1
|
Zamanian MY, Kamran Z, Tavakoli MR, Oghenemaro EF, Abohassan M, Kubaev A, Nathiya D, Kaur P, Zwamel AH, Abdulamer RS. The Role of ΔFosB in the Pathogenesis of Levodopa-Induced Dyskinesia: Mechanisms and Therapeutic Strategies. Mol Neurobiol 2025; 62:7393-7412. [PMID: 39890697 DOI: 10.1007/s12035-025-04720-z] [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: 09/13/2024] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Levodopa-induced dyskinesia (LID) represents a significant complication associated with the long-term administration of levodopa (L-DOPA) for the treatment of Parkinson's disease (PD). This review examines the critical role of ΔFosB, a transcription factor, in the pathogenesis of LID and explores potential therapeutic interventions. ΔFosB accumulates within the striatum in response to chronic dopaminergic stimulation, thereby driving maladaptive changes that culminate in dyskinesia. Its persistent expression modifies gene transcription, influencing neuronal plasticity and contributing to the sustained presence of dyskinetic movements. This study explains how ΔFosB functions at the molecular level, focusing on its connections with dopamine D1 receptors, the cAMP/PKA signaling pathway, and its regulatory effects on downstream targets such as DARPP-32 and GluA1 AMPA receptor subunits. Additionally, it examines how neuronal nitric oxide synthase (nNOS) affects ΔFosB levels and the development of LID. This review also considers the interactions between ΔFosB and other signaling pathways, such as ERK and mTOR, in the context of LID and striatal plasticity. Emerging therapeutic strategies targeting ΔFosB and its associated pathways include pharmacological interventions like ranitidine, 5-hydroxytryptophan, and carnosic acid. Furthermore, this study addresses the role of JunD, another component of the AP-1 transcription factor complex, in the pathogenesis of LID. Understanding the molecular mechanisms by which ΔFosB contributes to LID offers promising avenues for developing novel treatments that could mitigate dyskinesia and improve the quality of life for PD patients undergoing long-term L-DOPA therapy.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
| | - Zahra Kamran
- Department of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marziye Ranjbar Tavakoli
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology & Biotechnology, Faculty of Pharmacy, Delta State University, Abraka, Nigeria
| | - Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan
| | - Deepak Nathiya
- Department of Pharmacy Practice, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Ahmed Hussein Zwamel
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Resan Shakir Abdulamer
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq
| |
Collapse
|
2
|
Dubljević O, Pavković Ž, Srbovan M, Potrebić M, Stanojlović M, Pešić V. Attention-deficit/hyperactivity disorder-related psychomotor activity and altered neuronal activity in the medial prefrontal cortex and striatum in the A53T mouse model of Parkinson's disease and other synucleinopathies: Findings from an "endophenotype" approach. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111273. [PMID: 39870135 DOI: 10.1016/j.pnpbp.2025.111273] [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: 09/20/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 01/29/2025]
Abstract
Attention-Deficit/Hyperactivity Disorder (ADHD) is associated with an increased risk of Parkinson's disease (PD) and other synucleinopathies later in life. The severity of the ADHD phenotype may play a significant role in this association. There is no indication that any of the existing animal models can unify these disorders. Using the Open Field Test, amphetamine-challenge test, Western blot and immunohistochemical analysis of neuronal activity markers (c-Fos, FosB and ΔFosB) we performed a deliberate neurobehavioral characterization of 6-month-old hemizygous A53T carriers (A53T+) of the JAX006823 strain, evaluating the utility of this transgenic mouse model of PD and other synucleinopathies in ADHD/PD continuum research. Adhering to the "endophenotype" approach, non-transgenic littermates (A53T-) and C57BL/6J mice (used to maintain the colony) were examined with A53T+ mice, to differentiate between biomarkers of transgenicity and endophenotypic traits related to the genetic background of the strain. Obtained results revealed that increased behavioral and acute striatal response to novelty, increased basal neuronal activity of the ventromedial prefrontal cortex and rate-dependent calming effect of amphetamine were endophenotypic characteristics of the strain. Increased acute response of the medial prefrontal cortex to novelty and chronic increase in neuronal activity of the striatum appeared as the mark of transgenicity. To the best of our knowledge, this is the first study to indicate external validity of a transgenic mouse model of PD and other synucleinopathies with the neurobehavioral pathology associated with ADHD, hinting at its potential in preclinical research of ADHD/PD continuum. The full capacity of the model remains to be explored.
Collapse
Affiliation(s)
- Olga Dubljević
- Laboratory of Molecular Neurobiology and Behavior, Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Željko Pavković
- Laboratory of Molecular Neurobiology and Behavior, Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Maja Srbovan
- Laboratory of Molecular Neurobiology and Behavior, Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milica Potrebić
- Laboratory of Molecular Neurobiology and Behavior, Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Miloš Stanojlović
- Laboratory of Molecular Neurobiology and Behavior, Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia; Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Vesna Pešić
- Laboratory of Molecular Neurobiology and Behavior, Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
3
|
Jafri S, Ghani M, Stickle N, Virtanen C, Hazrati LN, Visanji NP. Translational profiling reveals novel gene expression changes in the direct and indirect pathways in a mouse model of levodopa induced dyskinesia. Front Cell Neurosci 2025; 18:1477511. [PMID: 40144773 PMCID: PMC11936753 DOI: 10.3389/fncel.2024.1477511] [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: 08/07/2024] [Accepted: 11/18/2024] [Indexed: 03/28/2025] Open
Abstract
Introduction The molecular mechanisms underlying L-dihydroxyphenylalanine (LDOPA) induced dyskinesia in Parkinson's disease are poorly understood. Here we employ two transgenic mouse lines, combining translating ribosomal affinity purification (TRAP) with bacterial artificial chromosome expression (Bac), to selectively isolate RNA from either DRD1A expressing striatonigral, or DRD2 expressing striatopallidal medium spiny neurons (MSNs) of the direct and indirect pathways respectively, to study changes in translational gene expression following repeated LDOPA treatment. Methods 6-OHDA lesioned DRD1A and DRD2 BacTRAP mice were treated with either saline or LDOPA bi-daily for 21 days over which time they developed abnormal involuntary movements reminiscent of dyskinesia. On day 22, all animals received LDOPA 40min prior to sacrifice. The striatum of the lesioned hemisphere was dissected and subject to TRAP. Extracted ribosomal RNA was amplified, purified, and gene expression was quantified using microarray. Results One hundred ninety-five significantly varying transcripts were identified among the four treatment groups. Pathway analysis revealed an overrepresentation of calcium signaling and long-term potentiation in the DRD1A expressing MSNs of the direct pathway, with significant involvement of long-term depression in the DRD2 expressing MSNs of the indirect pathway following chronic treatment with LDOPA. Several MAPK associated genes (NR4A1, GADD45G, STMN1, FOS, and DUSP1) differentiated the direct and indirect pathways following both acute and chronic LDOPA treatment. However, the MAPK pathway activator PAK1 was downregulated in the indirect pathway and upregulated in the direct pathway, strongly suggesting a role for PAK1 in regulating the opposing effects of LDOPA on these two pathways in dyskinesia. Discussion Future studies will assess the potential of targeting these genes and pathways to prevent the development of LDOPA-induced dyskinesia.
