1
|
Deng F, Yang D, Qing L, Chen Y, Zou J, Jia M, Wang Q, Jiang R, Huang L. Exploring the interaction between the gut microbiota and cyclic adenosine monophosphate-protein kinase A signaling pathway: a potential therapeutic approach for neurodegenerative diseases. Neural Regen Res 2025; 20:3095-3112. [PMID: 39589173 DOI: 10.4103/nrr.nrr-d-24-00607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 09/10/2024] [Indexed: 11/27/2024] Open
Abstract
The interaction between the gut microbiota and cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) signaling pathway in the host's central nervous system plays a crucial role in neurological diseases and enhances communication along the gut-brain axis. The gut microbiota influences the cAMP-PKA signaling pathway through its metabolites, which activates the vagus nerve and modulates the immune and neuroendocrine systems. Conversely, alterations in the cAMP-PKA signaling pathway can affect the composition of the gut microbiota, creating a dynamic network of microbial-host interactions. This reciprocal regulation affects neurodevelopment, neurotransmitter control, and behavioral traits, thus playing a role in the modulation of neurological diseases. The coordinated activity of the gut microbiota and the cAMP-PKA signaling pathway regulates processes such as amyloid-β protein aggregation, mitochondrial dysfunction, abnormal energy metabolism, microglial activation, oxidative stress, and neurotransmitter release, which collectively influence the onset and progression of neurological diseases. This study explores the complex interplay between the gut microbiota and cAMP-PKA signaling pathway, along with its implications for potential therapeutic interventions in neurological diseases. Recent pharmacological research has shown that restoring the balance between gut flora and cAMP-PKA signaling pathway may improve outcomes in neurodegenerative diseases and emotional disorders. This can be achieved through various methods such as dietary modifications, probiotic supplements, Chinese herbal extracts, combinations of Chinese herbs, and innovative dosage forms. These findings suggest that regulating the gut microbiota and cAMP-PKA signaling pathway may provide valuable evidence for developing novel therapeutic approaches for neurodegenerative diseases.
Collapse
Affiliation(s)
- Fengcheng Deng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Dan Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Lingxi Qing
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Yifei Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Jilian Zou
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Meiling Jia
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Qian Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Runda Jiang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Lihua Huang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| |
Collapse
|
2
|
Yan M, Wu H, Wu T, Wang Y, Su C, Li D, Han X. Microcystin-LR Exposure Damages Neurons by Inducing α-Syn Aggregation via MAPK4/GATA2/SNCA and PP2A/GRKs Pathways. Mol Neurobiol 2024:10.1007/s12035-024-04683-7. [PMID: 39738876 DOI: 10.1007/s12035-024-04683-7] [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: 06/06/2024] [Accepted: 12/23/2024] [Indexed: 01/02/2025]
Abstract
Microcystin-LR (MC-LR) is a natural neurotoxin with strong toxicity, and studies have demonstrated that chronic MC-LR exposure generated Parkinson-like dyskinesia in mice. Parkinson's disease (PD) is a neurologic degenerative disease mostly occurring in elderly people, and the progressive loss of dopaminergic neurons and the formation of Lewy bodies are the hallmark pathological features. The main component of Lewy bodies is α-synuclein (α-syn) encoded by the SNCA gene, and the copy number mutation of SNCA gene can promote the overexpression of α-syn. A mouse model of MC-LR exposure for 15 months was established to confirm the deposition of Lewy bodies. SH-SY5Y cells exposed to MC-LR were constructed as an in vitro model of PD, and the transcription factor that regulated the SNCA gene (the encoding gene of α-syn) was identified through the database. MC-LR enhanced the transcription level of SNCA gene and upregulated α-syn protein expression by promoting MAPK4 into the nucleus and binding to GATA2 295-480 fragment. In addition, MC-LR inhibited PP2A activity and activated GRKs kinase to promote α-syn phosphorylation at Ser129. These results suggest that MC-LR is involved in α-syn aggregate formation and PD pathogenesis by enhancing SNCA transcriptional activity to promote α-syn elevation via the MAPK4/GATA2 pathway and inducing α-syn phosphorylation via the PP2A/GRKs pathway.
