1
|
Mishra R, Upadhyay A. An update on mammalian and non-mammalian animal models for biomarker development in neurodegenerative disorders. Cell Mol Life Sci 2025; 82:147. [PMID: 40192808 PMCID: PMC11977071 DOI: 10.1007/s00018-025-05668-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 03/08/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025]
Abstract
Neurodegeneration is one of the leading factor for death globally, affecting millions of people. Developing animal models are critical to understand biological processes and comprehend pathological hallmarks of neurodegenerative diseases. For decades, many animal models have served as excellent tools to determine the disease progression, develop diagnostic methods and design novel therapies against distinct pathologies. Here, we provide a comprehensive overview of both, mammalian and non-mammalian animal models, with a focus on three most common and aggressive neurodegenerative disorders: Alzheimer's disease, Parkinson's disease and Spinocerebellar ataxia-1. We highlight various approaches including transgene, gene transfer, and chemically-induced methods used to develop disease models. In particular, we discuss applications of both non-mammalian and mammalian contributions in research on neurodegeneration. It is exciting to learn the roles of animal models in disease pathomechanisms, identifying biomarkers and hence devising novel interventions to treat neuropathological conditions.
Collapse
Affiliation(s)
- Ribhav Mishra
- School of Health Sciences, Purdue University, West Lafayette, IN, USA.
| | - Arun Upadhyay
- Department of Bioscience and Biomedical Engineering, Indian Institute of Technology Bhilai, Chhattisgarh, 491002, India
| |
Collapse
|
2
|
Mingo YB, Escobar Galvis ML, Henderson MX. α-Synuclein pathology and mitochondrial dysfunction: Toxic partners in Parkinson's disease. Neurobiol Dis 2025; 209:106889. [PMID: 40157617 DOI: 10.1016/j.nbd.2025.106889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/26/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025] Open
Abstract
Two major neuropathological features of Parkinson's disease (PD) are α-synuclein Lewy pathology and mitochondrial dysfunction. Although both α-synuclein pathology and mitochondrial dysfunction may independently contribute to PD pathogenesis, the interaction between these two factors is not yet fully understood. In this review, we discuss the physiological functions of α-synuclein and mitochondrial homeostasis in neurons as well as the pathological defects that ensue when these functions are disturbed in PD. Recent studies have highlighted that dysfunctional mitochondria can become sequestered within Lewy bodies, and cell biology studies have suggested that α-synuclein can directly impair mitochondrial function. There are also PD cases caused by genetic or environmental perturbation of mitochondrial homeostasis. Together, these studies suggest that mitochondrial dysfunction may be a common pathway to neurodegeneration in PD, triggered by multiple insults. We review the literature surrounding the interaction between α-synuclein and mitochondria and highlight open questions in the field that may be explored to advance our understanding of PD and develop novel, disease-modifying therapies.
Collapse
Affiliation(s)
- Yakum B Mingo
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States of America
| | | | - Michael X Henderson
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States of America.
| |
Collapse
|
3
|
Samidurai M, Chennakesavan K, Sarkar S, Malovic E, Nguyen HM, Singh L, Kumar A, Ealy A, Janarthanam C, Palanisamy BN, Kondru N, Zenitsky G, Jin H, Anantharam V, Kanthasamy A, Zhang H, Wulff H, Kanthasamy A. KCa3.1 Contributes to Neuroinflammation and Nigral Dopaminergic Neurodegeneration in Experimental models of Parkinson's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643982. [PMID: 40166152 PMCID: PMC11956954 DOI: 10.1101/2025.03.18.643982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Chronic neuroinflammation and misfolded α-synuclein (αSyn) have been identified as key pathological correlates driving Parkinson's disease (PD) pathogenesis; however, the contribution of ion channels to microglia activation in the context of α-synucleinopathy remains elusive. Herein, we show that KCa3.1, a calcium-activated potassium channel, is robustly upregulated within microglia in multiple preclinical models of PD and, most importantly, in human PD and dementia with Lewy bodies (DLB) brains. Pharmacological inhibition of KCa3.1 via senicapoc or TRAM-34 inhibits KCa3.1 channel activity and the associated reactive microglial phenotype in response to aggregated αSyn, as well as ameliorates of PD like pathology in diverse PD mouse models. Additionally, proteomic and transcriptomic profiling of microglia revealed that senicapoc ameliorates aggregated αSyn-induced, inflammation-associated pathways and dysregulated metabolism in primary microglial cells. Mechanistically, FYN kinase in a STAT1 dependent manner regulates KCa3.1 mediated the microglial reactive activation phenotype after α-synucleinopathy. Moreover, reduced neuroinflammation and subsequent PD-like neuropathology were observed in SYN AAV inoculated KCa3.1 knockout mice. Together, these findings suggest that KCa3.1 inhibition represents a novel therapeutic strategy for treating patients with PD and related α-synucleinopathies.
Collapse
|
4
|
Sáenz de Miera C, Bellefontaine N, Silveira MA, Fortin CN, Zampieri TT, Donato J, Williams KW, Mendes-da-Silva C, Heikkinen L, Broberger C, Frazao R, Elias CF. Nutritionally responsive PMv DAT neurons are dynamically regulated during pubertal transition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636271. [PMID: 39975315 PMCID: PMC11838509 DOI: 10.1101/2025.02.03.636271] [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
Pubertal development is tightly regulated by energy balance. The crosstalk between metabolism and reproduction is orchestrated by complex neural networks and leptin action in the hypothalamus plays a critical role. The ventral premammillary nucleus (PMv) leptin receptor (LepRb) neurons act as an essential relay for leptin action on reproduction. Here, we show that mouse PMv cells expressing the dopamine transporter (DAT) gene, Slc6a3 (PMvDAT) form a novel subpopulation of LepRb neurons. Virtually all PMvDAT neurons expressed Lepr mRNA and responded to acute leptin treatment. Electrophysiological recordings from DATCRE;tdTomato mice showed that PMvDAT cells in prepubertal females have a hyperpolarized resting membrane potential compared to diestrous females. Slc6a3 mRNA expression in the PMv was higher in prepubertal than in adult females. In prepubertal females Slc6a3 mRNA expression was higher in overnourished females from small size litters than in controls. Prepubertal Lep ob females showed decreased PMv Slc6a3 mRNA expression, that recovered to control levels after 3 days of leptin injections. Using a tracer adenoassociated virus in the PMv of adult DATCre;Kiss1hrGFP females, we observed PMvDAT projections in the anteroventral periventricular and periventricular nucleus (AVPV/PeN), surrounding Kiss1hrGFP neurons, a population critical for sexual maturation and positive estrogen feedback in females. The DATCRE;tdTomato projections to the AVPV were denser in adult than in prepubertal females. In adults, they surrounded tyrosine hydroxylase neurons. Overall, these findings suggest that the DAT expressing PMvLepRb subpopulation play a role in leptin regulation of sexual maturation via actions on AVPV kisspeptin/tyrosine hydroxylase neurons.
Collapse
Affiliation(s)
- Cristina Sáenz de Miera
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109
| | - Nicole Bellefontaine
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109
| | - Marina A Silveira
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, 78249
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil, 05508
| | - Chelsea N Fortin
- Department of Obstetrics and Gynecology University of Michigan, Ann Arbor, MI, 48109
| | - Thais T Zampieri
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil, 05508
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil, 05508
| | - Kevin W Williams
- Center for Hypothalamic Research, Department of Internal Medicine, Peter O’Donnell Jr. Brain Institute, The University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390
| | | | - Laura Heikkinen
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Christian Broberger
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Renata Frazao
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil, 05508
| | - Carol F Elias
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109
- Department of Obstetrics and Gynecology University of Michigan, Ann Arbor, MI, 48109
| |
Collapse
|
5
|
Drumond-Bock AL, Beckstead MJ. Ketones on the Brain: Can Diet Help Turn the Tables on Parkinson's?-Commentary on Mahajan et al. (2024). Eur J Neurosci 2025; 61:e70025. [PMID: 39937087 DOI: 10.1111/ejn.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/24/2025] [Accepted: 02/01/2025] [Indexed: 02/13/2025]
Affiliation(s)
- Ana Luiza Drumond-Bock
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Michael J Beckstead
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
- Oklahoma City VA Medical Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
6
|
Xiao X, Yeghiazaryan G, Eggersmann F, Cremer AL, Backes H, Kloppenburg P, Hausen AC. Deficiency of orexin receptor type 1 in dopaminergic neurons increases novelty-induced locomotion and exploration. eLife 2025; 12:RP91716. [PMID: 39841059 PMCID: PMC11753781 DOI: 10.7554/elife.91716] [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: 01/23/2025] Open
Abstract
Orexin signaling in the ventral tegmental area and substantia nigra promotes locomotion and reward processing, but it is not clear whether dopaminergic neurons directly mediate these effects. We show that dopaminergic neurons in these areas mainly express orexin receptor subtype 1 (Ox1R). In contrast, only a minor population in the medial ventral tegmental area express orexin receptor subtype 2 (Ox2R). To analyze the functional role of Ox1R signaling in dopaminergic neurons, we deleted Ox1R specifically in dopamine transporter-expressing neurons of mice and investigated the functional consequences. Deletion of Ox1R increased locomotor activity and exploration during exposure to novel environments or when intracerebroventricularely injected with orexin A. Spontaneous activity in home cages, anxiety, reward processing, and energy metabolism did not change. Positron emission tomography imaging revealed that Ox1R signaling in dopaminergic neurons affected distinct neural circuits depending on the stimulation mode. In line with an increase of neural activity in the lateral paragigantocellular nucleus (LPGi) of Ox1RΔDAT mice, we found that dopaminergic projections innervate the LPGi in regions where the inhibitory dopamine receptor subtype D2 but not the excitatory D1 subtype resides. These data suggest a crucial regulatory role of Ox1R signaling in dopaminergic neurons in novelty-induced locomotion and exploration.
Collapse
Affiliation(s)
- Xing Xiao
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of MetabolismCologneGermany
| | - Gagik Yeghiazaryan
- Department of Biology, Institute for Zoology, University of CologneCologneGermany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of CologneCologneGermany
| | - Fynn Eggersmann
- Department of Biology, Institute for Zoology, University of CologneCologneGermany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of CologneCologneGermany
| | - Anna Lena Cremer
- Max Planck Institute for Metabolism Research, Multimodal Imaging of Brain Metabolism GroupCologneGermany
| | - Heiko Backes
- Max Planck Institute for Metabolism Research, Multimodal Imaging of Brain Metabolism GroupCologneGermany
| | - Peter Kloppenburg
- Department of Biology, Institute for Zoology, University of CologneCologneGermany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of CologneCologneGermany
| | - Anne Christine Hausen
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of MetabolismCologneGermany
| |
Collapse
|
7
|
Berezhnoi D, Chehade HD, Simms G, Chen L, Chu HY. Sub-second characterization of locomotor activities of mouse models of Parkinsonism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.26.630411. [PMID: 39763733 PMCID: PMC11703164 DOI: 10.1101/2024.12.26.630411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
The degeneration of midbrain dopamine (DA) neurons disrupts the neural control of natural behavior, such as walking, posture, and gait in Parkinson's disease. While some aspects of motor symptoms can be managed by dopamine replacement therapies, others respond poorly. Recent advancements in machine learning-based technologies offer opportunities for unbiased segmentation and quantification of natural behavior in both healthy and diseased states. In the present study, we applied the motion sequencing (MoSeq) platform to study the spontaneous locomotor activities of neurotoxin and genetic mouse models of Parkinsonism as the midbrain DA neurons progressively degenerate. We also evaluated the treatment efficacy of levodopa (L-DOPA) on behavioral modules at fine time scales. We revealed robust changes in the kinematics and usage of the behavioral modules that encode spontaneous locomotor activity. Further analysis demonstrates that fast behavioral modules with higher velocities were more vulnerable to loss of DA and preferentially affected at early stages of Parkinsonism. Last, L-DOPA effectively improved the velocity, but not the usage and transition probability, of behavioral modules of Parkinsonian animals. In conclusion, the hypokinetic phenotypes in Parkinsonism are mediated by the decreased velocities of behavioral modules and the disrupted temporal organization of sub-second modules into actions. Moreover, we showed that the therapeutic effect of L-DOPA is mainly mediated by its effect on the velocities of behavior modules at fine time scales. This work documents robust changes in the velocity, usage, and temporal organization of behavioral modules and their responsiveness to dopaminergic treatment under the Parkinsonian state.
Collapse
Affiliation(s)
- Daniil Berezhnoi
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington DC, 20007, United States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, United States
| | - Hiba Douja Chehade
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington DC, 20007, United States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, United States
| | - Gabriel Simms
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States
| | - Liqiang Chen
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington DC, 20007, United States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, United States
| | - Hong-Yuan Chu
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington DC, 20007, United States
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, United States
| |
Collapse
|
8
|
Carbonell-Roig J, Aaltonen A, Wilson K, Molinari M, Cartocci V, McGuirt A, Mosharov E, Kehr J, Lieberman OJ, Sulzer D, Borgkvist A, Santini E. Dysregulated acetylcholine-mediated dopamine neurotransmission in the eIF4E Tg mouse model of autism spectrum disorders. Cell Rep 2024; 43:114997. [PMID: 39607825 DOI: 10.1016/j.celrep.2024.114997] [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: 02/05/2024] [Revised: 09/18/2024] [Accepted: 11/05/2024] [Indexed: 11/30/2024] Open
Abstract
Autism spectrum disorder (ASD) consists of diverse neurodevelopmental conditions where core behavioral symptoms are critical for diagnosis. Altered dopamine (DA) neurotransmission in the striatum has been suggested to contribute to the behavioral features of ASD. Here, we examine DA neurotransmission in a mouse model of ASD characterized by elevated expression of eukaryotic initiation factor 4E (eIF4E), a key regulator of cap-dependent translation, using a comprehensive approach that encompasses genetics, behavior, synaptic physiology, and imaging. The results indicate that increased eIF4E expression leads to behavioral inflexibility and impaired striatal DA release. The loss of normal DA neurotransmission is due to a defect in nicotinic receptor signaling that regulates calcium dynamics in dopaminergic axons. These findings provide a mechanistic understanding of ASD symptoms and offer a foundation for targeted therapeutic interventions by revealing the intricate interplay between eIF4E, DA neurotransmission, and behavioral flexibility.