Collapse
Affiliation(s)
- Sabika Jafri
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Mahdi Ghani
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Krembil Discovery Tower, Toronto, ON, Canada
| | - Natalie Stickle
- University Health Network Microarray Centre, Toronto Medical Discovery Tower, Toronto, ON, Canada
| | - Carl Virtanen
- University Health Network Microarray Centre, Toronto Medical Discovery Tower, Toronto, ON, Canada
| | - Lili-Naz Hazrati
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Naomi P. Visanji
- Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Krembil Discovery Tower, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Ahmed MR, Inayathullah M, Morton M, Pothineni VR, Kim K, Ahmed MS, Babar MM, Rajadas J. Intranasal delivery of liposome encapsulated flavonoids ameliorates l-DOPA induced dyskinesia in hemiparkinsonian mice. Biomaterials 2024; 311:122680. [PMID: 38959534 DOI: 10.1016/j.biomaterials.2024.122680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/25/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024]
Abstract
In the present study, we explored the development of a novel noninvasive liposomal drug delivery material for use in intranasal drug delivery applications in human diseases. We used drug entrapment into liposomal nanoparticle assembly to efficiently deliver the drugs to the nasal mucosa to be delivered to the brain. The naturally occurring flavonoid 7,8-dihydroxyflavone (7,8-DHF) has previously been shown to have beneficial effects in ameliorating Parkinson's disease (PD). We used both naturally occurring 7,8-DHF and the chemically modified form of DHF, the DHF-ME, to be used as a drug candidate for the treatment of PD and l-DOPA induced dyskinesia (LID), which is the debilitating side effect of l-DOPA therapy in PD. The ligand-protein interaction behavior for 7,8-DHF and 6,7-DHF-ME was found to be more effective with molecular docking and molecular stimulation studies of flavonoid compounds with TrkB receptor. Our study showed that 7,8-DHF delivered via intranasal route using a liposomal formulation ameliorated LID in hemiparkinsonian mice model when these mice were chronically administered with l-DOPA, which is the only current medication for relieving the clinical symptoms of PD. The present study also demonstrated that apart from reducing the LID, 7,8-DHF delivery directly to the brain via the intranasal route also corrected some long-term signaling adaptations involving ΔFosB and α Synuclein in the brain of dopamine (DA) depleted animals.
Collapse
Affiliation(s)
- Mohamed Rafiuddin Ahmed
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA
| | - Mohammed Inayathullah
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA
| | - Mithya Morton
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA; Children's Hospital of Orange County - UC Irvine School of Medicine, Department of Pediatrics, 505 S. Main St., Suite #525, Orange, CA, 92868, USA
| | - Venkata Raveendra Pothineni
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA
| | - Kwangmin Kim
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA; Department of Physiology, Gachon University College of Medicine, Incheon, 21999, South Korea
| | - Mohamed Sohail Ahmed
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, MCN 1161 21st Ave S. Nashville TN 37232, USA
| | - Mustafeez Mujtaba Babar
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA
| | - Jayakumar Rajadas
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA.
| |
Collapse
|
5
|
Kambey PA, Wu J, Liu W, Su M, Buberwa W, Tang C. Targeting serum response factor (SRF) deactivates ΔFosB and mitigates Levodopa-induced dyskinesia in a mouse model of Parkinson's disease. Gene Ther 2024; 31:614-624. [PMID: 39384937 DOI: 10.1038/s41434-024-00492-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024]
Abstract
L-3,4-dihydroxyphenylalanine (L-DOPA) is currently the preferred treatment for Parkinson's Disease (PD) and is considered the gold standard. However, prolonged use of L-DOPA in patients can result in involuntary movements known as Levodopa-induced dyskinesia (LID), which includes uncontrollable dystonia affecting the trunk, limbs, and face. The role of ΔFosB protein, a truncated splice variant of the FosB gene, in LID has been acknowledged, but its underlying mechanism has remained elusive. Here, using a mouse model of Parkinson's disease treated with chronic levodopa we demonstrate that serum response factor (SRF) binds to the FosB promoter, thereby activating FosB expression and levodopa induced-dyskinetic movements. Western blot analysis demonstrates a significant increase in SRF expression in the dyskinetic group compared to the control group. Knocking down SRF significantly reduced abnormal involuntary movements (AIMS) and ΔFosB expression compared to the control. Conversely, overexpression of SRF led to an increase in ΔFosB expression and worsened levodopa-induced dyskinesia. To shed light on the regulatory role of the Akt signaling pathway in this phenomenon, we administered the Akt agonist SC79 to PD mouse models via intraperitoneal injection, followed by L-DOPA administration. The expression of SRF, ΔFosB, and phosphorylated Akt (p-Akt) significantly increased in this group compared to the group receiving normal saline to signify that these happen through Akt signaling pathway. Collectively, our findings identify a promising therapeutic target for addressing levodopa-induced dyskinesia.
Collapse
Affiliation(s)
- Piniel Alphayo Kambey
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
- Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou Science Park, Huangpu District, Guangzhou, China.