Collapse
Affiliation(s)
- Minghao Yan
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Huifang Wu
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Tong Wu
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Yuhan Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Chengxiang Su
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Dongmei Li
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Xiaodong Han
- State Key Laboratory of Analytical Chemistry for Life Science, Division of Anatomy and Histo-Embryology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China.
| |
Collapse
|
3
|
Regoni M, Zanetti L, Sevegnani M, Domenicale C, Magnabosco S, Patel JC, Fernandes MK, Feeley RM, Monzani E, Mini C, Comai S, Cherchi L, De Gregorio D, Soliman I, Ruto F, Croci L, Consalez G, Rodighiero S, Ciammola A, Valtorta F, Morari M, Piccoli G, Rice ME, Sassone J. Dopamine neuron dysfunction and loss in the PrknR275W mouse model of juvenile parkinsonism. Brain 2024; 147:4017-4025. [PMID: 39350737 DOI: 10.1093/brain/awae276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 11/07/2024] Open
Abstract
Mutations in the PRKN gene encoding the protein parkin cause autosomal recessive juvenile parkinsonism (ARJP). Harnessing this mutation to create an early-onset Parkinson's disease mouse model would provide a unique opportunity to clarify the mechanisms involved in the neurodegenerative process and lay the groundwork for the development of neuroprotective strategies. To this end, we created a knock-in mouse carrying the homozygous PrknR275W mutation, which is the missense mutation with the highest allelic frequency in PRKN patients. We evaluated the anatomical and functional integrity of the nigrostriatal dopamine (DA) pathway, as well as motor behaviour in PrknR275W mice of both sexes. We report here that PrknR275W mice show early DA neuron dysfunction, age-dependent loss of DA neurons in the substantia nigra, decreased DA content and stimulus-evoked DA release in the striatum, and progressive motor impairment. Together, these data show that the PrknR275W mouse recapitulates key features of ARJP. Thus, these studies fill a critical need in the field by introducing a promising new Parkinson's disease model in which to study causative mechanisms of the disease and test therapeutic strategies.
Collapse
Affiliation(s)
- Maria Regoni
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Letizia Zanetti
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Martina Sevegnani
- Dulbecco Telethon Institute, Laboratory of Biology of Synapse, Center for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Chiara Domenicale
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| | - Stefano Magnabosco
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| | - Jyoti C Patel
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Megan K Fernandes
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ryan M Feeley
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Elena Monzani
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Cecilia Mini
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, 20132 Milan, Italy
- Department of Statistics, Computer Science, Applications (DiSIA), University of Florence, 50134 Florence, Italy
| | - Stefano Comai
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy
- Department of Biomedical Sciences, University of Padova, 35122 Padova, Italy
| | - Laura Cherchi
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Danilo De Gregorio
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Isabella Soliman
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Fabio Ruto
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Laura Croci
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giacomo Consalez
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Simona Rodighiero
- Department of Experimental Oncology, European Institute of Oncology (IEO) IRCCS, Milan 20139, Italy
| | - Andrea Ciammola
- IRCCS Istituto Auxologico Italiano, Department of Neurology and Laboratory of Neuroscience, 20149 Milan, Italy
| | - Flavia Valtorta
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Michele Morari
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy
| | - Giovanni Piccoli
- Dulbecco Telethon Institute, Laboratory of Biology of Synapse, Center for Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Margaret E Rice
- Department of Neurosurgery, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jenny Sassone
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, 20132 Milan, Italy
| |
Collapse
|
4
|
Zhang F, Ye Z, Xie Y, Liu M, Zhang L, Zhang J, Xu Z. Levodopa-induced dyskinesia: brain iron deposition as a new hypothesis. Biometals 2024; 37:1307-1323. [PMID: 39212870 DOI: 10.1007/s10534-024-00628-8] [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: 07/08/2024] [Accepted: 08/10/2024] [Indexed: 09/04/2024]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease in the older adults. The main pathological change in PD is the degenerative death of dopamine (DA) neurons in the midbrain substantia nigra, which causes a significant decrease in the DA content of the striatum. However, the exact etiology of this pathological change remains unclear. Genetic factors, environmental factors, aging, and oxidative stress may be involved in the degenerative death of dopaminergic neurons in PD. Pharmacological treatment using levodopa (L-DOPA) remains the main treatment for PD. Most patients with PD consuming L-DOPA for a long time usually develop levodopa-induced dyskinesia (LID) after 6.5 years of use, and LID seriously affects the quality of life and increases the risk of disability. Recently, studies have revealed that cerebral iron deposition may be involved in LID development and that iron deposition has neurotoxic effects and accelerates disease onset. However, the relationship between cerebral iron deposition and LID remains unclear. Herein, we reviewed the mechanisms by which iron deposition may be associated with LID development, which are mainly related to oxidative stress, neuroinflammation, and mitochondrial and lysosomal dysfunction. Using iron as an important target, the search and development of safe and effective brain iron scavengers, and thus the alleviation and treatment of LID, has a very important scientific and clinical value, as well as a good application prospect.