Collapse
Affiliation(s)
| | - Alina Aaltonen
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
| | - Karin Wilson
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
| | - Maya Molinari
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
| | - Veronica Cartocci
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden
| | - Avery McGuirt
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA
| | - Eugene Mosharov
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA
| | - Jan Kehr
- Pronexus Analytical AB, 16733 Stockholm-Bromma, Sweden
| | - Ori J Lieberman
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA; Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA 94143, USA
| | - David Sulzer
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA; New York State Psychiatric Institute, New York, NY 10032, USA
| | - Anders Borgkvist
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden.
| | - Emanuela Santini
- Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden.
| |
Collapse
|
9
|
Fushiki A, Ng D, Lewis ZR, Yadav A, Saraiva T, Hammond LA, Wirblich C, Tasic B, Menon V, da Silva JA, Costa RM. A Vulnerable Subtype of Dopaminergic Neurons Drives Early Motor Deficits in Parkinson's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.20.629776. [PMID: 39763754 PMCID: PMC11702755 DOI: 10.1101/2024.12.20.629776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
In Parkinson's disease, dopaminergic neurons (DANs) in the midbrain gradually degenerate, with ventral substantia nigra pars compacta (SNc) DANs exhibiting greater vulnerability. However, it remains unclear whether specific molecular subtypes of ventral SNc DANs are more susceptible to degeneration in PD, and if they contribute to the early motor symptoms associated with the disease. We identified a subtype of Sox6+ DANs, Anxa1+, which are selectively lost earlier than other DANs, and with a time course that aligns with the development of motor symptoms in MitoPark mice. We generated a knock-in Cre mouse line for Anxa1+ DANs and showed differential anatomical inputs and outputs of this population. Further, we found that the inhibition of transmitter release in Anxa1+ neurons led to bradykinesia and tremor. This study uncovers the existence of a selectively vulnerable subtype of DANs that is sufficient to drive early motor symptoms in Parkinson's disease.
Collapse
Affiliation(s)
- Akira Fushiki
- Allen Institute, Seattle, WA 98109, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| | - David Ng
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | | | - Archana Yadav
- Center for Translational and Computational Neuroimmunology, Department of Neurology Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Tatiana Saraiva
- Champalimaud Research, Champalimaud Foundation, Lisbon 1400-038, Portugal
- Department of Neurology, University Hospital of Würzburg, Würzburg 97080, Germany
| | - Luke A. Hammond
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
- Department of Neurology, The Ohio State University, Columbus, OH 43210, USA
| | - Christoph Wirblich
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | - Vilas Menon
- Center for Translational and Computational Neuroimmunology, Department of Neurology Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Joaquim Alves da Silva
- Champalimaud Research, Champalimaud Foundation, Lisbon 1400-038, Portugal
- NOVA Medical School, Universidade Nova de Lisboa, Lisbon 1169-056, Portugal
| | - Rui M. Costa
- Allen Institute, Seattle, WA 98109, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815
| |
Collapse
|
10
|
Mahajan VR, Nadel JA, King MT, Pawlosky RJ, Davis MI, Veech RL, Lovinger DM, Salinas AG. Ketone ester-enriched diet ameliorates motor and dopamine release deficits in MitoPark mice. Eur J Neurosci 2024; 60:6875-6890. [PMID: 39528410 PMCID: PMC11612846 DOI: 10.1111/ejn.16601] [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: 11/09/2023] [Revised: 10/07/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Parkinson's disease (PD) is a progressive, neurodegenerative disease characterized by motor dysfunction and dopamine deficits. The MitoPark (MP) mouse model of PD recapitulates several facets of Parkinson's disease, including gradual development of motor deficits, which enables the study of potential therapeutic interventions. One therapeutic strategy involves decreasing the mitochondrial metabolic load by inducing ketosis and providing an alternative energy source for neurons, leading to decreased neuronal oxidative stress. Thus, we hypothesized that administration of a ketone ester-enriched diet (KEED) would improve motor and dopamine release deficits in MP mice. Motor function (rotarod and open field tests), dopamine release (fast-scan cyclic voltammetry), tissue dopamine levels (gas chromatography-mass spectrometry) and dopamine neurons and axons (immunofluorescence) were assessed in MP, and control mice fed either the standard or a KEED. When started on the ketone diet before motor dysfunction onset, MP mice had improved motor function relative to standard diet (SD) MP mice. While the KEED did not preserve dopamine neurons or striatal dopamine axons, dopamine release in ketone diet MP mice was greater than SD MP mice but less than control mice. In a follow-up experiment, we began the ketone diet after motor dysfunction onset and observed a modest preservation of motor function in ketone diet MP mice relative to SD MP mice. The improvement in motor dysfunction indicates that a KEED or ketone supplement may have a beneficial effect on delaying motor deficit progression in Parkinson's disease.
Collapse
Affiliation(s)
- Vikrant R. Mahajan
- Laboratory for Integrative NeuroscienceNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthRockvilleMarylandUSA
- Laboratory for Metabolic ControlNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthRockvilleMarylandUSA
| | - Jacob A. Nadel
- Laboratory for Integrative NeuroscienceNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthRockvilleMarylandUSA
| | - M. Todd King
- Laboratory for Metabolic ControlNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthRockvilleMarylandUSA
| | - Robert J. Pawlosky
- Laboratory for Metabolic ControlNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthRockvilleMarylandUSA
| | - Margaret I. Davis
- Laboratory for Integrative NeuroscienceNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthRockvilleMarylandUSA
| | - Richard L. Veech
- Laboratory for Metabolic ControlNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthRockvilleMarylandUSA
| | - David M. Lovinger
- Laboratory for Integrative NeuroscienceNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthRockvilleMarylandUSA
| | - Armando G. Salinas
- Laboratory for Integrative NeuroscienceNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthRockvilleMarylandUSA
- Department of Pharmacology, Toxicology & NeuroscienceLouisiana State University Health Sciences Center – ShreveportShreveportLouisianaUSA
| |
Collapse
|
11
|
Hattori N, Sato S. Mitochondrial dysfunction in Parkinson's disease. J Neural Transm (Vienna) 2024; 131:1415-1428. [PMID: 39585446 DOI: 10.1007/s00702-024-02863-2] [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: 09/18/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024]
Abstract
The exact cause of nigral cell death in Parkinson's disease (PD) is still unknown. However, research on MPTP-induced experimental parkinsonism has significantly advanced our understanding. In this model, it is widely accepted that mitochondrial respiratory failure is the primary mechanism of cell death. Studies have shown that a toxic metabolite of MPTP inhibits Complex I and alpha-ketoglutarate dehydrogenase activities in mitochondria. Since then, many research groups have focused on mitochondrial dysfunction in PD, identifying deficiencies in Complex I or III in PD patients' brains, skeletal muscle, and platelets. There is some debate about the decline in mitochondrial function in peripheral organs. However, since α-synuclein, the main component protein of Lewy bodies, accumulates in peripheral organs, it is reasonable to consider PD a systemic disease. Additionally, mutant mitochondrial DNA with a 4,977 base pair deletion has been found in the brains of PD patients, suggesting that age-related accumulation of deleted mtDNA is accelerated in the striatum and may contribute to the pathophysiology of PD. While the cause of PD remains unknown, mitochondrial dysfunction is undoubtedly a factor in cell death in PD. In addition, the causative gene for familial PD, parkin (now PRKN), and PTEN-induced putative kinase 1 (PINK1), both gene products are also involved in mitochondrial quality control. Moreover, we have successfully isolated and identified CHCHD2, which is involved in the mitochondrial electron transfer system. There is no doubt that mitochondrial dysfunction contributes to cell death in PD.
Collapse
Affiliation(s)
- Nobutaka Hattori
- Department of Neurology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan.
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, 2-1-Hirosawa, Wako-Shi, Saitama, 351-0198, Japan.
| | - Shigeto Sato
- Department of Neurology, Faculty of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
- Center for Biomedical Research Resources, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan
| |
Collapse
|
12
|
Nielsen BE, Ford CP. Reduced striatal M4-cholinergic signaling following dopamine loss contributes to parkinsonian and l-DOPA-induced dyskinetic behaviors. SCIENCE ADVANCES 2024; 10:eadp6301. [PMID: 39565858 PMCID: PMC11578179 DOI: 10.1126/sciadv.adp6301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 10/17/2024] [Indexed: 11/22/2024]
Abstract
A dynamic equilibrium between dopamine and acetylcholine (ACh) is essential for striatal circuitry and motor function, as imbalances are associated with Parkinson's disease (PD) and levodopa-induced dyskinesia (LID). Conventional theories posit that cholinergic signaling is pathologically elevated in PD as a result of increased ACh release, which contributes to motor deficits. However, using approaches to measure receptor-mediated signaling, we found that, rather than the predicted enhancement, the strength of cholinergic transmission at muscarinic M4 receptor synapses on direct pathway medium spiny neurons was decreased in dopamine-depleted mice. This adaptation was due to a reduced postsynaptic M4 receptor function, resulting from down-regulated receptors and downstream signaling. Restoring M4 transmission unexpectedly led to a partial alleviation of motor deficits and LID dyskinetic behavior, revealing an unexpected prokinetic effect in addition to the canonical antikinetic role of M4 receptors. These findings indicate that decreased M4 function differentially contributes to parkinsonian and LID pathophysiology, representing a promising target for therapeutic intervention.
Collapse
Affiliation(s)
- Beatriz E. Nielsen
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Christopher P. Ford
- Department of Pharmacology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| |
Collapse
|
13
|
Charli A, Luo J, Palanisamy B, Malovic E, Riaz Z, Miller C, Chang YT, Samidurai M, Zenitsky G, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Mitochondrial Stress Disassembles Nuclear Architecture through Proteolytic Activation of PKCδ and Lamin B1 Phosphorylation in Neuronal Cells: Implications for Pathogenesis of Age-related Neurodegenerative Diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.01.621517. [PMID: 39554109 PMCID: PMC11565982 DOI: 10.1101/2024.11.01.621517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Mitochondrial dysfunction and oxidative stress are hallmarks of pathophysiological processes in age-related neurodegenerative diseases including Parkinson's, Alzheimer's and Huntington's diseases. Neuronal cells are highly vulnerable to mitochondrial stress, however, the cellular and molecular mechanisms underlying the enhanced vulnerability are not well understood. Previously, we demonstrated that the novel PKC isoform PKCδ is highly expressed in dopamin(DA)ergic neurons and plays a key role in inducing apoptotic cell death during neurotoxic stress via caspase-3-mediated proteolytic activation. Herein, we further uncovered a key downstream molecular event of PKCδ signaling following mitochondrial dysfunction that governs neuronal cell death by dissembling nuclear architecture. Exposing N27 DAergic cell line to the mitochondrial complex-1 inhibitor tebufenpyrad induced PKCδ phosphorylation at the T505 activation loop accompanied by caspase-3-dependent proteolytic activation of the kinase. Subcellular analysis using high-resolution 3D confocal microscopy revealed that proteolytically activated cleaved PKCδ translocates to the nuclear compartment, colocalizing with Lamin B1. Electron microscopy also enabled the visualization of nuclear membrane damage triggered by subjecting the DAergic neuronal cells by Tebufenpyrad (Tebu) toxicity. In silico analyses identified that the threonine site on Lamin B1 (T575) is likely phosphorylated by PKCδ, suggesting that Lamin B1 serves as a key downstream target of the kinase. Interestingly, N27 DAergic cells stably expressing the PKCδ proteolytic cleavage site-resistant mutant failed to induce nuclear damage, PKCδ activation, and Lamin B1 phosphorylation. Furthermore, CRISPR/Cas9-based stable knockdown of PKCδ greatly attenuated Tebu-induced Lamin B1 phosphorylation. Also, studies using Lamin B1 T575G mutated at phosphorylation and PKCδ-ΔNLS-overexpressing N27 cells showed that PKCδ activation and translocation to the nuclear membrane are critically required for phosphorylating Lamin B1 at T575 to induce nuclear membrane damage during Tebu insult. Additionally, Tebu failed to induce Lamin B1 damage and Lamin B1 phosphorylation in organotypic midbrain slices cultured from PKCδ -/- mouse pups. More importantly, we observed higher PKCδ activation, Lamin B1 phosphorylation and Lamin B1 loss in nigral DAergic neurons from the postmortem brains of PD patients as compared to age-matched healthy control brains, thus providing translational relevance of our finding. Collectively, our data reveal that PKCδ functions as a Lamin B1 kinase to disassemble the nuclear membrane during the neuronal cell death process triggered by mitochondrial stress. This mechanistic insight may have important implications for the etiology of age-related neurodegenerative diseases resulting from mitochondrial dysfunction as well as for the development of novel treatment strategies.
