, are important factors in the pathogenesis of TBI-induced secondary injury. Mitochondria extracted from healthy tissues and then transplanted have shown promise in models of a variety of diseases. However, the effect on recipient astrocytes is unclear. Here, we isolated primary astrocytes from newborn C57BL/6 mice, one portion of which was used to isolate mitochondria, and another was subjected to stretch injury (SI) followed by transplantation of the isolated mitochondria. After incubation for 12 h, cell viability, mitochondrial dysfunction, calcium overload, redox stress, inflammatory response, and apoptosis were improved. Live-cell imaging showed that the transplanted mitochondria were incorporated into injured astrocytes and fused with their mitochondrial networks, which was in accordance with the changes in the expression levels of markers of mitochondrial dynamics. The astrocytic IKK/NF-κB pathway was decelerated whereas the AMPK/PGC-1α pathway was accelerated by transplantation. Together, these results indicate that exogenous mitochondria from untreated astrocytes can be incorporated into injured astrocytes and fuse with their mitochondrial networks, improving cell viability by ameliorating mitochondrial dysfunction, redox stress, calcium overload, and inflammation.
Collapse
Affiliation(s)
- Qiu-Yuan Gong
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wei Wang
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lin Cai
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yao Jing
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Dian-Xu Yang
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Fang Yuan
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Heng-Li Tian
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jun Ding
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Hao Chen
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhi-Ming Xu
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
45
|
Tang T, Hu LB, Ding C, Zhang Z, Wang N, Wang T, Zhou H, Xia S, Fan L, Fu XJ, Yan F, Zhang X, Chen G, Li J. Src inhibition rescues FUNDC1-mediated neuronal mitophagy in ischaemic stroke. Stroke Vasc Neurol 2024; 9:367-379. [PMID: 37793899 PMCID: PMC11420917 DOI: 10.1136/svn-2023-002606] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 09/15/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Ischaemic stroke triggers neuronal mitophagy, while the involvement of mitophagy receptors in ischaemia/reperfusion (I/R) injury-induced neuronal mitophagy remain not fully elucidated. Here, we aimed to investigate the involvement of mitophagy receptor FUN14 domain-containing 1 (FUNDC1) and its modulation in neuronal mitophagy induced by I/R injury. METHODS Wild-type and FUNDC1 knockout mice were generated to establish models of neuronal I/R injury, including transient middle cerebral artery occlusion (tMCAO) in vivo and oxygen glucose deprivation/reperfusion in vitro. Stroke outcomes of mice with two genotypes were assessed. Neuronal mitophagy was analysed both in vivo and in vitro. Activities of FUNDC1 and its regulator Src were evaluated. The impact of Src on FUNDC1-mediated mitophagy was assessed through administration of Src antagonist PP1. RESULTS To our surprise, FUNDC1 knockout mice subjected to tMCAO showed stroke outcomes comparable to those of their wild-type littermates. Although neuronal mitophagy could be activated by I/R injury, FUNDC1 deletion did not disrupt neuronal mitophagy. Transient activation of FUNDC1, represented by dephosphorylation of Tyr18, was detected in the early stages (within 3 hours) of neuronal I/R injury; however, phosphorylated Tyr18 reappeared and even surpassed baseline levels in later stages (after 6 hours), accompanied by a decrease in FUNDC1-light chain 3 interactions. Spontaneous inactivation of FUNDC1 was associated with Src activation, represented by phosphorylation of Tyr416, which changed in parallel with the level of phosphorylated FUNDC1 (Tyr18) during neuronal I/R injury. Finally, FUNDC1-mediated mitophagy in neurons under I/R conditions can be rescued by pharmacological inhibition of Src. CONCLUSIONS FUNDC1 is inactivated by Src during the later stage (after 6 hours) of neuronal I/R injury, and rescue of FUNDC1-mediated mitophagy may serve as a potential therapeutic strategy for treating ischaemic stroke.