| | - Jiao Wu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - WenYa Liu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Mingyu Su
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Wokuheleza Buberwa
- Department of Neurology, The second affiliated hospital of Xi'an Jiaotong University, 710049, Xi'an, China
| | - Chuanxi Tang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
6
|
Roman KM, Dinasarapu AR, Cherian S, Fan X, Donsante Y, Aravind N, Chan CS, Jinnah H, Hess EJ. Striatal cell-type-specific molecular signatures reveal therapeutic targets in a model of dystonia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617010. [PMID: 39415987 PMCID: PMC11482807 DOI: 10.1101/2024.10.07.617010] [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: 10/19/2024]
Abstract
Striatal dysfunction is implicated in many forms of dystonia, including idiopathic, inherited and iatrogenic dystonias. The striatum is comprised largely of GABAergic spiny projection neurons (SPNs) that are defined by their long-range efferents. Direct SPNs (dSPNs) project to the internal globus pallidus/substantia nigra reticulata whereas indirect pathway SPNs (iSPNs) project to the external pallidum; the concerted activity of both SPN subtypes modulates movement. Convergent results from genetic, imaging and physiological studies in patients suggest that abnormalities of both dSPNs and iSPNs contribute to the expression of dystonia, but the molecular adaptations underlying these abnormalities are not known. Here we provide a comprehensive analysis of SPN cell-type-specific molecular signatures in a model of DOPA-responsive dystonia (DRD mice), which is caused by gene defects that reduce dopamine neurotransmission, resulting in dystonia that is specifically associated with striatal dysfunction. Individually profiling the translatome of dSPNs and iSPNs using translating ribosome affinity purification with RNA-seq revealed hundreds of differentially translating mRNAs in each SPN subtype in DRD mice, yet there was little overlap between the dysregulated genes in dSPNs and iSPNs. Despite the paucity of shared adaptations, a disruption in glutamatergic signaling was predicted for both dSPNs and iSPNs. Indeed, we found that both AMPA and NMDA receptor-mediated currents were enhanced in dSPNs but diminished in iSPNs in DRD mice. The pattern of mRNA dysregulation was specific to dystonia as the adaptations in DRD mice were distinct from those in parkinsonian mice where the dopamine deficit occurs in adults, suggesting that the phenotypic outcome is dependent on both the timing of the dopaminergic deficit and the SPN-specific adaptions. We leveraged the unique molecular signatures of dSPNs and iSPNs in DRD mice to identify biochemical mechanisms that may be targets for therapeutics, including LRRK2 inhibition. Administration of the LRRK2 inhibitor MLi-2 ameliorated the dystonia in DRD mice suggesting a novel target for therapeutics and demonstrating that the delineation of cell-type-specific molecular signatures provides a powerful approach to revealing both CNS dysfunction and therapeutic targets in dystonia.
Collapse
Affiliation(s)
- Kaitlyn M. Roman
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | | | - Suraj Cherian
- Department of Neuroscience, Northwestern University, Chicago, Illinois, USA
| | - Xueliang Fan
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Yuping Donsante
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Nivetha Aravind
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - C. Savio Chan
- Department of Neuroscience, Northwestern University, Chicago, Illinois, USA
| | - H.A. Jinnah
- Department of Neurology, Emory University, Atlanta, Georgia, USA
- Department of Human Genetics, Emory University, Atlanta, Georgia, USA
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - Ellen J. Hess
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
- Department of Neurology, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
7
|
Liu K, Zhang Z, Xu Y, Wu Y, Lian P, Ma Z, Tang Z, Zhang X, Yang X, Zhai H, Zhang L, Xu Y, Cao X. AMPK-mediated autophagy pathway activation promotes ΔFosB degradation to improve levodopa-induced dyskinesia. Cell Signal 2024; 118:111125. [PMID: 38432574 DOI: 10.1016/j.cellsig.2024.111125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/16/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Parkinson's disease patients on chronic levodopa often suffer from motor complications, which tend to reduce their quality of life. Levodopa-induced dyskinesia (LID) is one of the most prevalent motor complications, often characterized by abnormal involuntary movements, and the pathogenesis of LID is still unclear but recent studies have suggested the involvement of autophagy. METHODS The onset of LID was mimicked by chronic levodopa treatment in a unilateral 6-hydroxydopamine (6-OHDA) -lesion rat model. Overexpression of ΔFosB in HEK293 cells to mimic the state of ΔFosB accumulation. The modulation of the AMP-activated protein kinase (AMPK)-mediated autophagy pathway using by metformin, AICAR (an AMPK activator), Compound C (an AMPK inhibitor) and chloroquine (an autophagy pathway inhibitor). The severity of LID was assessed by axial, limb, and orofacial (ALO) abnormal involuntary movements (AIMs) score and in vivo electrophysiology. The activity of AMPK pathway as well as autophagy markers and FosB-ΔFosB levels were detected by western blotting. RT-qPCR was performed to detect the transcription level of FosB-ΔFosB. The mechanism of autophagy dysfunction was further explored by immunofluorescence and transmission electron microscopy. RESULTS In vivo experiments demonstrated that chronic levodopa treatment reduced AMPK phosphorylation, impaired autophagosome-lysosomal fusion and caused FosB-ΔFosB accumulation in the striatum of PD rats. Long-term metformin intervention improved ALO AIMs scores as well as reduced the mean power of high gamma (hγ) oscillations and the proportion of striatal projection neurons unstable in response to dopamine for LID rats. Moreover, the intervention of metformin promoted AMPK phosphorylation, ameliorated the impairment of autophagosome-lysosomal fusion, thus, promoting FosB-ΔFosB degradation to attenuate its accumulation in the striatum of LID rats. However, the aforementioned roles of metformin were reversed by Compound C and chloroquine. The results of in vitro studies demonstrated the ability of metformin and AICAR to attenuate ΔFosB levels by promoting its degradation, while Compound C and chloroquine could block this effect. CONCLUSIONS In conclusion, our results suggest that long-term metformin treatment could promote ΔFosB degradation and thus attenuate the development of LID through activating the AMPK-mediated autophagy pathway. Overall, our results support the AMPK-mediated autophagy pathway as a novel therapeutic target for LID and also indicate that metformin is a promising therapeutic candidate for LID.
Collapse
Affiliation(s)
- Ke Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhaoyuan Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yu Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Piaopiao Lian
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhuoran Ma
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhicheng Tang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoqian Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoman Yang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Heng Zhai
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lei Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
8
|
Cirillo R, Duperrier S, Parekh P, Millot M, Li Q, Thiolat ML, Morelli M, Xie J, Le Bars D, Redouté J, Bezard E, Sgambato V. Striatal Serotonin 4 Receptor is Increased in Experimental Parkinsonism and Dyskinesia. JOURNAL OF PARKINSON'S DISEASE 2024; 14:261-267. [PMID: 38339940 PMCID: PMC10977406 DOI: 10.3233/jpd-230331] [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: 01/07/2024] [Indexed: 02/12/2024]
Abstract
Alterations of serotonin type 4 receptor levels are linked to mood disorders and cognitive deficits in several conditions. However, few studies have investigated 5-HT4R alterations in movement disorders. We wondered whether striatal 5-HT4R expression is altered in experimental parkinsonism. We used a brain bank tissue from a rat and a macaque model of Parkinson's disease (PD). We then investigated its in vivo PET imaging regulation in a cohort of macaques. Dopaminergic depletion increases striatal 5-HT4R in the two models, further augmented after dyskinesia-inducing L-Dopa. Pending confirmation in PD patients, the 5-HT4R might offer a therapeutic target for dampening PD's symptoms.