Collapse
|
14
|
Shao M, Zhao C, Pan Z, Yang X, Gao C, Kam GHC, Zhou H, Lee SMY. Oxyphylla A exerts antiparkinsonian effects by ameliorating 6-OHDA-induced mitochondrial dysfunction and dyskinesia in vitro and in vivo. Chem Biol Interact 2024; 403:111224. [PMID: 39233265 DOI: 10.1016/j.cbi.2024.111224] [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: 05/14/2024] [Revised: 08/15/2024] [Accepted: 09/02/2024] [Indexed: 09/06/2024]
Abstract
Parkinson's disease (PD) poses a formidable challenge in neurology, marked by progressive neuronal loss in the substantia nigra. Despite extensive investigations, understanding PD's pathophysiology remains elusive, with no effective therapeutic intervention identified to alter its course. Oxyphylla A (OPA), a natural compound extracted from Alpinia oxyphylla, exhibits promise in experimental models of various neurodegenerative disorders (ND), notably through novel mechanisms like α-synuclein degradation. The purpose of this investigation was to explore the neuroprotective potential of OPA on 6-hydroxydopamine (6-OHDA)-induced neurotoxicity in PD models, with a focus on mitochondrial functions. Additionally, potential OPA targets for neuroprotection were explored. PC12 cells and C57BL/6 mice were lesioned with 6-OHDA as PD models. Impaired mitochondrial membrane potential (Δψm) was assessed using JC-1 staining. The oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were also detected to evaluate mitochondrial function and glucose metabolism in PC12 cells. Behavioral analysis and immunohistochemistry were performed to evaluate pathological lesions in the mouse brain. Moreover, bioinformatics tools predicted OPA targets. OPA restored cellular energy metabolism and mitochondrial biogenesis, preserving Δψm in 6-OHDA-induced neuronal damage. Pre-treatment mitigated loss of tyrosine hydroxylase (TH)-positive neurons in the substantia nigra and striatal dopaminergic fibers, restoring dopamine levels and ameliorating motor deficits in PD mice. Mechanistically, OPA may activate PKA/Akt/GSK-3β and CREB/PGC-1α/NRF-1/TFAM signaling cascades. Bioinformatics analysis identified potential OPA targets, including CTNNB1, ESR1, MAPK1, MAPK14, and SRC. OPA, derived from Alpinia oxyphylla, exhibited promising neuroprotective activity against PD through addressing mitochondrial dysfunction, suggesting its potential as a multi-targeted therapeutic for PD.
Collapse
Affiliation(s)
- Min Shao
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Chen Zhao
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Zhijian Pan
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Xuanjun Yang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China; Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Cheng Gao
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Gloria Hio-Cheng Kam
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Hefeng Zhou
- School of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, China.
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macau, China; Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China; Research Centre for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China; State Key Laboratory of Chemical Biology and Drug Discovery, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China; Research Institute for Smart Ageing, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China; Research Institute for Future Food, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China; PolyU-BGI Joint Research Centre for Genomics and Synthetic Biology in Global Deep Ocean Resource, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China.
| |
Collapse
|
15
|
Zhu Y, Kohli N, Young A, Sheldon M, Coni J, Rajasekaran M, Robinson L, Chroneos R, Riley S, Guarnieri JW, Jose J, Patel N, Wallace DC, Li S, Lee H, Mach RH, McManus MJ. PET Imaging with [ 18F]ROStrace Detects Oxidative Stress and Predicts Parkinson's Disease Progression in Mice. Antioxidants (Basel) 2024; 13:1226. [PMID: 39456479 PMCID: PMC11504722 DOI: 10.3390/antiox13101226] [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: 08/20/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Although the precise molecular mechanisms responsible for neuronal death and motor dysfunction in late-onset Parkinson's disease (PD) are unknown, evidence suggests that mitochondrial dysfunction and neuroinflammation occur early, leading to a collective increase in reactive oxygen species (ROS) production and oxidative stress. However, the lack of methods for tracking oxidative stress in the living brain has precluded its use as a potential biomarker. The goal of the current study is to address this need through the evaluation of the first superoxide (O2•-)-sensitive radioactive tracer, [18F]ROStrace, in a model of late-onset PD. To achieve this goal, MitoPark mice with a dopaminergic (DA) neuron-specific deletion of transcription factor A mitochondrial (Tfam) were imaged with [18F]ROStrace from the prodromal phase to the end-stage of PD-like disease. Our data demonstrate [18F]ROStrace was sensitive to increased oxidative stress during the early stages of PD-like pathology in MitoPark mice, which persisted throughout the disease course. Similarly to PD patients, MitoPark males had the most severe parkinsonian symptoms and metabolic impairment. [18F]ROStrace retention was also highest in MitoPark males, suggesting oxidative stress as a potential mechanism underlying the male sex bias of PD. Furthermore, [18F]ROStrace may provide a method to identify patients at risk of Parkinson's before irreparable neurodegeneration occurs and enhance clinical trial design by identifying patients most likely to benefit from antioxidant therapies.
Collapse
Affiliation(s)
- Yi Zhu
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Neha Kohli
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Anthony Young
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Malkah Sheldon
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jani Coni
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Meera Rajasekaran
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lozen Robinson
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Rea Chroneos
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Shaipreeah Riley
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Joseph W. Guarnieri
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Joshua Jose
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nisha Patel
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Douglas C. Wallace
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Shihong Li
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hsiaoju Lee
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert H. Mach
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Meagan J. McManus
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Center for Mitochondrial and Epigenomic Medicine, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
16
|
Lal R, Singh A, Watts S, Chopra K. Experimental models of Parkinson's disease: Challenges and Opportunities. Eur J Pharmacol 2024; 980:176819. [PMID: 39029778 DOI: 10.1016/j.ejphar.2024.176819] [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: 09/07/2023] [Revised: 05/29/2024] [Accepted: 07/17/2024] [Indexed: 07/21/2024]
Abstract
Parkinson's disease (PD) is a widespread neurodegenerative disorder occurs due to the degradation of dopaminergic neurons present in the substantia nigra pars compacta (SNpc). Millions of people are affected by this devastating disorder globally, and the frequency of the condition increases with the increase in the elderly population. A significant amount of progress has been made in acquiring more knowledge about the etiology and the pathogenesis of PD over the past decades. Animal models have been regarded to be a vital tool for the exploration of complex molecular mechanisms involved in PD. Various animals used as models for disease monitoring include vertebrates (zebrafish, rats, mice, guinea pigs, rabbits and monkeys) and invertebrate models (Drosophila, Caenorhabditis elegans). The animal models most relevant for study of PD are neurotoxin induction-based models (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), 6-Hydroxydopamine (6-OHDA) and agricultural pesticides (rotenone, paraquat), pharmacological models (reserpine or haloperidol treated rats), genetic models (α-synuclein, Leucine-rich repeat kinase 2 (LRRK2), DJ-1, PINK-1 and Parkin). Several non-mammalian genetic models such as zebrafish, Drosophila and Caenorhabditis elegance have also gained popularity in recent years due to easy genetic manipulation, presence of genes homologous to human PD, and rapid screening of novel therapeutic molecules. In addition, in vitro models (SH-SY5Y, PC12, Lund human mesencephalic (LUHMES) cells, Human induced pluripotent stem cell (iPSC), Neural organoids, organ-on-chip) are also currently in trend providing edge in investigating molecular mechanisms involved in PD as they are derived from PD patients. In this review, we explain the current situation and merits and demerits of the various animal models.
Collapse
Affiliation(s)
- Roshan Lal
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India.
| | - Aditi Singh
- TR(i)P for Health Laboratory, Centre for Excellence in Functional Foods, Department of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute (NABI), Knowledge City, Sector 81, SAS Nagar, Punjab, 140306, India.
| | - Shivam Watts
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India.
| | - Kanwaljit Chopra
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
17
|
Paß T, Ricke KM, Hofmann P, Chowdhury RS, Nie Y, Chinnery P, Endepols H, Neumaier B, Carvalho A, Rigoux L, Steculorum SM, Prudent J, Riemer T, Aswendt M, Liss B, Brachvogel B, Wiesner RJ. Preserved striatal innervation maintains motor function despite severe loss of nigral dopaminergic neurons. Brain 2024; 147:3189-3203. [PMID: 38574200 DOI: 10.1093/brain/awae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 01/22/2024] [Accepted: 02/09/2024] [Indexed: 04/06/2024] Open
Abstract
Degeneration of dopaminergic neurons in the substantia nigra and their striatal axon terminals causes cardinal motor symptoms of Parkinson's disease. In idiopathic cases, high levels of mitochondrial DNA alterations, leading to mitochondrial dysfunction, are a central feature of these vulnerable neurons. Here we present a mouse model expressing the K320E variant of the mitochondrial helicase Twinkle in dopaminergic neurons, leading to accelerated mitochondrial DNA mutations. These K320E-TwinkleDaN mice showed normal motor function at 20 months of age, although ∼70% of nigral dopaminergic neurons had perished. Remaining neurons still preserved ∼75% of axon terminals in the dorsal striatum and enabled normal dopamine release. Transcriptome analysis and viral tracing confirmed compensatory axonal sprouting of the surviving neurons. We conclude that a small population of substantia nigra dopaminergic neurons is able to adapt to the accumulation of mitochondrial DNA mutations and maintain motor control.
Collapse
Affiliation(s)
- Thomas Paß
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Konrad M Ricke
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Pierre Hofmann
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Roy S Chowdhury
- MRC Mitochondrial Biology Unit, University of Cambridge, CB2 0XY Cambridge, UK
| | - Yu Nie
- MRC Mitochondrial Biology Unit, University of Cambridge, CB2 0XY Cambridge, UK
| | - Patrick Chinnery
- MRC Mitochondrial Biology Unit, University of Cambridge, CB2 0XY Cambridge, UK
| | - Heike Endepols
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, 50937 Cologne, Germany
- Department of Nuclear Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
| | - Bernd Neumaier
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, 50937 Cologne, Germany
- Forschungszentrum Jülich GmbH, Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), 52425 Jülich, Germany
- Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
| | - André Carvalho
- Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD) and Centre for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Lionel Rigoux
- Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
| | - Sophie M Steculorum
- Max Planck Institute for Metabolism Research, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD) and Centre for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| | - Julien Prudent
- MRC Mitochondrial Biology Unit, University of Cambridge, CB2 0XY Cambridge, UK
| | - Trine Riemer
- Department of Paediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Markus Aswendt
- Department of Neurology, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
| | - Birgit Liss
- Institute of Applied Physiology, University of Ulm, 89081 Ulm, Germany
| | - Bent Brachvogel
- Department of Paediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, 50937 Cologne, Germany
| | - Rudolf J Wiesner
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD) and Centre for Molecular Medicine (CMMC), University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
18
|
He S, Ru Q, Chen L, Xu G, Wu Y. Advances in animal models of Parkinson's disease. Brain Res Bull 2024; 215:111024. [PMID: 38969066 DOI: 10.1016/j.brainresbull.2024.111024] [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: 05/12/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024]
Abstract
Parkinson's disease is a complex neurodegenerative disease characterized by progressive movement impairments. Predominant symptoms encompass resting tremor, bradykinesia, limb rigidity, and postural instability. In addition, it also includes a series of non-motor symptoms such as sleep disorders, hyposmia, gastrointestinal dysfunction, autonomic dysfunction and cognitive impairment. Pathologically, the disease manifests through dopaminergic neuronal loss and the presence of Lewy bodies. At present, no significant breakthrough has been achieved in clinical Parkinson's disease treatment. Exploring treatment modalities necessitate the establishment of scientifically sound animal models. In recent years, researchers have focused on replicating the symptoms of human Parkinson's disease, resulting in the establishment of various experimental animal models primarily through drugs and transgenic methods to mimic relevant pathologies and identify more effective treatments. This review examines traditional neurotoxin and transgenic animal models as well as α-synuclein pre-formed fibrils models, non-human primate models and non-mammalian specie models. Additionally, it introduces emerging models, including models based on optogenetics, induced pluripotent stem cells, and gene editing, aiming to provide a reference for the utilization of experimental animal models and clinical research for researchers in this field.
Collapse
Affiliation(s)
- Sui He
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Guodong Xu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
19
|
Medeiros D, Masini D, Plewnia C, Boi L, Rosati M, Scalbert N, Fisone G. Dopamine D2 receptor activation counteracts olfactory dysfunction and related cellular abnormalities in experimental parkinsonism. Heliyon 2024; 10:e35948. [PMID: 39224310 PMCID: PMC11366923 DOI: 10.1016/j.heliyon.2024.e35948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Olfactory dysfunction is a common non-motor symptom associated with Parkinson's disease (PD). This condition usually appears before the onset of the cardinal motor symptoms and is still poorly understood. Here, we generated a mouse model of early-stage PD based on partial 6-hydroxydopamine (6-OHDA) lesion of the dorsal striatum to reproduce the olfactory deficit and associated cellular and electrophysiological anomalies observed in patients. Using this model, we investigated the effect of long-term, continuous administration of pramipexole, a dopamine D2/3 selective agonist, on olfactory dysfunction. We found that pramipexole reverted the impairment of odor discrimination displayed by the mouse model in the habituation/dishabituation test. In line with similar observations in PD patients, the mouse model showed an increase of dopamine cells paralleled by augmented levels of the dopamine marker, tyrosine hydroxylase, in the olfactory bulb (OB). These changes, which have been proposed to contribute to olfactory dysfunction, were abolished by oral administration of pramipexole. Local field potential recording in the OB of 6-OHDA lesion mice showed reduced oscillations in the beta frequency range, in comparison to healthy control mice. This abnormality, which is suggestive of defective long range OB transmission, was also counteracted by pramipexole. Altogether these findings indicate that prolonged pharmacological stimulation of dopamine D2-like receptors rescues olfactory discrimination observed in experimental parkinsonism. Moreover, they show that this protective effect is exerted in parallel to a normalization of dopamine neurons and beta band oscillations in the OB, providing information on the potential mechanisms involved in PD-related olfactory dysfunction.