Collapse
Affiliation(s)
- Fanshi Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Zhuofan Ye
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
- Department of Neurology, Guizhou Pronvincial People's Hospital, Guiyang, China
| | - Yuanyang Xie
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Mei Liu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Li Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China
| | - Jun Zhang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China.
| | - Zucai Xu
- Department of Neurology, Affiliated Hospital of Zunyi Medical University, No. 149, Dalian Road, Huichuan District, Zunyi City, 563000, , Guizhou Province, China.
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
5
|
D'Addario SL, Rosina E, Massaro Cenere M, Bagni C, Mercuri NB, Ledonne A. ErbB inhibition rescues nigral dopamine neuron hyperactivity and repetitive behaviors in a mouse model of fragile X syndrome. Mol Psychiatry 2024:10.1038/s41380-024-02831-y. [PMID: 39543371 DOI: 10.1038/s41380-024-02831-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
Repetitive stereotyped behaviors are core symptoms of autism spectrum disorders (ASD) and fragile X syndrome (FXS), the prevalent genetic cause of intellectual disability and autism. The nigrostriatal dopamine (DA) circuit rules movement and creation of habits and sequential behaviors; therefore, its dysregulation could promote autistic repetitive behaviors. Nevertheless, inspection of substantia nigra pars compacta (SNpc) DA neurons in ASD models has been overlooked and specific evidence of their altered activity in ASD and FXS is absent. Here, we show that hyperactivity of SNpc DA neurons is an early feature of FXS. The underlying mechanism relies on an interplay between metabotropic glutamate receptor 1 (mGluR1) and ErbB tyrosine kinases, receptors for the neurotrophic and differentiation factors known as neuregulins. Up-regulation of ErbB4 and ErbB2 in nigral DA neurons drives neuronal hyperactivity and repetitive behaviors of the FXS mouse, concurrently rescued by ErbB inhibition. In conclusion, beyond providing the first evidence that nigral DA neuron hyperactivity is a signature of FXS and nigral mGluR1 and ErbB4/2 play a relevant role in FXS etiology, we demonstrate that inhibiting ErbB is a valuable pharmacological approach to attenuate stereotyped repetitive behaviors, thus opening an avenue toward innovative therapies for ASD and FXS treatment.
Collapse
Affiliation(s)
| | - Eleonora Rosina
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | | | - Claudia Bagni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Nicola B Mercuri
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Neurology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ada Ledonne
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy.
- Pharmacology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
6
|
Massaro Cenere M, Tiberi M, Paldino E, D'Addario SL, Federici M, Giacomet C, Cutuli D, Matteocci A, Cossa F, Zarrilli B, Casadei N, Ledonne A, Petrosini L, Berretta N, Fusco FR, Chiurchiù V, Mercuri NB. Systemic inflammation accelerates neurodegeneration in a rat model of Parkinson's disease overexpressing human alpha synuclein. NPJ Parkinsons Dis 2024; 10:213. [PMID: 39500895 PMCID: PMC11538257 DOI: 10.1038/s41531-024-00824-w] [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: 02/05/2024] [Accepted: 10/19/2024] [Indexed: 11/08/2024] Open
Abstract
Increasing efforts have been made to elucidate how genetic and environmental factors interact in Parkinson's disease (PD). In the present study, we assessed the development of symptoms on a genetic PD rat model that overexpresses human α-synuclein (Snca+/+) at a presymptomatic age, exposed to a pro-inflammatory insult by intraperitoneal injection of lipopolysaccharide (LPS), using immunohistology, high-dimensional flow cytometry, constant potential amperometry, and behavioral analyses. A single injection of LPS into WT and Snca+/+ rats triggered long-lasting increase in the activation of pro-inflammatory microglial markers, monocytes, and T lymphocytes. However, only LPS Snca+/+ rats showed dopaminergic neuronal loss in the substantia nigra pars compacta (SNpc), associated with a reduction in the release of evoked dopamine in the striatum. No significant changes were observed in the behavioral domain. We propose our double-hit animal as a reliable model to investigate the mechanisms whereby α-synuclein and inflammation interact to promote neurodegeneration in PD.