Collapse
Affiliation(s)
- Rossella Cirillo
- Institut des Sciences Cognitives Marc Jeannerod (ISCMJ), Unité Mixte de Recherche 5229 du Centre National de la Recherche Scientifique (CNRS), Bron, France
- Universitè de Lyon 1, Lyon, France
| | - Sandra Duperrier
- Institut des Sciences Cognitives Marc Jeannerod (ISCMJ), Unité Mixte de Recherche 5229 du Centre National de la Recherche Scientifique (CNRS), Bron, France
- Universitè de Lyon 1, Lyon, France
| | - Pathik Parekh
- Institut des Sciences Cognitives Marc Jeannerod (ISCMJ), Unité Mixte de Recherche 5229 du Centre National de la Recherche Scientifique (CNRS), Bron, France
- Universitè de Lyon 1, Lyon, France
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Monserrato, Italy
| | - Mathilde Millot
- Institut des Sciences Cognitives Marc Jeannerod (ISCMJ), Unité Mixte de Recherche 5229 du Centre National de la Recherche Scientifique (CNRS), Bron, France
- Universitè de Lyon 1, Lyon, France
| | - Qin Li
- Motac Beijing Services, Beijing, China
| | - Marie-Laure Thiolat
- Universitè de Bordeaux, Institut des Maladies Neurodégénératives, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, Bordeaux, France
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Monserrato, Italy
| | - Jing Xie
- Institut des Sciences Cognitives Marc Jeannerod (ISCMJ), Unité Mixte de Recherche 5229 du Centre National de la Recherche Scientifique (CNRS), Bron, France
- Institut du Vieillissement, Centrede Recherche Clinique Vieillissement Cerveau Fragilité, Hôpital des Charpennes, Villeurbanne, France
| | | | | | - Erwan Bezard
- Motac Beijing Services, Beijing, China
- Universitè de Bordeaux, Institut des Maladies Neurodégénératives, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, Bordeaux, France
| | - Véronique Sgambato
- Institut des Sciences Cognitives Marc Jeannerod (ISCMJ), Unité Mixte de Recherche 5229 du Centre National de la Recherche Scientifique (CNRS), Bron, France
- Universitè de Lyon 1, Lyon, France
| |
Collapse
|
9
|
Wang Q, Wang H, Meng W, Liu C, Li R, Zhang M, Liang K, Gao Y, Du T, Zhang J, Han C, Shi L, Meng F. The NONRATT023402.2/rno-miR-3065-5p/NGFR axis affects levodopa-induced dyskinesia in a rat model of Parkinson's disease. Cell Death Discov 2023; 9:342. [PMID: 37714835 PMCID: PMC10504256 DOI: 10.1038/s41420-023-01644-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 08/26/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023] Open
Abstract
Levodopa-induced dyskinesia (LID) is a common motor complication in Parkinson's disease. However, few studies have focused on the pathogenesis of LID at the transcriptional level. NONRATT023402.2, a long non-coding RNA (lncRNA) that may be related to LID was discovered in our previous study and characterized in rat models of LID. In the present study, NONRATT023402.2 was overexpressed by injection of adeno-associated virus (AAV) in striatum of LID rats, and 48 potential target genes, including nerve growth factor receptor (NGFR) were screened using next-generation sequencing and target gene predictions. The NONRATT023402.2/rno-miR-3065-5p/NGFR axis was verified using a dual luciferase reporter gene. Overexpression of NONRATT023402.2 significantly increased the abnormal involuntary movements (AIM) score of LID rats, activated the PI3K/Akt signaling pathway, and up-regulated c-Fos in the striatum. NGFR knockdown by injection of ShNGFR-AAV into the striatum of LID rats resulted in a significant decrease in the PI3K/Akt signaling pathway and c-Fos expression. The AIM score of LID rats was positively correlated with the expressions of NONRATT023402.2 and NGFR. A dual luciferase reporter assay showed that c-Fos, as a transcription factor, bound to the NONRATT023402.2 promoter and activated its expression. Together, the results showed that NONRATT023402.2 regulated NGFR expression via a competing endogenous RNA mechanism, which then activated the PI3K/Akt pathway and promoted c-Fos expression. This suggested that c-Fos acted as a transcription factor to activate NONRATT023402.2 expression, and form a positive feedback regulation loop in LID rats, thus, aggravating LID symptoms. NONRATT023402.2 is therefore a possible novel therapeutic target for LID.
Collapse
Affiliation(s)
- Qiao Wang
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Neurostimulation, Beijing, China
- Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Huizhi Wang
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Neurostimulation, Beijing, China
| | - Wenjia Meng
- Clinical School, Tianjin Medical University, Tianjin, China
| | - Chong Liu
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Neurostimulation, Beijing, China
| | - Renpeng Li
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Neurostimulation, Beijing, China
| | - Moxuan Zhang
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Neurostimulation, Beijing, China
| | - Kun Liang
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Neurostimulation, Beijing, China
| | - Yuan Gao
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Neurostimulation, Beijing, China
| | - Tingting Du
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Neurostimulation, Beijing, China
| | - Jianguo Zhang
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Neurostimulation, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chunlei Han
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Neurostimulation, Beijing, China.
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Lin Shi
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Neurostimulation, Beijing, China.
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Fangang Meng
- Department of Functional Neurosurgery, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
- Beijing Key Laboratory of Neurostimulation, Beijing, China.
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
- Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
10
|
Advani D, Kumar P. Deciphering the molecular mechanism and crosstalk between Parkinson's disease and breast cancer through multi-omics and drug repurposing approach. Neuropeptides 2022; 96:102283. [PMID: 35994781 DOI: 10.1016/j.npep.2022.102283] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/01/2022] [Accepted: 08/01/2022] [Indexed: 10/15/2022]
Abstract
Epidemiological studies indicate a higher occurrence of breast cancer (BRCA) in patients with Parkinson's disease. However, the exact molecular mechanism is still not precise. Herein, we tested the hypothesis that this inverse comorbidity result from shared genetic and molecular processes. We conducted an integrated omics analysis to identify the common gene signatures associated with PD and BRCA. Secondly, several dysregulated biological processes in both indications were analyzed by functional enrichment methods, and significant overlapping processes were identified. To establish common regulatory mechanisms, information about transcription factors and miRNAs associated with both the disorders was extracted. Finally, disease-specific gene expression signatures were compared through LINCS L1000 analysis to identify potential repurposing drugs for PD. The potential repurposed drug candidates were then correlated with PD-specific gene signatures by Cmap analysis. In conclusion, this study highlights the shared genes, biological pathways and regulatory signatures associated with PD and BRCA with an improved understanding of crosstalk involved. Additionally, the role of therapeutics was investigated in context with their comorbid associations. These findings could help to explain the complex molecular patterns of associations between PD and BRCA.