Collapse
Affiliation(s)
| | | | - Carina Plewnia
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Laura Boi
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Martha Rosati
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Nicolas Scalbert
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
20
|
Huang M, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. Mitochondrial stress-induced H4K12 hyperacetylation dysregulates transcription in Parkinson's disease. Front Cell Neurosci 2024; 18:1422362. [PMID: 39188570 PMCID: PMC11345260 DOI: 10.3389/fncel.2024.1422362] [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: 04/23/2024] [Accepted: 07/08/2024] [Indexed: 08/28/2024] Open
Abstract
Aberrant epigenetic modification has been implicated in the pathogenesis of Parkinson's disease (PD), which is characterized by the irreversible loss of dopaminergic (DAergic) neurons. However, the mechanistic landscape of histone acetylation (ac) in PD has yet to be fully explored. Herein, we mapped the proteomic acetylation profiling changes at core histones H4 and thus identified H4K12ac as a key epigenomic mark in dopaminergic neuronal cells as well as in MitoPark animal model of PD. Notably, the significantly elevated H4K12ac deposition in post-mortem PD brains highlights its clinical relevance to human PD. Increased histone acetyltransferase (HAT) activity and decreased histone deacetylase 2 (HDAC2) and HDAC4 were found in experimental PD cell models, suggesting the HAT/HDAC imbalance associated with mitochondrial stress. Following our delineation of the proteasome dysfunction that possibly contributes to H4K12ac deposition, we characterized the altered transcriptional profile and disease-associated pathways in the MitoPark mouse model of PD. Our study uncovers the axis of mitochondrial impairment-H4K12ac deposition-altered transcription/disease pathways as a neuroepigenetic mechanism underlying PD pathogenesis. These findings provide mechanistic information for the development of potential pharmacoepigenomic translational strategies for PD.
Collapse
Affiliation(s)
- Minhong Huang
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
| | - Huajun Jin
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States
| | - Vellareddy Anantharam
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States
| | - Arthi Kanthasamy
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States
| | - Anumantha G. Kanthasamy
- Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, United States
| |
Collapse
|
21
|
Recinto SJ, Premachandran S, Mukherjee S, Allot A, MacDonald A, Yaqubi M, Gruenheid S, Trudeau LE, Stratton JA. Characterizing enteric neurons in dopamine transporter (DAT)-Cre reporter mice reveals dopaminergic subtypes with dual-transmitter content. Eur J Neurosci 2024; 59:2465-2482. [PMID: 38487941 DOI: 10.1111/ejn.16307] [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: 07/31/2023] [Revised: 02/08/2024] [Accepted: 02/17/2024] [Indexed: 05/22/2024]
Abstract
The enteric nervous system (ENS) comprises a complex network of neurons whereby a subset appears to be dopaminergic although the characteristics, roles, and implications in disease are less understood. Most investigations relating to enteric dopamine (DA) neurons rely on immunoreactivity to tyrosine hydroxylase (TH)-the rate-limiting enzyme in the production of DA. However, TH immunoreactivity is likely to provide an incomplete picture. This study herein provides a comprehensive characterization of DA neurons in the gut using a reporter mouse line, expressing a fluorescent protein (tdTomato) under control of the DA transporter (DAT) promoter. Our findings confirm a unique localization of DA neurons in the gut and unveil the discrete subtypes of DA neurons in this organ, which we characterized using both immunofluorescence and single-cell transcriptomics, as well as validated using in situ hybridization. We observed distinct subtypes of DAT-tdTomato neurons expressing co-transmitters and modulators across both plexuses; some of them likely co-releasing acetylcholine, while others were positive for a slew of canonical DAergic markers (TH, VMAT2 and GIRK2). Interestingly, we uncovered a seemingly novel population of DA neurons unique to the ENS which was ChAT/DAT-tdTomato-immunoreactive and expressed Grp, Calcb, and Sst. Given the clear heterogeneity of DAergic gut neurons, further investigation is warranted to define their functional signatures and decipher their implication in disease.
Collapse
Affiliation(s)
- Sherilyn Junelle Recinto
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Shobina Premachandran
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Sriparna Mukherjee
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Pharmacology and Physiology, Department of Neurosciences, Université de Montreal, Faculty of Medicine, SNC and CIRCA Research Groups, Montreal, Quebec, Canada
| | - Alexis Allot
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
| | - Adam MacDonald
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Moein Yaqubi
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Samantha Gruenheid
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Louis-Eric Trudeau
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Pharmacology and Physiology, Department of Neurosciences, Université de Montreal, Faculty of Medicine, SNC and CIRCA Research Groups, Montreal, Quebec, Canada
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| |
Collapse
|
22
|
Radlicka-Borysewska A, Jabłońska J, Lenarczyk M, Szumiec Ł, Harda Z, Bagińska M, Barut J, Pera J, Kreiner G, Wójcik DK, Rodriguez Parkitna J. Non-motor symptoms associated with progressive loss of dopaminergic neurons in a mouse model of Parkinson's disease. Front Neurosci 2024; 18:1375265. [PMID: 38745938 PMCID: PMC11091341 DOI: 10.3389/fnins.2024.1375265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/09/2024] [Indexed: 05/16/2024] Open
Abstract
Parkinson's disease (PD) is characterized by three main motor symptoms: bradykinesia, rigidity and tremor. PD is also associated with diverse non-motor symptoms that may develop in parallel or precede motor dysfunctions, ranging from autonomic system dysfunctions and impaired sensory perception to cognitive deficits and depression. Here, we examine the role of the progressive loss of dopaminergic transmission in behaviors related to the non-motor symptoms of PD in a mouse model of the disease (the TIF-IADATCreERT2 strain). We found that in the period from 5 to 12 weeks after the induction of a gradual loss of dopaminergic neurons, mild motor symptoms became detectable, including changes in the distance between paws while standing as well as the swing speed and step sequence. Male mutant mice showed no apparent changes in olfactory acuity, no anhedonia-like behaviors, and normal learning in an instrumental task; however, a pronounced increase in the number of operant responses performed was noted. Similarly, female mice with progressive dopaminergic neuron degeneration showed normal learning in the probabilistic reversal learning task and no loss of sweet-taste preference, but again, a robustly higher number of choices were performed in the task. In both males and females, the higher number of instrumental responses did not affect the accuracy or the fraction of rewarded responses. Taken together, these data reveal discrete, dopamine-dependent non-motor symptoms that emerge in the early stages of dopaminergic neuron degeneration.
Collapse
Affiliation(s)
- Anna Radlicka-Borysewska
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| | - Judyta Jabłońska
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| | - Michał Lenarczyk
- Faculty of Management and Social Communication, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
| | - Łukasz Szumiec
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| | - Zofia Harda
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| | - Monika Bagińska
- Department of Brain Biochemistry, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| | - Justyna Barut
- Department of Brain Biochemistry, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| | - Joanna Pera
- Department of Neurology, Jagiellonian University Medical College, Kraków, Poland
| | - Grzegorz Kreiner
- Department of Brain Biochemistry, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| | - Daniel K. Wójcik
- Faculty of Management and Social Communication, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
- Laboratory of Neuroinformatics, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Jan Rodriguez Parkitna
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
23
|
Cardanho-Ramos C, Simões RA, Wang YZ, Faria-Pereira A, Bomba-Warczak E, Craessaerts K, Spinazzi M, Savas JN, Morais VA. Local mitochondrial replication in the periphery of neurons requires the eEF1A1 protein and thetranslation of nuclear-encoded proteins. iScience 2024; 27:109136. [PMID: 38510136 PMCID: PMC10951640 DOI: 10.1016/j.isci.2024.109136] [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: 09/10/2023] [Revised: 12/19/2023] [Accepted: 02/01/2024] [Indexed: 03/22/2024] Open
Abstract
In neurons, it is commonly assumed that mitochondrial replication only occurs in the cell body, after which the mitochondria must travel to the neuron's periphery. However, while mitochondrial DNA replication has been observed to occur away from the cell body, the specific mechanisms involved remain elusive. Using EdU-labelling in mouse primary neurons, we developed a tool to determine the mitochondrial replication rate. Taking of advantage of microfluidic devices, we confirmed that mitochondrial replication also occurs locally in the periphery of neurons. To achieve this, mitochondria require de novo nuclear-encoded, but not mitochondrial-encoded protein translation. Following a proteomic screen comparing synaptic with non-synaptic mitochondria, we identified two elongation factors - eEF1A1 and TUFM - that were upregulated in synaptic mitochondria. We found that mitochondrial replication is impaired upon the downregulation of eEF1A1, and this is particularly relevant in the periphery of neurons.
Collapse
Affiliation(s)
- Carlos Cardanho-Ramos
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rúben Alves Simões
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Yi-Zhi Wang
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Andreia Faria-Pereira
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ewa Bomba-Warczak
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Katleen Craessaerts
- VIB Center for Brain and Disease Research and KU Leuven, Department of Neurosciences, Leuven, Belgium
- Dementia Research Institute, University College London, London, UK
| | - Marco Spinazzi
- VIB Center for Brain and Disease Research and KU Leuven, Department of Neurosciences, Leuven, Belgium
- Dementia Research Institute, University College London, London, UK
- Neuromuscular Reference Center, Department of Neurology, Centre Hospitalier Universitaire d'Angers, Angers, France
| | - Jeffrey N. Savas
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Vanessa A. Morais
- Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
24
|
Wang V, Tseng KY, Kuo TT, Huang EYK, Lan KL, Chen ZR, Ma KH, Greig NH, Jung J, Choi HI, Olson L, Hoffer BJ, Chen YH. Attenuating mitochondrial dysfunction and morphological disruption with PT320 delays dopamine degeneration in MitoPark mice. J Biomed Sci 2024; 31:38. [PMID: 38627765 PMCID: PMC11022395 DOI: 10.1186/s12929-024-01025-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Mitochondria are essential organelles involved in cellular energy production. Changes in mitochondrial function can lead to dysfunction and cell death in aging and age-related disorders. Recent research suggests that mitochondrial dysfunction is closely linked to neurodegenerative diseases. Glucagon-like peptide-1 receptor (GLP-1R) agonist has gained interest as a potential treatment for Parkinson's disease (PD). However, the exact mechanisms responsible for the therapeutic effects of GLP-1R-related agonists are not yet fully understood. METHODS In this study, we explores the effects of early treatment with PT320, a sustained release formulation of the GLP-1R agonist Exenatide, on mitochondrial functions and morphology in a progressive PD mouse model, the MitoPark (MP) mouse. RESULTS Our findings demonstrate that administration of a clinically translatable dose of PT320 ameliorates the reduction in tyrosine hydroxylase expression, lowers reactive oxygen species (ROS) levels, and inhibits mitochondrial cytochrome c release during nigrostriatal dopaminergic denervation in MP mice. PT320 treatment significantly preserved mitochondrial function and morphology but did not influence the reduction in mitochondria numbers during PD progression in MP mice. Genetic analysis indicated that the cytoprotective effect of PT320 is attributed to a reduction in the expression of mitochondrial fission protein 1 (Fis1) and an increase in the expression of optic atrophy type 1 (Opa1), which is known to play a role in maintaining mitochondrial homeostasis and decreasing cytochrome c release through remodeling of the cristae. CONCLUSION Our findings suggest that the early administration of PT320 shows potential as a neuroprotective treatment for PD, as it can preserve mitochondrial function. Through enhancing mitochondrial health by regulating Opa1 and Fis1, PT320 presents a new neuroprotective therapy in PD.
Collapse
Affiliation(s)
- Vicki Wang
- Doctoral Degree Program in Translational Medicine, National Defense Medical Center and Academia Sinica, Taipei, 11490, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Kuan-Yin Tseng
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, 11490, Taiwan
- National Defense Medical Center, Taipei, 11490, Taiwan
| | - Tung-Tai Kuo
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, 11490, Taiwan
- Department of Pharmacology, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Eagle Yi-Kung Huang
- Department of Pharmacology, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Kuo-Lun Lan
- Department of Pathology, Tri-Service General Hospital, Taipei, 11490, Taiwan
| | - Zi-Rong Chen
- Department of Pathology, Tri-Service General Hospital, Taipei, 11490, Taiwan
| | - Kuo-Hsing Ma
- Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, National Institutes of Health (NIH), Baltimore, MD, 21224, USA
| | - Jin Jung
- Peptron, Inc., Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Ho-Ii Choi
- Peptron, Inc., Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Lars Olson
- Department of Neuroscience, Karolinska Institute, 171 77, Stockholm, Sweden
| | - Barry J Hoffer
- Department of Neurosurgery, University Hospitals of Cleveland, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Yuan-Hao Chen
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, 11490, Taiwan.
- National Defense Medical Center, Taipei, 11490, Taiwan.
| |
Collapse
|
25
|
Yaghmaeian Salmani B, Lahti L, Gillberg L, Jacobsen JK, Mantas I, Svenningsson P, Perlmann T. Transcriptomic atlas of midbrain dopamine neurons uncovers differential vulnerability in a Parkinsonism lesion model. eLife 2024; 12:RP89482. [PMID: 38587883 PMCID: PMC11001297 DOI: 10.7554/elife.89482] [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: 04/09/2024] Open
Abstract
Midbrain dopamine (mDA) neurons comprise diverse cells with unique innervation targets and functions. This is illustrated by the selective sensitivity of mDA neurons of the substantia nigra compacta (SNc) in patients with Parkinson's disease, while those in the ventral tegmental area (VTA) are relatively spared. Here, we used single nuclei RNA sequencing (snRNA-seq) of approximately 70,000 mouse midbrain cells to build a high-resolution atlas of mouse mDA neuron diversity at the molecular level. The results showed that differences between mDA neuron groups could best be understood as a continuum without sharp differences between subtypes. Thus, we assigned mDA neurons to several 'territories' and 'neighborhoods' within a shifting gene expression landscape where boundaries are gradual rather than discrete. Based on the enriched gene expression patterns of these territories and neighborhoods, we were able to localize them in the adult mouse midbrain. Moreover, because the underlying mechanisms for the variable sensitivities of diverse mDA neurons to pathological insults are not well understood, we analyzed surviving neurons after partial 6-hydroxydopamine (6-OHDA) lesions to unravel gene expression patterns that correlate with mDA neuron vulnerability and resilience. Together, this atlas provides a basis for further studies on the neurophysiological role of mDA neurons in health and disease.