Collapse
Affiliation(s)
- Mariangela Massaro Cenere
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy.
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Marta Tiberi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Laboratory of Resolution of Neuroinflammation, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Emanuela Paldino
- Laboratory of Neuroanatomy, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Sebastian Luca D'Addario
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Mauro Federici
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Cecilia Giacomet
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Debora Cutuli
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Psychology, Sapienza University of Rome, Rome, Italy
| | - Alessandro Matteocci
- Laboratory of Resolution of Neuroinflammation, Santa Lucia Foundation IRCCS, Rome, Italy
- PhD program in Immunology, Molecular Medicine and Applied biotechnologies, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Francesca Cossa
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Beatrice Zarrilli
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Ada Ledonne
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Laura Petrosini
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Nicola Berretta
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | | | - Valerio Chiurchiù
- Laboratory of Resolution of Neuroinflammation, Santa Lucia Foundation IRCCS, Rome, Italy
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - Nicola B Mercuri
- Department of Experimental Neuroscience, Santa Lucia Foundation IRCCS, Rome, Italy.
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
7
|
Zhou W, Daniels S, Singh V, Menard M, Escobar Galvis ML, Chu HY. α-Synuclein aggregation decreases cortico-amygdala connectivity and impairs social behavior in mice. Neurobiol Dis 2024; 202:106702. [PMID: 39406290 DOI: 10.1016/j.nbd.2024.106702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/03/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024] Open
Abstract
Abnormal accumulation of insoluble α-synuclein (α-Syn) inclusions in neurons, neurites, and glial cells is the defining neuropathology of synucleinopathies, including Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy. Accumulation of α-Syn inclusions in the amygdala has been well-documented in post-mortem studies of PD and DLB brains, as well as preclinical animal models of these conditions. Though α-Syn pathology is closely associated with neurodegeneration, there is a poor correlation between neuronal loss in the amygdala and the clinical features of PD and DLB. Moreover, functional interaction between the cerebral cortex and the amygdala is critical to regulating emotion, motivation, and social behaviors. The cortico-amygdala functional interaction is likely to be disrupted by the development of α-Syn pathology in the brain. Thus, we hypothesize that neuronal α-Syn inclusions disrupt cortical modulation of the amygdala circuits and are sufficient to drive social behavioral deficits. In the present work, we designed a series of longitudinal studies to rigorously measure the time courses of neurodegeneration, functional impairment of cortico-amygdala connectivity, and development of amygdala-dependent social behavioral deficits to test this hypothesis. We injected α-Syn preformed fibrils (PFFs) into the dorsal striatum to induce α-Syn aggregation in the amygdala and the medial prefrontal cortex (mPFC) of C57BL6 mice of both sexes, followed by a detailed analysis of temporal development of α-Syn pathology, synaptic deficits, and neuronal loss in the amygdala, as well as behavioral deficits at 3-12 months post injections. Development of α-Syn inclusions caused losses of cortical axon terminals and cell death in the basolateral amygdala (BLA) at 6- and 12-months post injections, respectively. At a relatively early stage of 3 months post injections, the connection strength of the mPFC-BLA synapse was decreased in PFFs-injection mice compared to controls. Meanwhile, the PFFs-injected mice showed impaired social interaction behavior, which was rescued by chemogenetic stimulation of mPFC-BLA connections. Altogether, we presented a series of evidence to delineate circuit events in the amygdala associated with the accumulation of α-Syn inclusions in the mouse brain, highlighting that functional impairment of the amygdala is sufficient to cause social behavior deficits. The present work further suggests that early circuit modulation could be an effective approach to alleviate symptoms associated with α-Syn pathology, necessitating studies of functional consequences of α-Syn aggregation.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503, USA; Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington DC 20007, USA
| | - Samuel Daniels
- Department of Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Vijay Singh
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Marissa Menard
- Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20852, United States
| | | | - Hong-Yuan Chu
- Department of Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI 49503, USA; Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington DC 20007, USA.
| |
Collapse
|
8
|
Mills KA, Phillips O, Mahajan A. Hope vs. Hype I: Spreading alpha-synuclein explains cognitive deficits in Parkinson disease. Parkinsonism Relat Disord 2024; 126:106042. [PMID: 38365523 DOI: 10.1016/j.parkreldis.2024.106042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/18/2024]
Abstract
The Parkinson Study Group (PSG) gathered North American experts in Parkinson disease during the 9th Annual Symposium on "Shaping the Management of Parkinson Disease: Debating Current Controversies". Debaters were tasked with agree or disagree positions to a particular prompt. This is the first in three-part series of "Hype vs. Hope" debates involving current trends and advances in Parkinson disease. With the prompt of "Spreading alpha-synuclein explains cognitive deficits in Parkinson disease," Dr. Kelly Mills, MD, MHS was tasked with the "agree" stance and Dr. Abhimanyu Mahajan, MD, MHS was tasked with the "disagree" stance. The following point-of-view article is an adaptation of this debate.