Collapse
Affiliation(s)
- Tianchi Tang
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Li-Bin Hu
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Chao Ding
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhihua Zhang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ning Wang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tingting Wang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hang Zhou
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Siqi Xia
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Linfeng Fan
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Xiong-Jie Fu
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Feng Yan
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Xiangnan Zhang
- Zhejiang University Department of Pharmacology, Hangzhou, Zhejiang, China
| | - Gao Chen
- Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Jianru Li
- Department of Neurosurgery, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
46
|
Yu SB, Wang H, Sanchez RG, Carlson NM, Nguyen K, Zhang A, Papich ZD, Abushawish AA, Whiddon Z, Matysik W, Zhang J, Whisenant TC, Ghassemian M, Koberstein JN, Stewart ML, Myers SA, Pekkurnaz G. Neuronal activity-driven O-GlcNAcylation promotes mitochondrial plasticity. Dev Cell 2024; 59:2143-2157.e9. [PMID: 38843836 PMCID: PMC11338717 DOI: 10.1016/j.devcel.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/15/2024] [Accepted: 05/09/2024] [Indexed: 06/18/2024]
Abstract
Neuronal activity is an energy-intensive process that is largely sustained by instantaneous fuel utilization and ATP synthesis. However, how neurons couple ATP synthesis rate to fuel availability is largely unknown. Here, we demonstrate that the metabolic sensor enzyme O-linked N-acetyl glucosamine (O-GlcNAc) transferase regulates neuronal activity-driven mitochondrial bioenergetics in hippocampal and cortical neurons. We show that neuronal activity upregulates O-GlcNAcylation in mitochondria. Mitochondrial O-GlcNAcylation is promoted by activity-driven glucose consumption, which allows neurons to compensate for high energy expenditure based on fuel availability. To determine the proteins that are responsible for these adjustments, we mapped the mitochondrial O-GlcNAcome of neurons. Finally, we determine that neurons fail to meet activity-driven metabolic demand when O-GlcNAcylation dynamics are prevented. Our findings suggest that O-GlcNAcylation provides a fuel-dependent feedforward control mechanism in neurons to optimize mitochondrial performance based on neuronal activity. This mechanism thereby couples neuronal metabolism to mitochondrial bioenergetics and plays a key role in sustaining energy homeostasis.
Collapse
Affiliation(s)
- Seungyoon B Yu
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Haoming Wang
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Richard G Sanchez
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Natasha M Carlson
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Khanh Nguyen
- Laboratory for Immunochemical Circuits, Center of Autoimmunity and Inflammation, and Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA 92093, USA
| | - Andrew Zhang
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Zachary D Papich
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Ahmed A Abushawish
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Zachary Whiddon
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Weronika Matysik
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Jie Zhang
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Thomas C Whisenant
- Center for Computational Biology and Bioinformatics, University of California San Diego, La Jolla, CA 92093, USA
| | - Majid Ghassemian
- Biomolecular and Proteomics Mass Spectrometry Facility, University of California San Diego, La Jolla, CA 92093, USA
| | - John N Koberstein
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Melissa L Stewart
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Samuel A Myers
- Laboratory for Immunochemical Circuits, Center of Autoimmunity and Inflammation, and Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA 92093, USA; Department of Pharmacology, Program in Immunology, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Gulcin Pekkurnaz
- Neurobiology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
47
|
Begeman A, Smolka JA, Shami A, Waingankar TP, Lewis SC. A spatial atlas of mitochondrial gene expression reveals dynamic translation hubs and remodeling in stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.604215. [PMID: 39149346 PMCID: PMC11326164 DOI: 10.1101/2024.08.05.604215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Mitochondrial genome expression is important for cellular bioenergetics. How mitochondrial RNA processing and translation are spatially organized across dynamic mitochondrial networks is not well understood. Here, we report that processed mitochondrial RNAs are consolidated with mitoribosome components into translation hubs distal to either nucleoids or processing granules in human cells. During stress, these hubs are remodeled into translationally repressed mesoscale bodies containing messenger, ribosomal, and double-stranded RNA. We show that the highly conserved helicase SUV3 contributes to the distribution of processed RNA within mitochondrial networks, and that stress bodies form downstream of proteostatic stress in cells lacking SUV3 unwinding activity. We propose that the spatial organization of nascent chain synthesis into discrete domains serves to throttle the flow of genetic information in stress to ensure mitochondrial quality control.
Collapse
Affiliation(s)
- Adam Begeman
- Department of Molecular and Cell Biology, University of California, Berkeley, CA USA
| | - John A. Smolka
- Department of Molecular and Cell Biology, University of California, Berkeley, CA USA
| | - Ahmad Shami
- Department of Molecular and Cell Biology, University of California, Berkeley, CA USA
| | | | - Samantha C. Lewis
- Department of Molecular and Cell Biology, University of California, Berkeley, CA USA
- Innovative Genomics Institute, Berkeley, CA, USA
- Helen Wills Neuroscience Institute, Berkeley, CA USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA USA
| |
Collapse
|
48
|
Lu Y, Jiang Y, Wang F, Wu H, Hua Y. Electron Microscopic Mapping of Mitochondrial Morphology in the Cochlear Nerve Fibers. J Assoc Res Otolaryngol 2024; 25:341-354. [PMID: 38937328 PMCID: PMC11349726 DOI: 10.1007/s10162-024-00957-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: 01/08/2024] [Accepted: 06/12/2024] [Indexed: 06/29/2024] Open
Abstract
To enable nervous system function, neurons are powered in a use-dependent manner by mitochondria undergoing morphological-functional adaptation. In a well-studied model system-the mammalian cochlea, auditory nerve fibers (ANFs) display distinct electrophysiological properties, which is essential for collectively sampling acoustic information of a large dynamic range. How exactly the associated mitochondrial networks are deployed in functionally differentiated ANFs remains scarcely interrogated. Here, we leverage volume electron microscopy and machine-learning-assisted image analysis to phenotype mitochondrial morphology and distribution along ANFs of full-length in the mouse cochlea inner spiral bundle. This reveals greater variance in mitochondrial size with increased ANF habenula to terminal path length. Particularly, we analyzed the ANF terminal-residing mitochondria, which are critical for local calcium uptake during sustained afferent activities. Our results suggest that terminal-specific enrichment of mitochondria, in addition to terminal size and overall mitochondrial abundance of the ANF, correlates with heterogenous mitochondrial contents of the terminal.