Collapse
Affiliation(s)
| | - Laura Lahti
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholmSweden
| | - Linda Gillberg
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholmSweden
| | - Jesper Kjaer Jacobsen
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholmSweden
- Department of Neurology, Karolinska University HospitalStockholmSweden
| | - Ioannis Mantas
- Department of Clinical Neuroscience, Karolinska InstitutetStockholmSweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska InstitutetStockholmSweden
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska InstitutetStockholmSweden
| |
Collapse
|
26
|
Stagkourakis S, Williams P, Spigolon G, Khanal S, Ziegler K, Heikkinen L, Fisone G, Broberger C. Maternal Aggression Driven by the Transient Mobilisation of a Dormant Hormone-Sensitive Circuit. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.02.526862. [PMID: 38585740 PMCID: PMC10996482 DOI: 10.1101/2023.02.02.526862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Aggression, a sexually dimorphic behaviour, is prevalent in males and typically absent in virgin females. Following parturition, however, the transient expression of aggression in adult female mice protects pups from predators and infanticide by male conspecifics. While maternal hormones are known to elicit nursing, their potential role in maternal aggression remains elusive. Here, we show in mice that a molecularly defined subset of ventral premammillary (PMvDAT) neurons, instrumental for intermale aggression, switch from quiescence to a hyperexcitable state during lactation. We identify that the maternal hormones prolactin and oxytocin excite these cells through actions that include T-type Ca2+ channels. Optogenetic manipulation or genetic ablation of PMvDAT neurons profoundly affects maternal aggression, while activation of these neurons impairs the expression of non-aggression-related maternal behaviours. This work identifies a monomorphic neural substrate that can incorporate hormonal cues to enable the transient expression of a dormant behavioural program in lactating females.
Collapse
Affiliation(s)
- Stefanos Stagkourakis
- Department of Neuroscience, Biomedicum B:4, Karolinska Institutet, Solnavägen 9, 171 65 Stockholm, Sweden
| | - Paul Williams
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16C, 104 05 Stockholm, Sweden
| | - Giada Spigolon
- Department of Neuroscience, Biomedicum B:4, Karolinska Institutet, Solnavägen 9, 171 65 Stockholm, Sweden
| | - Shreya Khanal
- Department of Neuroscience, Biomedicum B:4, Karolinska Institutet, Solnavägen 9, 171 65 Stockholm, Sweden
| | - Katharina Ziegler
- Department of Neuroscience, Biomedicum B:4, Karolinska Institutet, Solnavägen 9, 171 65 Stockholm, Sweden
| | - Laura Heikkinen
- Department of Neuroscience, Biomedicum B:4, Karolinska Institutet, Solnavägen 9, 171 65 Stockholm, Sweden
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16C, 104 05 Stockholm, Sweden
| | - Gilberto Fisone
- Department of Neuroscience, Biomedicum B:4, Karolinska Institutet, Solnavägen 9, 171 65 Stockholm, Sweden
| | - Christian Broberger
- Department of Neuroscience, Biomedicum B:4, Karolinska Institutet, Solnavägen 9, 171 65 Stockholm, Sweden
- Department of Biochemistry and Biophysics, Stockholm University, Svante Arrhenius väg 16C, 104 05 Stockholm, Sweden
| |
Collapse
|
27
|
Ellioff KJ, Osting SM, Lentine A, Welper AD, Burger C, Greenspan DS. Ablation of Mitochondrial RCC1-L Induces Nigral Dopaminergic Neurodegeneration and Parkinsonian-like Motor Symptoms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.01.567409. [PMID: 38585782 PMCID: PMC10996473 DOI: 10.1101/2023.12.01.567409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Mitochondrial dysfunction has been linked to both idiopathic and familial forms of Parkinson's disease (PD). We have previously identified RCC1-like (RCC1L) as a protein of the inner mitochondrial membrane important to mitochondrial fusion. Herein, to test whether deficits in RCC1L mitochondrial function might be involved in PD pathology, we have selectively ablated the Rcc1l gene in the dopaminergic (DA) neurons of mice. A PD-like phenotype resulted that includes progressive movement abnormalities, paralleled by progressive degeneration of the nigrostriatal tract. Experimental and control groups were examined at 2, 3-4, and 5-6 months of age. Animals were tested in the open field task to quantify anxiety, exploratory drive, locomotion, and immobility; and in the cylinder test to quantify rearing behavior. Beginning at 3-4 months, both female and male Rcc1l knockout mice show rigid muscles and resting tremor, kyphosis and a growth deficit compared with heterozygous or wild type littermate controls. Rcc1l knockout mice begin showing locomotor impairments at 3-4 months, which progress until 5-6 months of age, at which age the Rcc1l knockout mice die. The progressive motor impairments were associated with progressive and significantly reduced tyrosine hydroxylase immunoreactivity in the substantia nigra pars compacta (SNc), and dramatic loss of nigral DA projections in the striatum. Dystrophic spherical mitochondria are apparent in the soma of SNc neurons in Rcc1l knockout mice as early as 1.5-2.5 months of age and become progressively more pronounced until 5-6 months. Together, the results reveal the RCC1L protein to be essential to in vivo mitochondrial function in DA neurons. Further characterization of this mouse model will determine whether it represents a new model for in vivo study of PD, and the putative role of the human RCC1L gene as a risk factor that might increase PD occurrence and severity in humans.
Collapse
Affiliation(s)
- Kaylin J. Ellioff
- Department of Neurology, University of Wisconsin, Madison WI, 53706
- Present Address, Department of Pharmacology, University of Washington, Seattle WA, 98195
| | | | - Alyssa Lentine
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison WI, 53705
| | - Ashley D. Welper
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison WI, 53705
| | - Corinna Burger
- Department of Neurology, University of Wisconsin, Madison WI, 53706
| | - Daniel S. Greenspan
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison WI, 53705
| |
Collapse
|
28
|
Jia Y, Li J, Wang Y, Ma Y, Chen L, Zhang H, Xue M, Liang H. Folic Acid Rescues Dopaminergic Neurons in MPTP-Induced Mice by Inhibiting the NLRP3 Inflammasome and Ameliorating Mitochondrial Impairment. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:5734-5745. [PMID: 38453725 DOI: 10.1021/acs.jafc.3c06337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Parkinson's disease (PD) is marked by the degeneration of dopaminergic neurons of the substantia nigra (SN), with neuroinflammation and mitochondrial dysfunction being key contributors. The neuroprotective potential of folic acid (FA) in the dopaminergic system of PD was assessed in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model. MPTP (20 mg/kg of body weight) was administered to C57BL/6J mice to simulate PD symptoms followed by FA treatment (5 mg/kg of body weight). Behavioral tests, pole, rotarod, and open-field tests, evaluated motor function, while immunohistochemistry, ELISA, RT-qPCR, and Western blotting quantified neuroinflammation, oxidative stress markers, and mitochondrial function. FA supplementation considerably improved motor performance, reduced homocysteine levels and mitigated oxidative damage in the SN. The FA-attenuated activation of the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome lessened glial cell activity and reduced neuroinflammation. At the molecular level, FA reduced DNA damage, downregulated phosphorylated p53, and induced the expression of peroxisome proliferator-activated receptor α coactivator 1α (PGC-1α), enhancing mitochondrial function. Therefore, FA exerts neuroprotection in MPTP-induced PD by inhibiting neuroinflammation via NLRP3 inflammasome suppression and promoting mitochondrial integrity through the p53-PGC-1α pathway. Notable limitations of our study include its reliance on a single animal model and the incompletely elucidated mechanisms underlying the impact of FA on mitochondrial dynamics. Future investigations will explore the clinical utility of FA and its molecular mechanisms, further advancing it as a potential therapeutic for managing and delaying the progression of PD.
Collapse
Affiliation(s)
- Yina Jia
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Jing Li
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Yutong Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Yiqing Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Lei Chen
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Huaqi Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Meilan Xue
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Hui Liang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| |
Collapse
|
29
|
Bartman S, Coppotelli G, Ross JM. Mitochondrial Dysfunction: A Key Player in Brain Aging and Diseases. Curr Issues Mol Biol 2024; 46:1987-2026. [PMID: 38534746 DOI: 10.3390/cimb46030130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/28/2024] Open
Abstract
Mitochondria are thought to have become incorporated within the eukaryotic cell approximately 2 billion years ago and play a role in a variety of cellular processes, such as energy production, calcium buffering and homeostasis, steroid synthesis, cell growth, and apoptosis, as well as inflammation and ROS production. Considering that mitochondria are involved in a multitude of cellular processes, mitochondrial dysfunction has been shown to play a role within several age-related diseases, including cancers, diabetes (type 2), and neurodegenerative diseases, although the underlying mechanisms are not entirely understood. The significant increase in lifespan and increased incidence of age-related diseases over recent decades has confirmed the necessity to understand the mechanisms by which mitochondrial dysfunction impacts the process of aging and age-related diseases. In this review, we will offer a brief overview of mitochondria, along with structure and function of this important organelle. We will then discuss the cause and consequence of mitochondrial dysfunction in the aging process, with a particular focus on its role in inflammation, cognitive decline, and neurodegenerative diseases, such as Huntington's disease, Parkinson's disease, and Alzheimer's disease. We will offer insight into therapies and interventions currently used to preserve or restore mitochondrial functioning during aging and neurodegeneration.
Collapse
Affiliation(s)
- Sydney Bartman
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Giuseppe Coppotelli
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Jaime M Ross
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| |
Collapse
|
30
|
Yao MF, Dang T, Wang HJ, Zhu XZ, Qiao C. Mitochondrial homeostasis regulation: A promising therapeutic target for Parkinson's disease. Behav Brain Res 2024; 459:114811. [PMID: 38103871 DOI: 10.1016/j.bbr.2023.114811] [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: 10/20/2023] [Revised: 12/10/2023] [Accepted: 12/10/2023] [Indexed: 12/19/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc) and the presence of Lewy bodies (LBs) or Lewy neurites (LNs) which consist of α-synuclein (α-syn) and a complex mix of other biomolecules. Mitochondrial dysfunction is widely believed to play an essential role in the pathogenesis of PD and other related neurodegenerative diseases. But mitochondrial dysfunction is subject to complex genetic regulation. There is increasing evidence that PD-related genes directly or indirectly affect mitochondrial integrity. Therefore, targeted regulation of mitochondrial function has great clinical application prospects in the treatment of PD. However, lots of PD drugs targeting mitochondria have been developed but their clinical therapeutic effects are not ideal. This review aims to reveal the role of mitochondrial dysfunction in the pathogenesis of neurodegenerative diseases based on the mitochondrial structure and function, which may highlight potential interventions and therapeutic targets for the development of PD drugs to recover mitochondrial dysfunction in neurodegenerative diseases.
Collapse
Affiliation(s)
- Meng-Fan Yao
- Department of Clinical Pharmabcy, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China; College of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Tao Dang
- Department of Clinical Pharmabcy, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China; College of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Hua-Jun Wang
- Department of Clinical Pharmabcy, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Xiao-Zhong Zhu
- Department of Cardiothoracic Surgery, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Chen Qiao
- Department of Clinical Pharmabcy, the Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, Jiangsu 212001, China; College of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
31
|
Ay M, Charli A, Langley M, Jang A, Padhi P, Jin H, Anantharam V, Kalyanaraman B, Kanthasamy A, Kanthasamy AG. Mito-metformin protects against mitochondrial dysfunction and dopaminergic neuronal degeneration by activating upstream PKD1 signaling in cell culture and MitoPark animal models of Parkinson's disease. Front Neurosci 2024; 18:1356703. [PMID: 38449738 PMCID: PMC10915001 DOI: 10.3389/fnins.2024.1356703] [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: 12/16/2023] [Accepted: 02/07/2024] [Indexed: 03/08/2024] Open
Abstract
Impaired mitochondrial function and biogenesis have strongly been implicated in the pathogenesis of Parkinson's disease (PD). Thus, identifying the key signaling mechanisms regulating mitochondrial biogenesis is crucial to developing new treatment strategies for PD. We previously reported that protein kinase D1 (PKD1) activation protects against neuronal cell death in PD models by regulating mitochondrial biogenesis. To further harness the translational drug discovery potential of targeting PKD1-mediated neuroprotective signaling, we synthesized mito-metformin (Mito-Met), a mitochondria-targeted analog derived from conjugating the anti-diabetic drug metformin with a triphenylphosphonium functional group, and then evaluated the preclinical efficacy of Mito-Met in cell culture and MitoPark animal models of PD. Mito-Met (100-300 nM) significantly activated PKD1 phosphorylation, as well as downstream Akt and AMPKα phosphorylation, more potently than metformin, in N27 dopaminergic neuronal cells. Furthermore, treatment with Mito-Met upregulated the mRNA and protein expression of mitochondrial transcription factor A (TFAM) implying that Mito-Met can promote mitochondrial biogenesis. Interestingly, Mito-Met significantly increased mitochondrial bioenergetics capacity in N27 dopaminergic cells. Mito-Met also reduced mitochondrial fragmentation induced by the Parkinsonian neurotoxicant MPP+ in N27 cells and protected against MPP+-induced TH-positive neurite loss in primary neurons. More importantly, Mito-Met treatment (10 mg/kg, oral gavage for 8 week) significantly improved motor deficits and reduced striatal dopamine depletion in MitoPark mice. Taken together, our results demonstrate that Mito-Met possesses profound neuroprotective effects in both in vitro and in vivo models of PD, suggesting that pharmacological activation of PKD1 signaling could be a novel neuroprotective translational strategy in PD and other related neurocognitive diseases.