Collapse
Affiliation(s)
- Kelly A Mills
- Johns Hopkins Department of Neurology, Baltimore, MD600 N. Wolfe Street, Meyer 6-181D, 21287, United States.
| | - Oliver Phillips
- Geisel School of Medicine at Dartmouth, Hanover, NH, 18 Old Etna Road, 03756, United States.
| | - Abhimanyu Mahajan
- The University of Cincinnati James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, OH, 260 Stetson St., Suite 2300, Cincinnati, 45219, United States.
| |
Collapse
|
9
|
Carli S, Brugnano L, Caligiore D. Simulating combined monoaminergic depletions in a PD animal model through a bio-constrained differential equations system. Front Comput Neurosci 2024; 18:1386841. [PMID: 39247252 PMCID: PMC11378529 DOI: 10.3389/fncom.2024.1386841] [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: 02/16/2024] [Accepted: 07/31/2024] [Indexed: 09/10/2024] Open
Abstract
Introduction Historically, Parkinson's Disease (PD) research has focused on the dysfunction of dopamine-producing cells in the substantia nigra pars compacta, which is linked to motor regulation in the basal ganglia. Therapies have mainly aimed at restoring dopamine (DA) levels, showing effectiveness but variable outcomes and side effects. Recent evidence indicates that PD complexity implicates disruptions in DA, noradrenaline (NA), and serotonin (5-HT) systems, which may underlie the variations in therapy effects. Methods We present a system-level bio-constrained computational model that comprehensively investigates the dynamic interactions between these neurotransmitter systems. The model was designed to replicate experimental data demonstrating the impact of NA and 5-HT depletion in a PD animal model, providing insights into the causal relationships between basal ganglia regions and neuromodulator release areas. Results The model successfully replicates experimental data and generates predictions regarding changes in unexplored brain regions, suggesting avenues for further investigation. It highlights the potential efficacy of alternative treatments targeting the locus coeruleus and dorsal raphe nucleus, though these preliminary findings require further validation. Sensitivity analysis identifies critical model parameters, offering insights into key factors influencing brain area activity. A stability analysis underscores the robustness of our mathematical formulation, bolstering the model validity. Discussion Our holistic approach emphasizes that PD is a multifactorial disorder and opens promising avenues for early diagnostic tools that harness the intricate interactions among monoaminergic systems. Investigating NA and 5-HT systems alongside the DA system may yield more effective, subtype-specific therapies. The exploration of multisystem dysregulation in PD is poised to revolutionize our understanding and management of this complex neurodegenerative disorder.
Collapse
Affiliation(s)
- Samuele Carli
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council (CTNLab-ISTC-CNR), Rome, Italy
- Entersys s.r.l., Padua, Italy
- AI2Life s.r.l., Innovative Start-Up, ISTC-CNR Spin-Off, Rome, Italy
- Department of Mathematics and Computer Science "U. Dini", University of Florence, Florence, Italy
| | - Luigi Brugnano
- Department of Mathematics and Computer Science "U. Dini", University of Florence, Florence, Italy
| | - Daniele Caligiore
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council (CTNLab-ISTC-CNR), Rome, Italy
- AI2Life s.r.l., Innovative Start-Up, ISTC-CNR Spin-Off, Rome, Italy
| |
Collapse
|
10
|
Chen L, Chehade HD, Chu HY. Motor Cortical Neuronal Hyperexcitability Associated with α-Synuclein Aggregation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604995. [PMID: 39091827 PMCID: PMC11291145 DOI: 10.1101/2024.07.24.604995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Dysfunction of the cerebral cortex is thought to underlie motor and cognitive impairments in Parkinson disease (PD). While cortical function is known to be suppressed by abnormal basal ganglia output following dopaminergic degeneration, it remains to be determined how the deposition of Lewy pathology disrupts cortical circuit integrity and function. Moreover, it is also unknown whether cortical Lewy pathology and midbrain dopaminergic degeneration interact to disrupt cortical function in late-stage. To begin to address these questions, we injected α-synuclein (αSyn) preformed fibrils (PFFs) into the dorsolateral striatum of mice to seed αSyn pathology in the cortical cortex and induce degeneration of midbrain dopaminergic neurons. Using this model system, we reported that αSyn aggregates accumulate in the motor cortex in a layer- and cell-subtype-specific pattern. Particularly, intratelencephalic neurons (ITNs) showed earlier accumulation and greater extent of αSyn aggregates relative to corticospinal neurons (CSNs). Moreover, we demonstrated that the intrinsic excitability and inputs resistance of αSyn aggregates-bearing ITNs in the secondary motor cortex (M2) are increased, along with a noticeable shrinkage of cell bodies and loss of dendritic spines. Last, neither the intrinsic excitability of CSNs nor their thalamocortical input was altered by a partial striatal dopamine depletion associated with αSyn pathology. Our results documented motor cortical neuronal hyperexcitability associated with αSyn aggregation and provided a novel mechanistic understanding of cortical circuit dysfunction in PD.