Collapse
Affiliation(s)
- Dia Advani
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), Shahbad Daulatpur, Bawana Road, Delhi 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), Shahbad Daulatpur, Bawana Road, Delhi 110042, India.
| |
Collapse
|
11
|
dos Santos Pereira M, do Nascimento GC, Bortolanza M, Michel PP, Raisman-Vozari R, Del Bel E. Doxycycline attenuates l-DOPA-induced dyskinesia through an anti-inflammatory effect in a hemiparkinsonian mouse model. Front Pharmacol 2022; 13:1045465. [PMID: 36506543 PMCID: PMC9728610 DOI: 10.3389/fphar.2022.1045465] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/11/2022] [Indexed: 11/24/2022] Open
Abstract
The pharmacological manipulation of neuroinflammation appears to be a promising strategy to alleviate l-DOPA-induced dyskinesia (LID) in Parkinson's disease (PD). Doxycycline (Doxy), a semisynthetic brain-penetrant tetracycline antibiotic having interesting anti-inflammatory properties, we addressed the possibility that this compound could resolve LID in l-DOPA-treated C57BL/6 mice presenting either moderate or intermediate lesions of the mesostriatal dopaminergic pathway generated by intrastriatal injections of 6-OHDA. Doxy, when given subcutaneously before l-DOPA at doses of 20 mg kg-1 and 40 mg kg-1, led to significant LID reduction in mice with moderate and intermediate dopaminergic lesions, respectively. Importantly, Doxy did not reduce locomotor activity improved by l-DOPA. To address the molecular mechanism of Doxy, we sacrificed mice with mild lesions 1) to perform the immunodetection of tyrosine hydroxylase (TH) and Fos-B and 2) to evaluate a panel of inflammation markers in the striatum, such as cyclooxygenase-2 and its downstream product Prostaglandin E2 along with the cytokines TNF-α, IL-1β and IL-6. TH-immunodetection revealed that vehicle and Doxy-treated mice had similar striatal lesions, excluding that LID improvement by Doxy could result from neurorestorative effects. Importantly, LID inhibition by Doxy was associated with decreased Fos-B and COX-2 expression and reduced levels of PGE2, TNF-α, and IL-1β in the dorsolateral striatum of dyskinetic mice. We conclude 1) that Doxy has the potential to prevent LID regardless of the intensity of dopaminergic lesioning and 2) that the anti-inflammatory effects of Doxy probably account for LID attenuation. Overall, the present results further indicate that Doxy might represent an attractive and alternative treatment for LID in PD.
Collapse
Affiliation(s)
| | | | - Mariza Bortolanza
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil
| | - Patrick Pierre Michel
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de La Pitié Salpêtrière, Paris, France
| | - Rita Raisman-Vozari
- Sorbonne Université, Paris Brain Institute-ICM, Inserm, CNRS, APHP, Hôpital de La Pitié Salpêtrière, Paris, France
| | - Elaine Del Bel
- Department of Basic and Oral Biology, FORP, Campus USP, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
12
|
Cao W, Liang S, Yang Y, Zhu C, Sun L, Zhang L. Fisetin Ameliorates Levodopa-Induced Dyskinesia in Experimental Model Parkinson's Disease: Role of Mitochondrial Activities and Monoamines Turnover. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221136674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background: Levodopa (or l-DOPA) is the current standard of care for the management of Parkinson's disease (PD), but its chronic administration has been associated with the development of LID (l-DOPA-induced dyskinesia). Fisetin is a dietary flavonoid known for its neuroprotective efficacy. Aim: To determine the neuroprotective potential of fisetin in 6-hydroxydopamine (6-OHDA)-lesioned LID animals. Methods: 6-OHDA (12 µg and L-ascorbic acid [10 µL]) was injected in a substantial nigra of Sprague-Dawley rat to develop PD followed by l-DOPA (20 mg/kg and benserazide HCl [5 mg/kg], 42 days) to induce LID. Rats were concomitantly administered with vehicle or amantadine (40 mg/kg), or fisetin (5, 10, and 25 mg/kg, p.o.) for 42 days with l-DOPA. Results: Chronic l-DOPA administration resulted in progressive abnormal involuntary movements (AIMs viz. axial, forelimb, and orolingual), akinesia (forelimb adjusting steps, FAS), muscular rigidity (catalepsy bar test), muscular coordination, and neurological impairments. Fisetin at doses of 10 and 25 mg/kg effectively reduced ( P < .05) these LID-induced AIMs and behavioral changes. Furthermore, fisetin treatment markedly ( P < .05) attenuated LID-induced diminished striatal mitochondrial complex activities, striatal monoamines (serotonin [5-HT] and dopamine [DA]), elevated monoamines turnover (DA: DOPAC and 5-HT: 5-HIAA). In addition, fisetin treatment effectively ( P < .05) reversed the upregulated expressions of striatal cFOS, FosB, Homer, Nurr-77, Parkin, and Pdyn. Conclusion: Our study demonstrated that fisetin offered neuroprotection via amelioration of striatum mitochondrial dysfunction and monoamine (5-HT and DA) turnover to halt further development of abnormal involuntary movement and dyskinesia.
Collapse
Affiliation(s)
- Wenhui Cao
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Nangang District, Harbin Heilongjiang Province, China
- Department of Neurology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Shaodong Liang
- Department of Neurosurgery, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Yindong Yang
- Department of Neurology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Chuanzhen Zhu
- Graduate School, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Li Sun
- Department of Neurology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Liming Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Nangang District, Harbin Heilongjiang Province, China
| |
Collapse
|
13
|
Kumar A, Aglyamova G, Yim Y, Bailey AO, Lynch H, Powell R, Nguyen N, Rosenthal Z, Zhao WN, Li Y, Chen J, Fan S, Lee H, Russell W, Stephan C, Robison A, Haggarty S, Nestler E, Zhou J, Machius M, Rudenko G. Chemically targeting the redox switch in AP1 transcription factor ΔFOSB. Nucleic Acids Res 2022; 50:9548-9567. [PMID: 36039764 PMCID: PMC9458432 DOI: 10.1093/nar/gkac710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/21/2022] [Accepted: 08/08/2022] [Indexed: 12/24/2022] Open
Abstract
The AP1 transcription factor ΔFOSB, a splice variant of FOSB, accumulates in the brain in response to chronic insults such as exposure to drugs of abuse, depression, Alzheimer's disease and tardive dyskinesias, and mediates subsequent long-term neuroadaptations. ΔFOSB forms heterodimers with other AP1 transcription factors, e.g. JUND, that bind DNA under control of a putative cysteine-based redox switch. Here, we reveal the structural basis of the redox switch by determining a key missing crystal structure in a trio, the ΔFOSB/JUND bZIP domains in the reduced, DNA-free form. Screening a cysteine-focused library containing 3200 thiol-reactive compounds, we identify specific compounds that target the redox switch, validate their activity biochemically and in cell-based assays, and show that they are well tolerated in different cell lines despite their general potential to bind to cysteines covalently. A crystal structure of the ΔFOSB/JUND bZIP domains in complex with a redox-switch-targeting compound reveals a deep compound-binding pocket near the DNA-binding site. We demonstrate that ΔFOSB, and potentially other, related AP1 transcription factors, can be targeted specifically and discriminately by exploiting unique structural features such as the redox switch and the binding partner to modulate biological function despite these proteins previously being thought to be undruggable.