Collapse
Affiliation(s)
- Muhammet Ay
- Parkinson’s Disorder Research Laboratory, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA, United States
| | - Adhithiya Charli
- Parkinson’s Disorder Research Laboratory, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA, United States
| | - Monica Langley
- Parkinson’s Disorder Research Laboratory, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA, United States
| | - Ahyoung Jang
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| | - Piyush Padhi
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| | - Huajun Jin
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| | - Vellareddy Anantharam
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| | | | - Arthi Kanthasamy
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| | - Anumantha G. Kanthasamy
- Parkinson’s Disorder Research Laboratory, Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, IA, United States
- Department of Physiology and Pharmacology, Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, United States
| |
Collapse
|
32
|
Granat L, Knorr DY, Ranson DC, Chakrabarty RP, Chandel NS, Bateman JM. A Drosophila model of mitochondrial disease phenotypic heterogeneity. Biol Open 2024; 13:bio060278. [PMID: 38304969 PMCID: PMC10924217 DOI: 10.1242/bio.060278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/22/2024] [Indexed: 02/03/2024] Open
Abstract
Mutations in genes that affect mitochondrial function cause primary mitochondrial diseases. Mitochondrial diseases are highly heterogeneous and even patients with the same mitochondrial disease can exhibit broad phenotypic heterogeneity, which is poorly understood. Mutations in subunits of mitochondrial respiratory complex I cause complex I deficiency, which can result in severe neurological symptoms and death in infancy. However, some complex I deficiency patients present with much milder symptoms. The most common nuclear gene mutated in complex I deficiency is the highly conserved core subunit NDUFS1. To model the phenotypic heterogeneity in complex I deficiency, we used RNAi lines targeting the Drosophila NDUFS1 homolog ND-75 with different efficiencies. Strong knockdown of ND-75 in Drosophila neurons resulted in severe behavioural phenotypes, reduced lifespan, altered mitochondrial morphology, reduced endoplasmic reticulum (ER)-mitochondria contacts and activation of the unfolded protein response (UPR). By contrast, weak ND-75 knockdown caused much milder behavioural phenotypes and changes in mitochondrial morphology. Moreover, weak ND-75 did not alter ER-mitochondria contacts or activate the UPR. Weak and strong ND-75 knockdown resulted in overlapping but distinct transcriptional responses in the brain, with weak knockdown specifically affecting proteosome activity and immune response genes. Metabolism was also differentially affected by weak and strong ND-75 knockdown including gamma-aminobutyric acid (GABA) levels, which may contribute to neuronal dysfunction in ND-75 knockdown flies. Several metabolic processes were only affected by strong ND-75 knockdown including the pentose phosphate pathway and the metabolite 2-hydroxyglutarate (2-HG), suggesting 2-HG as a candidate biomarker of severe neurological mitochondrial disease. Thus, our Drosophila model provides the means to dissect the mechanisms underlying phenotypic heterogeneity in mitochondrial disease.
Collapse
Affiliation(s)
- Lucy Granat
- Maurice Wohl Clinical Neuroscience Institute, King's College London, 5 Cutcombe Road, London SE5 9RX, UK
| | - Debbra Y. Knorr
- Maurice Wohl Clinical Neuroscience Institute, King's College London, 5 Cutcombe Road, London SE5 9RX, UK
| | - Daniel C. Ranson
- Maurice Wohl Clinical Neuroscience Institute, King's College London, 5 Cutcombe Road, London SE5 9RX, UK
| | - Ram Prosad Chakrabarty
- Department of Medicine, Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Navdeep S. Chandel
- Department of Medicine, Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Joseph M. Bateman
- Maurice Wohl Clinical Neuroscience Institute, King's College London, 5 Cutcombe Road, London SE5 9RX, UK
| |
Collapse
|
33
|
Carbonell-Roig J, Aaltonen A, Cartocci V, McGuirt A, Mosharov E, Kehr J, Lieberman OJ, Sulzer D, Borgkvist A, Santini E. Dysregulated acetylcholine-mediated dopamine neurotransmission in the eIF4E Tg mouse model of autism spectrum disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.29.577831. [PMID: 38352367 PMCID: PMC10862723 DOI: 10.1101/2024.01.29.577831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2024]
Abstract
Autism Spectrum Disorders (ASD) consist of diverse neurodevelopmental conditions where core behavioral symptoms are critical for diagnosis. Altered dopamine neurotransmission in the striatum has been suggested to contribute to the behavioral features of ASD. Here, we examine dopamine neurotransmission in a mouse model of ASD characterized by elevated expression of the eukaryotic initiation factor 4E (eIF4E), a key regulator of cap-dependent translation, using a comprehensive approach that encompasses genetics, behavior, synaptic physiology, and imaging. The results indicate that increased eIF4E expression leads to behavioral inflexibility and impaired striatal dopamine release. The loss of normal dopamine neurotransmission is due to a defective nicotinic receptor signaling that regulates calcium dynamics in dopaminergic axons. These findings reveal an intricate interplay between eIF4E, DA neurotransmission, and behavioral flexibility, provide a mechanistic understanding of ASD symptoms and offer a foundation for targeted therapeutic interventions.
Collapse
|
34
|
Chen YH, Kuo TT, Wang V, Cheng PW, Huang EYK, Ma KH, Greig NH, Olson L, Hoffer BJ, Tseng KY. Serotonergic Regulation of Synaptic Dopamine Levels Mitigates L-DOPA-Induced Dyskinesia in a Mouse Model of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2024; 14:941-964. [PMID: 38905058 PMCID: PMC11307072 DOI: 10.3233/jpd-240080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/20/2024] [Indexed: 06/23/2024]
Abstract
Background The serotonin (5-HT) system can manipulate the processing of exogenous L-DOPA in the DA-denervated striatum, resulting in the modulation of L-DOPA-induced dyskinesia (LID). Objective To characterize the effects of the serotonin precursor 5-hydroxy-tryptophan (5-HTP) or the serotonin transporter (SERT) inhibitor, Citalopram on L-DOPA-induced behavior, neurochemical signals, and underlying protein expressions in an animal model of Parkinson's disease. Methods MitoPark (MP) mice at 20 weeks of age, subjected to a 14-day administration of L-DOPA/Carbidopa, displayed dyskinesia, referred to as LID. Subsequent investigations explored the effects of 5-HT-modifying agents, such as 5-HTP and Citalopram, on abnormal involuntary movements (AIMs), locomotor activity, neurochemical signals, serotonin transporter activity, and protein expression in the DA-denervated striatum of LID MP mice. Results 5-HTP exhibited duration-dependent suppressive effects on developing and established LID, especially related to abnormal limb movements observed in L-DOPA-primed MP mice. However, Citalopram, predominantly suppressed abnormal axial movement induced by L-DOPA in LID MP mice. We demonstrated that 5-HTP could decrease L-DOPA-upregulation of DA turnover rates while concurrently upregulating 5-HT metabolism. Additionally, 5-HTP was shown to reduce the expressions of p-ERK and p-DARPP-32 in the striatum of LID MP mice. The effect of Citalopram in alleviating LID development may be attributed to downregulation of SERT activity in the dorsal striatum of LID MP mice. Conclusions While both single injection of 5-HTP and Citalopram effectively mitigated the development of LID, the difference in mitigation of AIM subtypes may be linked to the unique effects of these two serotonergic agents on L-DOPA-derived DA and 5-HT metabolism.
Collapse
Affiliation(s)
- Yuan-Hao Chen
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan
- National Defense Medical Center, Taipei, Taiwan
| | - Tung-Tai Kuo
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Vicki Wang
- Doctoral Degree Program in Translational Medicine, National Defense Medical Center and Academia Sinica, Taipei, Taiwan
| | - Pin-Wen Cheng
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan
| | | | - Kuo-Hsing Ma
- Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, National Institutes of Health (NIH), Baltimore, MD, USA
| | - Lars Olson
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Barry J. Hoffer
- Department of Neurosurgery, University Hospitals of Cleveland, Cleveland, OH, USA
| | - Kuan-Yin Tseng
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan
- National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
35
|
Venkatesan D, Iyer M, Narayanasamy A, Gopalakrishnan AV, Vellingiri B. Plausible Role of Mitochondrial DNA Copy Number in Neurodegeneration-a Need for Therapeutic Approach in Parkinson's Disease (PD). Mol Neurobiol 2023; 60:6992-7008. [PMID: 37523043 DOI: 10.1007/s12035-023-03500-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/11/2023] [Indexed: 08/01/2023]
Abstract
Parkinson's disease (PD) is an advancing age-associated progressive brain disorder which has various diverse factors, among them mitochondrial dysfunction involves in dopaminergic (DA) degeneration. Aging causes a rise in mitochondrial abnormalities which leads to structural and functional modifications in neuronal activity and cell death in PD. This ends in deterioration of mitochondrial function, mitochondrial alterations, mitochondrial DNA copy number (mtDNA CN) and oxidative phosphorylation (OXPHOS) capacity. mtDNA levels or mtDNA CN in PD have reported that mtDNA depletion would be a predisposing factor in PD pathogenesis. To maintain the mtDNA levels, therapeutic approaches have been focused on mitochondrial biogenesis in PD. The depletion of mtDNA levels in PD can be influenced by autophagic dysregulation, apoptosis, neuroinflammation, oxidative stress, sirtuins, and calcium homeostasis. The current review describes the regulation of mtDNA levels and discusses the plausible molecular pathways in mtDNA CN depletion in PD pathogenesis. We conclude by suggesting further research on mtDNA depletion which might show a promising effect in predicting and diagnosing PD.
Collapse
Affiliation(s)
- Dhivya Venkatesan
- Centre for Neuroscience, Department of Biotechnology, Karpagam Academy of Higher Education (Deemed to Be University), Coimbatore, 641021, India
| | - Mahalaxmi Iyer
- Centre for Neuroscience, Department of Biotechnology, Karpagam Academy of Higher Education (Deemed to Be University), Coimbatore, 641021, India
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632014, India
| | - Balachandar Vellingiri
- Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India.
| |
Collapse
|
36
|
Pogorelov VM, Martini ML, Jin J, Wetsel WC, Caron MG. Dopamine-Depleted Dopamine Transporter Knockout (DDD) Mice: Dyskinesia with L-DOPA and Dopamine D1 Agonists. Biomolecules 2023; 13:1658. [PMID: 38002340 PMCID: PMC10669682 DOI: 10.3390/biom13111658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
L-DOPA is the mainstay of treatment for Parkinson's disease (PD). However, over time this drug can produce dyskinesia. A useful acute PD model for screening novel compounds for anti-parkinsonian and L-DOPA-induced dyskinesia (LID) are dopamine-depleted dopamine-transporter KO (DDD) mice. Treatment with α-methyl-para-tyrosine rapidly depletes their brain stores of DA and renders them akinetic. During sensitization in the open field (OF), their locomotion declines as vertical activities increase and upon encountering a wall they stand on one leg or tail and engage in climbing behavior termed "three-paw dyskinesia". We have hypothesized that L-DOPA induces a stereotypic activation of locomotion in DDD mice, where they are unable to alter the course of their locomotion, and upon encountering walls engage in "three-paw dyskinesia" as reflected in vertical counts or beam-breaks. The purpose of our studies was to identify a valid index of LID in DDD mice that met three criteria: (a) sensitization with repeated L-DOPA administration, (b) insensitivity to a change in the test context, and (c) stimulatory or inhibitory responses to dopamine D1 receptor agonists (5 mg/kg SKF81297; 5 and 10 mg/kg MLM55-38, a novel compound) and amantadine (45 mg/kg), respectively. Responses were compared between the OF and a circular maze (CM) that did not hinder locomotion. We found vertical counts and climbing were specific for testing in the OF, while oral stereotypies were sensitized to L-DOPA in both the OF and CM and responded to D1R agonists and amantadine. Hence, in DDD mice oral stereotypies should be used as an index of LID in screening compounds for PD.
Collapse
Affiliation(s)
- Vladimir M. Pogorelov
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, 354 Sands Building, 303 Research Drive, Durham, NC 27710, USA
| | - Michael L. Martini
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.L.M.); (J.J.)
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (M.L.M.); (J.J.)
| | - William C. Wetsel
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, 354 Sands Building, 303 Research Drive, Durham, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA;
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Marc G. Caron
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA;
| |
Collapse
|
37
|
Tresse E, Marturia-Navarro J, Sew WQG, Cisquella-Serra M, Jaberi E, Riera-Ponsati L, Fauerby N, Hu E, Kretz O, Aznar S, Issazadeh-Navikas S. Mitochondrial DNA damage triggers spread of Parkinson's disease-like pathology. Mol Psychiatry 2023; 28:4902-4914. [PMID: 37779111 PMCID: PMC10914608 DOI: 10.1038/s41380-023-02251-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 10/03/2023]
Abstract
In the field of neurodegenerative diseases, especially sporadic Parkinson's disease (sPD) with dementia (sPDD), the question of how the disease starts and spreads in the brain remains central. While prion-like proteins have been designated as a culprit, recent studies suggest the involvement of additional factors. We found that oxidative stress, damaged DNA binding, cytosolic DNA sensing, and Toll-Like Receptor (TLR)4/9 activation pathways are strongly associated with the sPDD transcriptome, which has dysregulated type I Interferon (IFN) signaling. In sPD patients, we confirmed deletions of mitochondrial (mt)DNA in the medial frontal gyrus, suggesting a potential role of damaged mtDNA in the disease pathophysiology. To explore its contribution to pathology, we used spontaneous models of sPDD caused by deletion of type I IFN signaling (Ifnb-/-/Ifnar-/- mice). We found that the lack of neuronal IFNβ/IFNAR leads to oxidization, mutation, and deletion in mtDNA, which is subsequently released outside the neurons. Injecting damaged mtDNA into mouse brain induced PDD-like behavioral symptoms, including neuropsychiatric, motor, and cognitive impairments. Furthermore, it caused neurodegeneration in brain regions distant from the injection site, suggesting that damaged mtDNA triggers spread of PDD characteristics in an "infectious-like" manner. We also discovered that the mechanism through which damaged mtDNA causes pathology in healthy neurons is independent of Cyclic GMP-AMP synthase and IFNβ/IFNAR, but rather involves the dual activation of TLR9/4 pathways, resulting in increased oxidative stress and neuronal cell death, respectively. Our proteomic analysis of extracellular vesicles containing damaged mtDNA identified the TLR4 activator, Ribosomal Protein S3 as a key protein involved in recognizing and extruding damaged mtDNA. These findings might shed light on new molecular pathways through which damaged mtDNA initiates and spreads PD-like disease, potentially opening new avenues for therapeutic interventions or disease monitoring.