Collapse
Affiliation(s)
- Liqiang Chen
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, United States
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington DC, 20007, United States
| | - Hiba Douja Chehade
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, United States
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington DC, 20007, United States
| | - Hong-Yuan Chu
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, 20852, United States
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, United States
- Department of Pharmacology and Physiology, Georgetown University of Medical Center, Washington DC, 20007, United States
| |
Collapse
|
11
|
Negi S, Khurana N, Duggal N. The misfolding mystery: α-synuclein and the pathogenesis of Parkinson's disease. Neurochem Int 2024; 177:105760. [PMID: 38723900 DOI: 10.1016/j.neuint.2024.105760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024]
Abstract
Neurodegenerative diseases such as Parkinson's disease (PD) are characterized by the death of neurons in specific areas of the brain. One of the proteins that is involved in the pathogenesis of PD is α-synuclein (α-syn). α-Syn is a normal protein that is found in all neurons, but in PD, it misfolds and aggregates into toxic fibrils. These fibrils can then coalesce into pathological inclusions, such as Lewy bodies and Lewy neurites. The pathogenic pathway of PD is thought to involve a number of steps, including misfolding and aggregation of α-syn, mitochondrial dysfunction, protein clearance impairment, neuroinflammation and oxidative stress. A deeper insight into the structure of α-syn and its fibrils could aid in understanding the disease's etiology. The prion-like nature of α-syn is also an important area of research. Prions are misfolded proteins that can spread from cell to cell, causing other proteins to misfold as well. It is possible that α-syn may behave in a similar way, spreading from cell to cell and causing a cascade of misfolding and aggregation. Various post-translational alterations have also been observed to play a role in the pathogenesis of PD. These alterations can involve a variety of nuclear and extranuclear activities, and they can lead to the misfolding and aggregation of α-syn. A better understanding of the pathogenic pathway of PD could lead to the development of new therapies for the treatment of this disease.
Collapse
Affiliation(s)
- Samir Negi
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi, G.T. Road, Phagwara, Punjab, 144411, India
| | - Navneet Khurana
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi, G.T. Road, Phagwara, Punjab, 144411, India
| | - Navneet Duggal
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi, G.T. Road, Phagwara, Punjab, 144411, India.
| |
Collapse
|
12
|
Meccariello R, Bellenchi GC, Pulcrano S, D’Addario SL, Tafuri D, Mercuri NB, Guatteo E. Neuronal dysfunction and gene modulation by non-coding RNA in Parkinson's disease and synucleinopathies. Front Cell Neurosci 2024; 17:1328269. [PMID: 38249528 PMCID: PMC10796818 DOI: 10.3389/fncel.2023.1328269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/07/2023] [Indexed: 01/23/2024] Open
Abstract
Over the last few decades, emerging evidence suggests that non-coding RNAs (ncRNAs) including long-non-coding RNA (lncRNA), microRNA (miRNA) and circular-RNA (circRNA) contribute to the molecular events underlying progressive neuronal degeneration, and a plethora of ncRNAs have been identified significantly misregulated in many neurodegenerative diseases, including Parkinson's disease and synucleinopathy. Although a direct link between neuropathology and causative candidates has not been clearly established in many cases, the contribution of ncRNAs to the molecular processes leading to cellular dysfunction observed in neurodegenerative diseases has been addressed, suggesting that they may play a role in the pathophysiology of these diseases. Aim of the present Review is to overview and discuss recent literature focused on the role of RNA-based mechanisms involved in different aspects of neuronal pathology in Parkinson's disease and synucleinopathy models.