Collapse
Affiliation(s)
| | | | - Yun Young Yim
- Nash Family Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Aaron O Bailey
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Haley M Lynch
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Reid T Powell
- HTS Screening Core, Texas A&M University School of Medicine, Institute of Biosciences and Technology, Center for Translational Cancer Research, Houston, TX 77030, USA
| | - Nghi D Nguyen
- HTS Screening Core, Texas A&M University School of Medicine, Institute of Biosciences and Technology, Center for Translational Cancer Research, Houston, TX 77030, USA
| | - Zachary Rosenthal
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Departments of Psychiatry & Neurology, Harvard Medical School, Boston, MA 02114, USA
| | - Wen-Ning Zhao
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Departments of Psychiatry & Neurology, Harvard Medical School, Boston, MA 02114, USA
| | - Yi Li
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Jianping Chen
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Shanghua Fan
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA,Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Hubert Lee
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA,Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - William K Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Clifford Stephan
- HTS Screening Core, Texas A&M University School of Medicine, Institute of Biosciences and Technology, Center for Translational Cancer Research, Houston, TX 77030, USA
| | - Alfred J Robison
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Departments of Psychiatry & Neurology, Harvard Medical School, Boston, MA 02114, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience and the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA,Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Mischa Machius
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA,Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Gabby Rudenko
- To whom correspondence should be addressed. Tel: +1 409 772 6292; Fax: +1 409 772 9642;
| |
Collapse
|
14
|
Wan Y, Han L, Rong L, Yang S, Song L, Wu N, Liu Z, Gan J. Inhibition of BET Protein Function Suppressed the Overactivation of the Canonical NF-κB Signaling Pathway in 6-OHDA-Lesioned Rat Model of Levodopa-Induced Dyskinesia. Front Neurosci 2022; 16:896322. [PMID: 35801173 PMCID: PMC9253514 DOI: 10.3389/fnins.2022.896322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundNeuroinflammation is involved in the mechanisms of levodopa-induced dyskinesia (LID). The canonical NF-κB activation signaling pathway plays a critical role in the neuroinflammation development and BET protein-induced NF-κB-mediated neuroinflammation. The inhibition of the BET protein function has been reported to alleviate LID; however, its association with the canonical NF-κB signaling pathway in the 6-OHDA-lesioned striatum of the LID rat model remains unknown. Accordingly, we identified the status of the canonical NF-κB signaling pathway in the 6-OHDA-lesioned striatum of the LID rat model and whether the anti-dyskinetic effect of the BET inhibitor JQ1 was associated with its suppression on NF-κB-mediated neuroinflammation.Methods6-OHDA PD rat models were treated with either L-dopa plus JQ1 or L-dopa alone. L-dopa treatment was given for 2 weeks, and the JQ1 treatment was given for 3 weeks and was initiated a week prior to L-dopa treatment. As a control, the sham rats were treated with JQ1 or Veh for 3 weeks. The ALO AIM assessment and cylinder test were performed during the treatment. Glial activation markers, pro-inflammatory substances, and critical proteins in the canonical NF-κB signaling pathway were tested in the lesioned striatum after the final treatment.ResultsJQ1 effectively alleviated LID without influencing motor improvement. In the lesioned striatum, L-dopa triggered an overactivation of the canonical NF-κB signaling pathway, with an increase in the phospho-IKKα/β, phospho-IκBα, and NF-κB nuclear translocation and its phosphorylation at Ser 536 and Ser 276 sites (p < 0.01 vs. sham group). L-dopa induced an overexpression of the pro-inflammatory substances of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, and inducible nitric oxide synthase (iNOS), and the glial activation markers CD68 and GFAP. All the molecular changes were greatly inhibited by JQ1.ConclusionL-dopa triggered an overactivation of the canonical NF-κB signaling pathway, leading to an enhanced neuroinflammation response in the 6-OHDA-lesioned striatum of LID rat models. The inhibition of the BET protein function significantly suppressed the activation of the canonical NF-κB signaling pathway in the striatum, alleviating the neuroinflammation response and the severity of LID.
Collapse
Affiliation(s)
- Ying Wan
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Han
- Department of Gerontology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu Rong
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuyuan Yang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu Song
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Na Wu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenguo Liu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Zhenguo Liu,
| | - Jing Gan
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Jing Gan,
| |
Collapse
|
15
|
Coutant B, Frontera JL, Perrin E, Combes A, Tarpin T, Menardy F, Mailhes-Hamon C, Perez S, Degos B, Venance L, Léna C, Popa D. Cerebellar stimulation prevents Levodopa-induced dyskinesia in mice and normalizes activity in a motor network. Nat Commun 2022; 13:3211. [PMID: 35680891 PMCID: PMC9184492 DOI: 10.1038/s41467-022-30844-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 05/23/2022] [Indexed: 11/09/2022] Open
Abstract
Chronic Levodopa therapy, the gold-standard treatment for Parkinson's Disease (PD), leads to the emergence of involuntary movements, called levodopa-induced dyskinesia (LID). Cerebellar stimulation has been shown to decrease LID severity in PD patients. Here, in order to determine how cerebellar stimulation induces LID alleviation, we performed daily short trains of optogenetic stimulations of Purkinje cells (PC) in freely moving LID mice. We demonstrated that these stimulations are sufficient to suppress LID or even prevent their development. This symptomatic relief is accompanied by the normalization of aberrant neuronal discharge in the cerebellar nuclei, the motor cortex and the parafascicular thalamus. Inhibition of the cerebello-parafascicular pathway counteracted the beneficial effects of cerebellar stimulation. Moreover, cerebellar stimulation reversed plasticity in D1 striatal neurons and normalized the overexpression of FosB, a transcription factor causally linked to LID. These findings demonstrate LID alleviation and prevention by daily PC stimulations, which restore the function of a wide motor network, and may be valuable for LID treatment.