Collapse
Affiliation(s)
- Emilie Tresse
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Biocentre, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Joana Marturia-Navarro
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Biocentre, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Wei Qi Guinevere Sew
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Biocentre, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Marina Cisquella-Serra
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Biocentre, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Elham Jaberi
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Biocentre, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Lluis Riera-Ponsati
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Biocentre, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Natasha Fauerby
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Biocentre, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Erling Hu
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Biocentre, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark
| | - Oliver Kretz
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susana Aznar
- Centre for Neuroscience and Stereology, University Hospital Bispebjerg-Frederiksberg, 2400, Copenhagen, Denmark
| | - Shohreh Issazadeh-Navikas
- Neuroinflammation Unit, Biotech Research & Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Biocentre, Ole Maaløes Vej 5, DK-2200, Copenhagen N, Denmark.
| |
Collapse
|
38
|
Xue J, Zhang J, Zhang J, Liu J, Wang F, Li K, Liu C. The Parkinson's disease-associated mutation LRRK2 G2385R alters mitochondrial biogenesis via the PGC-1α-TFAM pathway. Mitochondrion 2023; 73:10-18. [PMID: 37708949 DOI: 10.1016/j.mito.2023.09.002] [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: 07/10/2023] [Revised: 08/21/2023] [Accepted: 09/12/2023] [Indexed: 09/16/2023]
Abstract
Mutations in the Leucine-rich repeat protein kinase 2 (LRRK2) gene are the most frequent cause of familial Parkinson's disease (PD). Although LRRK2 has been extensively studied, the pathogenic mechanism of the LRRK2 G2385R mutation, which is most common in Asian populations, especially in the Chinese Han population, remains unclear. In this study, we demonstrated that the LRRK2 G2385R mutation in HEK293T cells led to a reduction in cellular PGC-1α protein expression and inhibition of mitochondrial biogenesis through the PGC-1α-TFAM pathway. This resulted in a decrease in mitochondrial genome expression, which in turn impaired the normal electron transfer process of the oxidative phosphorylation respiratory chain, leading to mitochondrial dysfunction and onset of apoptosis. The mitochondrial dysfunction and apoptosis caused by the LRRK2 G2385R mutation were significantly alleviated by antioxidant Idebenone, which provides a theoretical basis for the subsequent development of precise treatment specifically for PD patients with LRRK2 G2385R mutation. Further validation of our findings in neurons and animal models are necessary.
Collapse
Affiliation(s)
- Jian Xue
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China; Department of Neurology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Jinbao Zhang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Jinru Zhang
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junyi Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China; Department of Neurology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Fen Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Kai Li
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Chunfeng Liu
- Department of Neurology and Suzhou Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China.
| |
Collapse
|
39
|
Heo JY, Park AH, Lee MJ, Ryu MJ, Kim YK, Jang YS, Kim SJ, Shin SY, Son HJ, Stein TD, Huh YH, Chung SK, Choi SY, Kim JM, Hwang O, Shong M, Hyeon SJ, Lee J, Ryu H, Kim D, Kweon GR. Crif1 deficiency in dopamine neurons triggers early-onset parkinsonism. Mol Psychiatry 2023; 28:4474-4484. [PMID: 37648779 DOI: 10.1038/s41380-023-02234-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 08/07/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023]
Abstract
Mitochondrial dysfunction has been implicated in Parkinson's Disease (PD) progression; however, the mitochondrial factors underlying the development of PD symptoms remain unclear. One candidate is CR6-interacting factor1 (CRIF1), which controls translation and membrane insertion of 13 mitochondrial proteins involved in oxidative phosphorylation. Here, we found that CRIF1 mRNA and protein expression were significantly reduced in postmortem brains of elderly PD patients compared to normal controls. To evaluate the effect of Crif1 deficiency, we produced mice lacking the Crif1 gene in dopaminergic neurons (DAT-CRIF1-KO mice). From 5 weeks of age, DAT-CRIF1-KO mice began to show decreased dopamine production with progressive neuronal degeneration in the nigral area. At ~10 weeks of age, they developed PD-like behavioral deficits, including gait abnormalities, rigidity, and resting tremor. L-DOPA, a medication used to treat PD, ameliorated these defects at an early stage, although it was ineffective in older mice. Taken together, the observation that CRIF1 expression is reduced in human PD brains and deletion of CRIF1 in dopaminergic neurons leads to early-onset PD with stepwise PD progression support the conclusion that CRIF1-mediated mitochondrial function is important for the survival of dopaminergic neurons.
Collapse
Affiliation(s)
- Jun Young Heo
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Ah Hyung Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Min Joung Lee
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Min Jeong Ryu
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Yong Kyung Kim
- Research Center for Endocrine and Metabolic Diseases, Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Yun Seon Jang
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Soo Jeong Kim
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - So Yeon Shin
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Hyo Jin Son
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Thor D Stein
- Boston University Alzheimer's Disease Research Center and Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
- VA Bedford Healthcare System, Bedford, MA, 01730, USA
- VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Yang Hoon Huh
- Electron Microscopy Research center, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Sookja K Chung
- Faculty of Medicine & Dr Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
| | - Song Yi Choi
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Jin Man Kim
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea
| | - Onyou Hwang
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Minho Shong
- Graduate School of Medical Science and Education, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Seung Jae Hyeon
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Junghee Lee
- Boston University Alzheimer's Disease Research Center and Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Hoon Ryu
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| | - Daesoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea.
| | - Gi Ryang Kweon
- Department of Biochemistry, Chungnam National University School of Medicine, Daejeon, 35015, Republic of Korea.
| |
Collapse
|
40
|
Averina OA, Kuznetsova SA, Permyakov OA, Sergiev PV. Animal Models of Mitochondrial Diseases Associated with Nuclear Gene Mutations. Acta Naturae 2023; 15:4-22. [PMID: 38234606 PMCID: PMC10790356 DOI: 10.32607/actanaturae.25442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 10/05/2023] [Indexed: 01/19/2024] Open
Abstract
Mitochondrial diseases (MDs) associated with nuclear gene mutations are part of a large group of inherited diseases caused by the suppression of energy metabolism. These diseases are of particular interest, because nuclear genes encode not only most of the structural proteins of the oxidative phosphorylation system (OXPHOS), but also all the proteins involved in the OXPHOS protein import from the cytoplasm and their assembly in mitochondria. Defects in any of these proteins can lead to functional impairment of the respiratory chain, including dysfunction of complex I that plays a central role in cellular respiration and oxidative phosphorylation, which is the most common cause of mitopathologies. Mitochondrial diseases are characterized by an early age of onset and a progressive course and affect primarily energy-consuming tissues and organs. The treatment of MDs should be initiated as soon as possible, but the diagnosis of mitopathologies is extremely difficult because of their heterogeneity and overlapping clinical features. The molecular pathogenesis of mitochondrial diseases is investigated using animal models: i.e. animals carrying mutations causing MD symptoms in humans. The use of mutant animal models opens new opportunities in the study of genes encoding mitochondrial proteins, as well as the molecular mechanisms of mitopathology development, which is necessary for improving diagnosis and developing approaches to drug therapy. In this review, we present the most recent information on mitochondrial diseases associated with nuclear gene mutations and animal models developed to investigate them.
Collapse
Affiliation(s)
- O. A. Averina
- Institute of Functional Genomics, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
| | - S. A. Kuznetsova
- Institute of Functional Genomics, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
| | - O. A. Permyakov
- Institute of Functional Genomics, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
| | - P. V. Sergiev
- Institute of Functional Genomics, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
| |
Collapse
|
41
|
Rocha E, Chamoli M, Chinta SJ, Andersen JK, Wallis R, Bezard E, Goldberg M, Greenamyre T, Hirst W, Kuan WL, Kirik D, Niedernhofer L, Rappley I, Padmanabhan S, Trudeau LE, Spillantini M, Scott S, Studer L, Bellantuono I, Mortiboys H. Aging, Parkinson's Disease, and Models: What Are the Challenges? AGING BIOLOGY 2023; 1:e20230010. [PMID: 38978807 PMCID: PMC11230631 DOI: 10.59368/agingbio.20230010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Parkinson's disease (PD) is a chronic, neurodegenerative condition characterized by motor symptoms such as bradykinesia, rigidity, and tremor, alongside multiple nonmotor symptoms. The appearance of motor symptoms is linked to progressive dopaminergic neuron loss within the substantia nigra. PD incidence increases sharply with age, suggesting a strong association between mechanisms driving biological aging and the development and progression of PD. However, the role of aging in the pathogenesis of PD remains understudied. Numerous models of PD, including cell models, toxin-induced models, and genetic models in rodents and nonhuman primates (NHPs), reproduce different aspects of PD, but preclinical studies of PD rarely incorporate age as a factor. Studies using patient neurons derived from stem cells via reprogramming methods retain some aging features, but their characterization, particularly of aging markers and reproducibility of neuron type, is suboptimal. Investigation of age-related changes in PD using animal models indicates an association, but this is likely in conjunction with other disease drivers. The biggest barrier to drawing firm conclusions is that each model lacks full characterization and appropriate time-course assessments. There is a need to systematically investigate whether aging increases the susceptibility of mouse, rat, and NHP models to develop PD and understand the role of cell models. We propose that a significant investment in time and resources, together with the coordination and sharing of resources, knowledge, and data, is required to accelerate progress in understanding the role of biological aging in PD development and improve the reliability of models to test interventions.
Collapse
Affiliation(s)
- Emily Rocha
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Shankar J Chinta
- Buck Institute for Research on Aging, Novato, CA, USA
- Touro University California, College of Pharmacy, Vallejo, CA, USA
| | | | - Ruby Wallis
- The Healthy Lifespan Institute, Sheffield, United Kingdom
| | | | | | - Tim Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - We-Li Kuan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (BRAINS), Lund, Sweden
| | - Laura Niedernhofer
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Irit Rappley
- Recursion pharmaceuticals, Salt Lake City, UT, USA
| | | | - Louis-Eric Trudeau
- Department of pharmacology and physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Maria Spillantini
- Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | | | - Lorenz Studer
- The Center for Stem Cell Biology and Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Ilaria Bellantuono
- The Healthy Lifespan Institute, Sheffield, United Kingdom
- Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
| | - Heather Mortiboys
- The Healthy Lifespan Institute, Sheffield, United Kingdom
- Department of Neuroscience, Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kindgom
| |
Collapse
|
42
|
Olkhova EA, Smith LA, Bradshaw C, Gorman GS, Erskine D, Ng YS. Neurological Phenotypes in Mouse Models of Mitochondrial Disease and Relevance to Human Neuropathology. Int J Mol Sci 2023; 24:ijms24119698. [PMID: 37298649 DOI: 10.3390/ijms24119698] [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: 05/13/2023] [Revised: 05/24/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Mitochondrial diseases represent the most common inherited neurometabolic disorders, for which no effective therapy currently exists for most patients. The unmet clinical need requires a more comprehensive understanding of the disease mechanisms and the development of reliable and robust in vivo models that accurately recapitulate human disease. This review aims to summarise and discuss various mouse models harbouring transgenic impairments in genes that regulate mitochondrial function, specifically their neurological phenotype and neuropathological features. Ataxia secondary to cerebellar impairment is one of the most prevalent neurological features of mouse models of mitochondrial dysfunction, consistent with the observation that progressive cerebellar ataxia is a common neurological manifestation in patients with mitochondrial disease. The loss of Purkinje neurons is a shared neuropathological finding in human post-mortem tissues and numerous mouse models. However, none of the existing mouse models recapitulate other devastating neurological phenotypes, such as refractory focal seizures and stroke-like episodes seen in patients. Additionally, we discuss the roles of reactive astrogliosis and microglial reactivity, which may be driving the neuropathology in some of the mouse models of mitochondrial dysfunction, as well as mechanisms through which cellular death may occur, beyond apoptosis, in neurons undergoing mitochondrial bioenergy crisis.
Collapse
Affiliation(s)
- Elizaveta A Olkhova
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Laura A Smith
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Carla Bradshaw
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Gráinne S Gorman
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Daniel Erskine
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| | - Yi Shiau Ng
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre, Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, UK
| |
Collapse
|
43
|
Gan Q, Ding Y, Peng M, Chen L, Dong J, Hu J, Ma Y. The Potential of Edible and Medicinal Resource Polysaccharides for Prevention and Treatment of Neurodegenerative Diseases. Biomolecules 2023; 13:biom13050873. [PMID: 37238743 DOI: 10.3390/biom13050873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/30/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
As natural medicines in complementary and alternative medicine, edible and medicinal resources are being gradually recognized throughout the world. According to statistics from the World Health Organization, about 80% of the worldwide population has used edible and medicinal resource products to prevent and treat diseases. Polysaccharides, one of the main effective components in edible and medicinal resources, are considered ideal regulators of various biological responses due to their high effectiveness and low toxicity, and they have a wide range of possible applications for the development of functional foods for the regulation of common, frequently occurring, chronic and severe diseases. Such applications include the development of polysaccharide products for the prevention and treatment of neurodegenerative diseases that are difficult to control by a single treatment, which is of great value to the aging population. Therefore, we evaluated the potential of polysaccharides to prevent neurodegeneration by their regulation of behavioral and major pathologies, including abnormal protein aggregation and neuronal damage caused by neuronal apoptosis, autophagy, oxidative damage, neuroinflammation, unbalanced neurotransmitters, and poor synaptic plasticity. This includes multi-target and multi-pathway regulation involving the mitochondrial pathway, MAPK pathway, NF-κB pathway, Nrf2 pathway, mTOR pathway, PI3K/AKT pathway, P53/P21 pathway, and BDNF/TrkB/CREB pathway. In this paper, research into edible and medicinal resource polysaccharides for neurodegenerative diseases was reviewed in order to provide a basis for the development and application of polysaccharide health products and promote the recognition of functional products of edible and medicinal resources.