Collapse
Affiliation(s)
- Rosaria Meccariello
- Department of Medical and Movement Sciences and Wellness, University of Naples Parthenope, Naples, Italy
| | - Gian Carlo Bellenchi
- Institute of Genetics and Biophysics, CNR, Naples, Italy
- Experimental Neurology Laboratory, Santa Lucia Foundation IRCCS, Rome, Italy
| | | | - Sebastian Luca D’Addario
- Experimental Neurology Laboratory, Santa Lucia Foundation IRCCS, Rome, Italy
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, CNR, Rome, Italy
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Domenico Tafuri
- Department of Medical and Movement Sciences and Wellness, University of Naples Parthenope, Naples, Italy
| | - Nicola B. Mercuri
- Experimental Neurology Laboratory, Santa Lucia Foundation IRCCS, Rome, Italy
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Ezia Guatteo
- Department of Medical and Movement Sciences and Wellness, University of Naples Parthenope, Naples, Italy
- Experimental Neurology Laboratory, Santa Lucia Foundation IRCCS, Rome, Italy
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| |
Collapse
|
13
|
Pulcrano S, De Gregorio R, De Sanctis C, Volpicelli F, Piscitelli RM, Speranza L, Perrone-Capano C, di Porzio U, Caiazzo M, Martini A, Giacomet C, Medina D, Awatramani R, Viggiano D, Federici M, Mercuri NB, Guatteo E, Bellenchi GC. miR-218 Promotes Dopaminergic Differentiation and Controls Neuron Excitability and Neurotransmitter Release through the Regulation of a Synaptic-Related Genes Network. J Neurosci 2023; 43:8104-8125. [PMID: 37816598 PMCID: PMC10697421 DOI: 10.1523/jneurosci.0431-23.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/25/2023] [Accepted: 08/10/2023] [Indexed: 10/12/2023] Open
Abstract
In the brain, microRNAs (miRNAs) are believed to play a role in orchestrating synaptic plasticity at a higher level by acting as an additional mechanism of translational regulation, alongside the mRNA/polysome system. Despite extensive research, our understanding of the specific contribution of individual miRNA to the function of dopaminergic neurons (DAn) remains limited. By performing a dopaminergic-specific miRNA screening, we have identified miR-218 as a critical regulator of DAn activity in male and female mice. We have found that miR-218 is specifically expressed in mesencephalic DAn and is able to promote dopaminergic differentiation of embryonic stem cells and functional maturation of transdifferentiated induced DA neurons. Midbrain-specific deletion of both genes encoding for miR-218 (referred to as miR-218-1 and mir218-2) affects the expression of a cluster of synaptic-related mRNAs and alters the intrinsic excitability of DAn, as it increases instantaneous frequencies of evoked action potentials, reduces rheobase current, affects the ionic current underlying the action potential after hyperpolarization phase, and reduces dopamine efflux in response to a single electrical stimulus. Our findings provide a comprehensive understanding of the involvement of miR-218 in the dopaminergic system and highlight its role as a modulator of dopaminergic transmission.SIGNIFICANCE STATEMENT In the past decade, several miRNAs have emerged as potential regulators of synapse activity through the modulation of specific gene expression. Among these, we have identified a dopaminergic-specific miRNA, miR-218, which is able to promote dopaminergic differentiation and regulates the translation of an entire cluster of synapse related mRNAs. Deletion of miR-218 has notable effects on dopamine release and alters the intrinsic excitability of dopaminergic neurons, indicating a direct control of dopaminergic activity by miR-218.