Collapse
Affiliation(s)
- Bérénice Coutant
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, 75005, Paris, France
| | - Jimena Laura Frontera
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, 75005, Paris, France
| | - Elodie Perrin
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France
| | - Adèle Combes
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, 75005, Paris, France
| | - Thibault Tarpin
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, 75005, Paris, France
| | - Fabien Menardy
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, 75005, Paris, France
| | - Caroline Mailhes-Hamon
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, 75005, Paris, France
| | - Sylvie Perez
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France
| | - Bertrand Degos
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France
| | - Laurent Venance
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France
| | - Clément Léna
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, 75005, Paris, France.
| | - Daniela Popa
- Neurophysiology of Brain Circuits Team, Institut de biologie de l'Ecole normale supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, PSL Research University, 75005, Paris, France.
| |
Collapse
|
16
|
Hutny M, Hofman J, Klimkowicz-Mrowiec A, Gorzkowska A. Current Knowledge on the Background, Pathophysiology and Treatment of Levodopa-Induced Dyskinesia-Literature Review. J Clin Med 2021; 10:jcm10194377. [PMID: 34640395 PMCID: PMC8509231 DOI: 10.3390/jcm10194377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/02/2021] [Accepted: 09/22/2021] [Indexed: 02/07/2023] Open
Abstract
Levodopa remains the primary drug for controlling motor symptoms in Parkinson’s disease through the whole course, but over time, complications develop in the form of dyskinesias, which gradually become more frequent and severe. These abnormal, involuntary, hyperkinetic movements are mainly characteristic of the ON phase and are triggered by excess exogenous levodopa. They may also occur during the OFF phase, or in both phases. Over the past 10 years, the issue of levodopa-induced dyskinesia has been the subject of research into both the substrate of this pathology and potential remedial strategies. The purpose of the present study was to review the results of recent research on the background and treatment of dyskinesia. To this end, databases were reviewed using a search strategy that included both relevant keywords related to the topic and appropriate filters to limit results to English language literature published since 2010. Based on the selected papers, the current state of knowledge on the morphological, functional, genetic and clinical features of levodopa-induced dyskinesia, as well as pharmacological, genetic treatment and other therapies such as deep brain stimulation, are described.
Collapse
Affiliation(s)
- Michał Hutny
- Students’ Scientific Society, Department of Neurorehabilitation, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland;
- Correspondence:
| | - Jagoda Hofman
- Students’ Scientific Society, Department of Neurorehabilitation, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland;
| | - Aleksandra Klimkowicz-Mrowiec
- Department of Internal Medicine and Gerontology, Faculty of Medicine, Medical College, Jagiellonian University, 30-688 Kraków, Poland;
| | - Agnieszka Gorzkowska
- Department of Neurorehabilitation, Faculty of Medical Sciences, School of Medicine, Medical University of Silesia, 40-752 Katowice, Poland;
| |
Collapse
|
17
|
Fieblinger T. Striatal Control of Movement: A Role for New Neuronal (Sub-) Populations? Front Hum Neurosci 2021; 15:697284. [PMID: 34354577 PMCID: PMC8329243 DOI: 10.3389/fnhum.2021.697284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/29/2021] [Indexed: 12/19/2022] Open
Abstract
The striatum is a very heterogenous brain area, composed of different domains and compartments, albeit lacking visible anatomical demarcations. Two populations of striatal spiny projection neurons (SPNs) build the so-called direct and indirect pathway of the basal ganglia, whose coordinated activity is essential to control locomotion. Dysfunction of striatal SPNs is part of many movement disorders, such as Parkinson’s disease (PD) and L-DOPA-induced dyskinesia. In this mini review article, I will highlight recent studies utilizing single-cell RNA sequencing to investigate the transcriptional profiles of striatal neurons. These studies discover that SPNs carry a transcriptional signature, indicating both their anatomical location and compartmental identity. Furthermore, the transcriptional profiles reveal the existence of additional distinct neuronal populations and previously unknown SPN sub-populations. In a parallel development, studies in rodent models of PD and L-DOPA-induced dyskinesia (LID) report that direct pathway SPNs do not react uniformly to L-DOPA therapy, and that only a subset of these neurons is underlying the development of abnormal movements. Together, these studies demonstrate a new level of cellular complexity for striatal (dys-) function and locomotor control.
Collapse
Affiliation(s)
- Tim Fieblinger
- Institute for Synaptic Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
18
|
Bordone MP, Damianich A, Bernardi MA, Eidelman T, Sanz-Blasco S, Gershanik OS, Avale ME, Ferrario JE. Fyn knockdown prevents levodopa-induced dyskinesia in a mouse model of Parkinson's disease. eNeuro 2021; 8:ENEURO.0559-20.2021. [PMID: 34099487 PMCID: PMC8281260 DOI: 10.1523/eneuro.0559-20.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 12/23/2022] Open
Abstract
Dopamine replacement by levodopa is the most widely used therapy for Parkinson's disease (PD), however patients often develop side effects, known as levodopa-induced dyskinesia (LID), that usually need therapeutic intervention. There are no suitable therapeutic options for LID, except for the use of the NMDA receptor antagonist amantadine, which has limited efficacy. The NMDA receptor is indeed the most plausible target to manage LID in PD and recently the kinase Fyn- one of its key regulators- became a new putative molecular target involved in LID. The aim of this work was to reduce Fyn expression to alleviate LID in a mouse model of PD. We performed intra-striatal delivery of a designed micro-RNA against Fyn (miRNA-Fyn) in 6-OHDA-lesioned mice treated with levodopa. The miRNA-Fyn was delivered either before or after levodopa exposure to assess its ability to prevent or revert dyskinesia. Pre-administration of miRNA-Fyn reduced LID with a concomitant reduction of FosB-ΔFosB protein levels -a marker of LID- as well as decreased phosphorylation of the NR2B-NMDA subunit, which is a main target of Fyn. On the other hand, post L-DOPA delivery of miRNA-Fyn was less effective to revert already established dyskinesia, suggesting that early blocking of Fyn activity might be a more efficient therapeutic approach. Together, our results provide proof of concept about Fyn as a plausible therapeutic target to manage LID, and validate RNA silencing as a potential approach to locally reduce striatal Fyn, rising new perspectives for RNA therapy interventions in PD.Significance StatementLevodopa induced dyskinesia (LID) is an incapacitant side effect of treatment in Parkinson's disease (PD). LID is a therapeutic challenge, lacking an effective pharmacological treatment, except for the use of inhibitors of the NMDA receptor, which have limited efficacy and may trigger untoward side effects. The kinase Fyn is a key regulator of NMDA function and a potential therapeutic target to control LID. Here, we show that RNA interference therapy to reduce the amount of Fyn mRNA in the adult brain is effective to prevent LID in a mouse model of PD, setting the grounds for future biomedical interventions to manage LID in PD.