Collapse
Affiliation(s)
- Yan Lu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Jiang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangfang Wang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunfeng Hua
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai, China.
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Translational Medicine on Ear and Nose Diseases, Shanghai, China.
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
49
|
Wang Y, Yang JS, Zhao M, Chen JQ, Xie HX, Yu HY, Liu NH, Yi ZJ, Liang HL, Xing L, Jiang HL. Mitochondrial endogenous substance transport-inspired nanomaterials for mitochondria-targeted gene delivery. Adv Drug Deliv Rev 2024; 211:115355. [PMID: 38849004 DOI: 10.1016/j.addr.2024.115355] [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: 03/18/2024] [Revised: 05/16/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Mitochondrial genome (mtDNA) independent of nuclear gene is a set of double-stranded circular DNA that encodes 13 proteins, 2 ribosomal RNAs and 22 mitochondrial transfer RNAs, all of which play vital roles in functions as well as behaviors of mitochondria. Mutations in mtDNA result in various mitochondrial disorders without available cures. However, the manipulation of mtDNA via the mitochondria-targeted gene delivery faces formidable barriers, particularly owing to the mitochondrial double membrane. Given the fact that there are various transport channels on the mitochondrial membrane used to transfer a variety of endogenous substances to maintain the normal functions of mitochondria, mitochondrial endogenous substance transport-inspired nanomaterials have been proposed for mitochondria-targeted gene delivery. In this review, we summarize mitochondria-targeted gene delivery systems based on different mitochondrial endogenous substance transport pathways. These are categorized into mitochondrial steroid hormones import pathways-inspired nanomaterials, protein import pathways-inspired nanomaterials and other mitochondria-targeted gene delivery nanomaterials. We also review the applications and challenges involved in current mitochondrial gene editing systems. This review delves into the approaches of mitochondria-targeted gene delivery, providing details on the design of mitochondria-targeted delivery systems and the limitations regarding the various technologies. Despite the progress in this field is currently slow, the ongoing exploration of mitochondrial endogenous substance transport and mitochondrial biological phenomena may act as a crucial breakthrough in the targeted delivery of gene into mitochondria and even the manipulation of mtDNA.
Collapse
Affiliation(s)
- Yi Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Jing-Song Yang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Min Zhao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Jia-Qi Chen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Hai-Xin Xie
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Hao-Yuan Yu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Na-Hui Liu
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Zi-Juan Yi
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Hui-Lin Liang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China; College of Pharmacy, Yanbian University, Yanji 133002, China.
| |
Collapse
|
50
|
Kuznetsov IA, Kuznetsov AV. Mitochondrial transport in symmetric and asymmetric axons with multiple branching junctions: a computational study. Comput Methods Biomech Biomed Engin 2024; 27:1071-1090. [PMID: 37424316 PMCID: PMC10776827 DOI: 10.1080/10255842.2023.2226787] [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: 04/03/2023] [Revised: 05/10/2023] [Accepted: 06/10/2023] [Indexed: 07/11/2023]
Abstract
Mitochondrial aging has been proposed to be involved in a variety of neurodegenerative disorders, such as Parkinson's disease. Here, we explore the impact of multiple branching junctions in axons on the mean age of mitochondria and their age density distributions in demand sites. The study examined mitochondrial concentration, mean age, and age density distribution in relation to the distance from the soma. We developed models for a symmetric axon containing 14 demand sites and an asymmetric axon containing 10 demand sites. We investigated how the concentration of mitochondria changes when an axon splits into two branches at the branching junction. Additionally, we studied whether mitochondrial concentrations in the branches are affected by what proportion of mitochondrial flux enters the upper branch versus the lower branch. Furthermore, we explored whether the distributions of mitochondrial mean age and age density in branching axons are affected by how the mitochondrial flux splits at the branching junction. When the mitochondrial flux is unevenly split at the branching junction of an asymmetric axon, with a greater proportion of the flux entering the longer branch, the average age of mitochondria (system age) in the axon increases. Our findings elucidate the effects of axonal branching on the mitochondrial age.
Collapse
Affiliation(s)
- Ivan A Kuznetsov
- Perelman School of Medicine, University of PA, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrey V Kuznetsov
- Department of Mechanical and Aerospace Engineering, NC State University, Raleigh, NC, USA
| |
Collapse
|