Collapse
Affiliation(s)
- Salvatore Pulcrano
- Institute of Genetics and Biophysics, Consiglio Nazionale delle Ricerche, Naples, 80131, Italy
| | - Roberto De Gregorio
- Institute of Genetics and Biophysics, Consiglio Nazionale delle Ricerche, Naples, 80131, Italy
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine and New York Presbyterian, New York, New York 10021
| | - Claudia De Sanctis
- Institute of Genetics and Biophysics, Consiglio Nazionale delle Ricerche, Naples, 80131, Italy
- Neuropathology Brain Bank at Mount Sinai, New York, New York 10029
| | - Floriana Volpicelli
- Institute of Genetics and Biophysics, Consiglio Nazionale delle Ricerche, Naples, 80131, Italy
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, 80131, Italy
| | - Rosa Maria Piscitelli
- Fondazione Santa Lucia Istituto Di Ricovero e Cura a Carattere Scientifico, Rome, 00143, Italy
| | - Luisa Speranza
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, New York 10461
| | - Carla Perrone-Capano
- Institute of Genetics and Biophysics, Consiglio Nazionale delle Ricerche, Naples, 80131, Italy
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, 80131, Italy
| | - Umberto di Porzio
- Institute of Genetics and Biophysics, Consiglio Nazionale delle Ricerche, Naples, 80131, Italy
| | - Massimiliano Caiazzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, 80131, Italy
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Alessandro Martini
- Fondazione Santa Lucia Istituto Di Ricovero e Cura a Carattere Scientifico, Rome, 00143, Italy
| | - Cecilia Giacomet
- Fondazione Santa Lucia Istituto Di Ricovero e Cura a Carattere Scientifico, Rome, 00143, Italy
| | - Diego Medina
- Telethon Institute of Genetics and Medicine, Pozzuoli, 80078, Italy
- Department of Medical and Translational Science, Federico II University, Naples, 80131, Italy
| | | | - Davide Viggiano
- Department of Translational Medical Sciences, University of Campania "L. Vanvitelli," Naples, 80131, Italy
| | - Mauro Federici
- Fondazione Santa Lucia Istituto Di Ricovero e Cura a Carattere Scientifico, Rome, 00143, Italy
| | - Nicola B Mercuri
- Fondazione Santa Lucia Istituto Di Ricovero e Cura a Carattere Scientifico, Rome, 00143, Italy
- University of Tor Vergata, Department of Systems Medicine, Rome, 00133, Italy
| | - Ezia Guatteo
- Fondazione Santa Lucia Istituto Di Ricovero e Cura a Carattere Scientifico, Rome, 00143, Italy
- Department of Motor Science and Wellness, Parthenope University, Naples, 80133, Italy
| | - Gian Carlo Bellenchi
- Institute of Genetics and Biophysics, Consiglio Nazionale delle Ricerche, Naples, 80131, Italy
- Fondazione Santa Lucia Istituto Di Ricovero e Cura a Carattere Scientifico, Rome, 00143, Italy
| |
Collapse
|
14
|
Mazzeo F, Meccariello R, Guatteo E. Molecular and Epigenetic Aspects of Opioid Receptors in Drug Addiction and Pain Management in Sport. Int J Mol Sci 2023; 24:ijms24097831. [PMID: 37175536 PMCID: PMC10178540 DOI: 10.3390/ijms24097831] [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: 03/31/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Opioids are substances derived from opium (natural opioids). In its raw state, opium is a gummy latex extracted from Papaver somniferum. The use of opioids and their negative health consequences among people who use drugs have been studied. Today, opioids are still the most commonly used and effective analgesic treatments for severe pain, but their use and abuse causes detrimental side effects for health, including addiction, thus impacting the user's quality of life and causing overdose. The mesocorticolimbic dopaminergic circuitry represents the brain circuit mediating both natural rewards and the rewarding aspects of nearly all drugs of abuse, including opioids. Hence, understanding how opioids affect the function of dopaminergic circuitry may be useful for better knowledge of the process and to develop effective therapeutic strategies in addiction. The aim of this review was to summarize the main features of opioids and opioid receptors and focus on the molecular and upcoming epigenetic mechanisms leading to opioid addiction. Since synthetic opioids can be effective for pain management, their ability to induce addiction in athletes, with the risk of incurring doping, is also discussed.
Collapse
Affiliation(s)
- Filomena Mazzeo
- Department of Economics, Law, Cybersecurity and Sports Sciences, University of Naples "Parthenope", 80133 Naples, Italy
- Department of Movement Sciences and Wellbeing, University of Naples "Parthenope", 80133 Naples, Italy
| | - Rosaria Meccariello
- Department of Movement Sciences and Wellbeing, University of Naples "Parthenope", 80133 Naples, Italy
| | - Ezia Guatteo
- Department of Movement Sciences and Wellbeing, University of Naples "Parthenope", 80133 Naples, Italy
- IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy
| |
Collapse
|