Collapse
Affiliation(s)
- Qingxia Gan
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory of Traditional Chinese Medicine Processing Technology, State Administration of Traditional Chinese Medicine, No. 1166, Wenjiang District, Chengdu 611137, China
| | - Yugang Ding
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory of Traditional Chinese Medicine Processing Technology, State Administration of Traditional Chinese Medicine, No. 1166, Wenjiang District, Chengdu 611137, China
| | - Maoyao Peng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory of Traditional Chinese Medicine Processing Technology, State Administration of Traditional Chinese Medicine, No. 1166, Wenjiang District, Chengdu 611137, China
| | - Linlin Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory of Traditional Chinese Medicine Processing Technology, State Administration of Traditional Chinese Medicine, No. 1166, Wenjiang District, Chengdu 611137, China
| | - Jijing Dong
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory of Traditional Chinese Medicine Processing Technology, State Administration of Traditional Chinese Medicine, No. 1166, Wenjiang District, Chengdu 611137, China
| | - Jiaxi Hu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuntong Ma
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory of Traditional Chinese Medicine Processing Technology, State Administration of Traditional Chinese Medicine, No. 1166, Wenjiang District, Chengdu 611137, China
| |
Collapse
|
44
|
Kwon YJ, Kwon OI, Hwang HJ, Shin HC, Yang S. Therapeutic effects of phlorotannins in the treatment of neurodegenerative disorders. Front Mol Neurosci 2023; 16:1193590. [PMID: 37305552 PMCID: PMC10249478 DOI: 10.3389/fnmol.2023.1193590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 04/27/2023] [Indexed: 06/13/2023] Open
Abstract
Phlorotannins are natural polyphenolic compounds produced by brown marine algae and are currently found in nutritional supplements. Although they are known to cross the blood-brain barrier, their neuropharmacological actions remain unclear. Here we review the potential therapeutic benefits of phlorotannins in the treatment of neurodegenerative diseases. In mouse models of Alzheimer's disease, ethanol intoxication and fear stress, the phlorotannin monomer phloroglucinol and the compounds eckol, dieckol and phlorofucofuroeckol A have been shown to improve cognitive function. In a mouse model of Parkinson's disease, phloroglucinol treatment led to improved motor performance. Additional neurological benefits associated with phlorotannin intake have been demonstrated in stroke, sleep disorders, and pain response. These effects may stem from the inhibition of disease-inducing plaque synthesis and aggregation, suppression of microglial activation, modulation of pro-inflammatory signaling, reduction of glutamate-induced excitotoxicity, and scavenging of reactive oxygen species. Clinical trials of phlorotannins have not reported significant adverse effects, suggesting these compounds to be promising bioactive agents in the treatment of neurological diseases. We therefore propose a putative biophysical mechanism of phlorotannin action in addition to future directions for phlorotannin research.
Collapse
Affiliation(s)
- Yoon Ji Kwon
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Oh Ig Kwon
- Botamedi Brain Health and Medical Care Company Limited, Central, Hong Kong SAR, China
| | - Hye Jeong Hwang
- Center for Molecular Intelligence, SUNY Korea, Incheon, Republic of Korea
| | - Hyeon-Cheol Shin
- Botamedi Brain Health and Medical Care Company Limited, Central, Hong Kong SAR, China
- Center for Molecular Intelligence, SUNY Korea, Incheon, Republic of Korea
| | - Sungchil Yang
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| |
Collapse
|
45
|
Moradi Vastegani S, Nasrolahi A, Ghaderi S, Belali R, Rashno M, Farzaneh M, Khoshnam SE. Mitochondrial Dysfunction and Parkinson's Disease: Pathogenesis and Therapeutic Strategies. Neurochem Res 2023:10.1007/s11064-023-03904-0. [PMID: 36943668 DOI: 10.1007/s11064-023-03904-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/21/2023] [Accepted: 02/28/2023] [Indexed: 03/23/2023]
Abstract
Parkinson's disease (PD) is a common age-related neurodegenerative disorder whose pathogenesis is not completely understood. Mitochondrial dysfunction and increased oxidative stress have been considered as major causes and central events responsible for the progressive degeneration of dopaminergic (DA) neurons in PD. Therefore, investigating mitochondrial disorders plays a role in understanding the pathogenesis of PD and can be an important therapeutic target for this disease. This study discusses the effect of environmental, genetic and biological factors on mitochondrial dysfunction and also focuses on the mitochondrial molecular mechanisms underlying neurodegeneration, and its possible therapeutic targets in PD, including reactive oxygen species generation, calcium overload, inflammasome activation, apoptosis, mitophagy, mitochondrial biogenesis, and mitochondrial dynamics. Other potential therapeutic strategies such as mitochondrial transfer/transplantation, targeting microRNAs, using stem cells, photobiomodulation, diet, and exercise were also discussed in this review, which may provide valuable insights into clinical aspects. A better understanding of the roles of mitochondria in the pathophysiology of PD may provide a rationale for designing novel therapeutic interventions in our fight against PD.
Collapse
Affiliation(s)
- Sadegh Moradi Vastegani
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ava Nasrolahi
- Infectious Ophthalmologic Research Center, Imam Khomeini Hospital Clinical Research Development Unit, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shahab Ghaderi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rafie Belali
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masome Rashno
- Asadabad School of Medical Sciences, Asadabad, Iran
- Student Research Committee, Asadabad School of Medical Sciences, Asadabad, Iran
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Esmaeil Khoshnam
- Persian Gulf Physiology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
46
|
Zhou J, Li J, Papaneri AB, Cui G. AJ76 and UH232 as potential agents for diagnosing early-stage Parkinson's disease. Neuropharmacology 2023; 226:109397. [PMID: 36623805 PMCID: PMC9901527 DOI: 10.1016/j.neuropharm.2022.109397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023]
Abstract
We recently reported that the "Dopamine Neuron Challenge Test" (DNC Test), a diagnostic method that measures the levels of dopamine metabolites in cerebrospinal fluid (CSF) and plasma samples after pharmacologically inducing a transient dopamine release, can detect early-stage Parkinson's disease (PD) with high sensitivity and selectivity in mouse models. The use of haloperidol in the original DNC test to challenge dopamine neurons was less than ideal, as it may cause extrapyramidal motor symptoms. Here we report an improved DNC Test, in which the original challenging agents, haloperidol and methylphenidate, are replaced by a single challenging agent, a dopamine autoreceptor preferring antagonist AJ76 or UH232. We show that the improved DNC Test can achieve the same level of sensitivity and selectivity in detecting early PD in a mouse model without causing motor side effects. These findings significantly improve the practicality of using the DNC Test as a screening or diagnostic test for detecting early-stage PD in the high-risk population in humans.
Collapse
Affiliation(s)
- Jingheng Zhou
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Drive, Research Triangle Park, NC, 27709, USA
| | - Jicheng Li
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Drive, Research Triangle Park, NC, 27709, USA
| | - Amy B Papaneri
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Drive, Research Triangle Park, NC, 27709, USA
| | - Guohong Cui
- In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Drive, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
47
|
PT320, a Sustained-Release GLP-1 Receptor Agonist, Ameliorates L-DOPA-Induced Dyskinesia in a Mouse Model of Parkinson's Disease. Int J Mol Sci 2023; 24:ijms24054687. [PMID: 36902115 PMCID: PMC10002999 DOI: 10.3390/ijms24054687] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/15/2023] [Accepted: 02/24/2023] [Indexed: 03/04/2023] Open
Abstract
To determine the efficacy of PT320 on L-DOPA-induced dyskinetic behaviors, and neurochemistry in a progressive Parkinson's disease (PD) MitoPark mouse model. To investigate the effects of PT320 on the manifestation of dyskinesia in L-DOPA-primed mice, a clinically translatable biweekly PT320 dose was administered starting at either 5 or 17-weeks-old mice. The early treatment group was given L-DOPA starting at 20 weeks of age and longitudinally evaluated up to 22 weeks. The late treatment group was given L-DOPA starting at 28 weeks of age and longitudinally observed up to 29 weeks. To explore dopaminergic transmission, fast scan cyclic voltammetry (FSCV) was utilized to measure presynaptic dopamine (DA) dynamics in striatal slices following drug treatments. Early administration of PT320 significantly mitigated the severity L-DOPA-induced abnormal involuntary movements; PT320 particularly improved excessive numbers of standing as well as abnormal paw movements, while it did not affect L-DOPA-induced locomotor hyperactivity. In contrast, late administration of PT320 did not attenuate any L-DOPA-induced dyskinesia measurements. Moreover, early treatment with PT320 was shown to not only increase tonic and phasic release of DA in striatal slices in L-DOPA-naïve MitoPark mice, but also in L-DOPA-primed animals. Early treatment with PT320 ameliorated L-DOPA-induced dyskinesia in MitoPark mice, which may be related to the progressive level of DA denervation in PD.
Collapse
|
48
|
Reduced OPA1, Mitochondrial Fragmentation and Increased Susceptibility to Apoptosis in Granular Corneal Dystrophy Type 2 Corneal Fibroblasts. Genes (Basel) 2023; 14:genes14030566. [PMID: 36980838 PMCID: PMC10048436 DOI: 10.3390/genes14030566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
The progressive degeneration of granular corneal dystrophy type 2 (GCD2) corneal fibroblasts is associated with altered mitochondrial function, but the underlying mechanisms are incompletely understood. We investigated whether an imbalance of mitochondrial dynamics contributes to mitochondrial dysfunction of GCD2 corneal fibroblasts. Transmission electron microscopy revealed several small, structurally abnormal mitochondria with altered cristae morphology in GCD2 corneal fibroblasts. Confocal microscopy showed enhanced mitochondrial fission and fragmented mitochondrial tubular networks. Western blotting revealed higher levels of MFN1, MFN2, and pDRP1 and decreased levels of OPA1 and FIS1 in GCD2. OPA1 reduction by short hairpin RNA (shRNA) resulted in fragmented mitochondrial tubular networks and increased susceptibility to mitochondrial stress-induced apoptosis. A decrease in the mitochondrial biogenesis-related transcription factors NRF1 and PGC1α was observed, while there was an increase in the mitochondrial membrane proteins TOM20 and TIM23. Additionally, reduced levels of mitochondrial DNA (mtDNA) were exhibited in GCD2 corneal fibroblasts. These observations suggest that altered mitochondrial fission/fusion and biogenesis are the critical molecular mechanisms that cause mitochondrial dysfunction contributing to the degeneration of GCD2 corneal fibroblasts.
Collapse
|
49
|
Zheng Y, Zhang J, Zhu X, Wei Y, Zhao W, Si S, Li Y. A Mitochondrial Perspective on Noncommunicable Diseases. Biomedicines 2023; 11:biomedicines11030647. [PMID: 36979626 PMCID: PMC10045938 DOI: 10.3390/biomedicines11030647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/05/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Mitochondria are the center of energy metabolism in eukaryotic cells and play a central role in the metabolism of living organisms. Mitochondrial diseases characterized by defects in oxidative phosphorylation are the most common congenital diseases. Meanwhile, mitochondrial dysfunction caused by secondary factors such as non-inherited genetic mutations can affect normal physiological functions of human cells, induce apoptosis, and lead to the development of various diseases. This paper reviewed several major factors and mechanisms that contribute to mitochondrial dysfunction and discussed the development of diseases closely related to mitochondrial dysfunction and drug treatment strategies discovered in recent years.
Collapse
Affiliation(s)
- Yifan Zheng
- Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jing Zhang
- Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaohong Zhu
- Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yuanjuan Wei
- Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wuli Zhao
- NHC Key Laboratory of Antibiotic Bioengineering, Laboratory of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Correspondence: (W.Z.); (S.S.); (Y.L.)
| | - Shuyi Si
- Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Correspondence: (W.Z.); (S.S.); (Y.L.)
| | - Yan Li
- Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Correspondence: (W.Z.); (S.S.); (Y.L.)
| |
Collapse
|
50
|
Fifel K, Yanagisawa M, Deboer T. Mechanisms of Sleep/Wake Regulation under Hypodopaminergic State: Insights from MitoPark Mouse Model of Parkinson's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2203170. [PMID: 36515271 PMCID: PMC9929135 DOI: 10.1002/advs.202203170] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 11/16/2022] [Indexed: 06/17/2023]
Abstract
Sleep/wake alterations are predominant in neurological and neuropsychiatric disorders involving dopamine dysfunction. Unfortunately, specific, mechanisms-based therapies for these debilitating sleep problems are currently lacking. The pathophysiological mechanisms of sleep/wake alterations within a hypodopaminergic MitoPark mouse model of Parkinson's disease (PD) are investigated. MitoPark mice replicate most PD-related sleep alterations, including sleep fragmentation, hypersomnia, and daytime sleepiness. Surprisingly, these alterations are not accounted for by a dysfunction in the circadian or homeostatic regulatory processes of sleep, nor by acute masking effects of light or darkness. Rather, the sleep phenotype is linked with the impairment of instrumental arousal and sleep modulation by behavioral valence. These alterations correlate with changes in high-theta (8-11.5 Hz) electroencephalogram power density during motivationally-charged wakefulness. These results demonstrate that sleep/wake alterations induced by dopamine dysfunction are mediated by impaired modulation of sleep by motivational valence and provide translational insights into sleep problems associated with disorders linked to dopamine dysfunction.
Collapse
Affiliation(s)
- Karim Fifel
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of Tsukuba1‐1‐1 TennodaiTsukubaIbaraki305–8575Japan
- Department of Cell and Chemical BiologyLaboratory of NeurophysiologyLeiden University Medical CenterP.O. Box 9600Leiden2300 RCThe Netherlands
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI‐IIIS)University of Tsukuba1‐1‐1 TennodaiTsukubaIbaraki305–8575Japan
| | - Tom Deboer
- Department of Cell and Chemical BiologyLaboratory of NeurophysiologyLeiden University Medical CenterP.O. Box 9600Leiden2300 RCThe Netherlands
| |
Collapse
|