Collapse
Affiliation(s)
- Melina P Bordone
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Instituto de Biociencias, Biotecnología y Biología traslacional (iB3), Ciudad Autónoma de Buenos Aires, Argentina (C1428EGA)
- CONICET, Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
| | - Ana Damianich
- CONICET - Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), "Dr. Héctor N. Torres", Ciudad Autónoma de Buenos Aires, Argentina (C1428ADN)
| | - M Alejandra Bernardi
- CONICET, Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
| | - Tomas Eidelman
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Instituto de Biociencias, Biotecnología y Biología traslacional (iB3), Ciudad Autónoma de Buenos Aires, Argentina (C1428EGA)
| | - Sara Sanz-Blasco
- CONICET, Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
| | - Oscar S Gershanik
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
| | - M Elena Avale
- CONICET - Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI), "Dr. Héctor N. Torres", Ciudad Autónoma de Buenos Aires, Argentina (C1428ADN)
| | - Juan E Ferrario
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Instituto de Biociencias, Biotecnología y Biología traslacional (iB3), Ciudad Autónoma de Buenos Aires, Argentina (C1428EGA).
- CONICET, Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Investigaciones Farmacológicas (ININFA), Ciudad Autónoma de Buenos Aires, Argentina (C1113AAD)
| |
Collapse
|
19
|
Liu Z, Yan A, Zhao J, Yang S, Song L, Liu Z. The p75 neurotrophin receptor as a novel intermediate in L-dopa-induced dyskinesia in experimental Parkinson's disease. Exp Neurol 2021; 342:113740. [PMID: 33971218 DOI: 10.1016/j.expneurol.2021.113740] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 04/14/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022]
Abstract
In Parkinson's disease (PD), long-term administration of L-dopa often leads to L-dopa-induced dyskinesia (LID), a debilitating motor complication. The p75 neurotrophin receptor (p75NTR) is likely to play a critical role in the regulation of dendritic spine density and morphology and appears to be associated with neuroinflammation, which previously has been identified as a crucial mechanism in LID. While aberrant modifications of p75NTR in neurological diseases have been extensively documented, only a few studies report p75NTR dysfunction in PD, and no data are available in LID. Here, we explored the functional role of p75NTR in LID. In LID rats, we identified that p75NTR was significantly increased in the lesioned striatum. In 6-hydroxydopamine (6-OHDA)-hemilesioned rats, specific knockdown of striatal p75NTR levels achieved by viral vector injection into the striatum prevented the development of LID and increased striatal structural plasticity. By contrast, we found that in 6-OHDA-hemilesioned rats, striatal p75NTR overexpression exacerbated LID and facilitated striatal dendritic spine losses. Moreover, we observed that the immunomodulatory drug fingolimod attenuated LID without lessening the therapeutic efficacy of L-dopa and normalized p75NTR levels. Together, these data demonstrate for the first time that p75NTR plays a pivotal role in the development of LID and that p75NTR may act as a potential novel target for the management of LID.
Collapse
Affiliation(s)
- Zhihua Liu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200082, China
| | - Aijuan Yan
- Department of Neurology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China
| | - Jiahao Zhao
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200082, China
| | - Shuyuan Yang
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200082, China
| | - Lu Song
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200082, China
| | - Zhenguo Liu
- Department of Neurology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200082, China.
| |
Collapse
|
20
|
Beck G, Zhang J, Fong K, Mochizuki H, Mouradian MM, Papa SM. Striatal ΔFosB gene suppression inhibits the development of abnormal involuntary movements induced by L-Dopa in rats. Gene Ther 2021; 28:760-770. [PMID: 33707771 PMCID: PMC8433270 DOI: 10.1038/s41434-021-00249-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 02/07/2021] [Accepted: 02/19/2021] [Indexed: 11/24/2022]
Abstract
L-Dopa-induced dyskinesia (LID) is associated with upregulation of striatal ΔFosB in animal models and patients with Parkinson’s disease (PD). A mechanistic role of ΔFosB is suspected because its transgenic overexpression leads to early appearance of LID in rodents and primates. The present study in rodents is aimed at exploring the therapeutic potential of striatal ΔFosB gene suppression to control LID in patients with PD. To determine the effect of reducing striatal ΔFosB expression, we used RNAi gene knockdown in a rat model of PD and assessed abnormal involuntary movements (AIMs) in response to L-Dopa. Rats with dopamine depletion received striatal injections of rAAV-ΔFosB shRNA or a control virus before exposure to chronic L-Dopa treatment. Development of AIMs during the entire L-Dopa treatment period was markedly inhibited by ΔFosB gene knockdown and its associated molecular changes. The antiparkinsonian action of L-Dopa was unchanged by ΔFosB gene knockdown. These results suggest a major role for ΔFosB in the development of LID, and support exploring strategies to reduce striatal ΔFosB levels in patients with PD.
Collapse
Affiliation(s)
- Goichi Beck
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.,Department of Neurology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Jie Zhang
- RWJMS Institute for Neurological Therapeutics and Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Kayoko Fong
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Japan
| | - M Maral Mouradian
- RWJMS Institute for Neurological Therapeutics and Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, USA.
| | - Stella M Papa
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA. .,Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
21
|
Yang K, Zhao X, Wang C, Zeng C, Luo Y, Sun T. Circuit Mechanisms of L-DOPA-Induced Dyskinesia (LID). Front Neurosci 2021; 15:614412. [PMID: 33776634 PMCID: PMC7988225 DOI: 10.3389/fnins.2021.614412] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/05/2021] [Indexed: 12/25/2022] Open
Abstract
L-DOPA is the criterion standard of treatment for Parkinson disease. Although it alleviates some of the Parkinsonian symptoms, long-term treatment induces L-DOPA–induced dyskinesia (LID). Several theoretical models including the firing rate model, the firing pattern model, and the ensemble model are proposed to explain the mechanisms of LID. The “firing rate model” proposes that decreasing the mean firing rates of the output nuclei of basal ganglia (BG) including the globus pallidus internal segment and substantia nigra reticulata, along the BG pathways, induces dyskinesia. The “firing pattern model” claimed that abnormal firing pattern of a single unit activity and local field potentials may disturb the information processing in the BG, resulting in dyskinesia. The “ensemble model” described that dyskinesia symptoms might represent a distributed impairment involving many brain regions, but the number of activated neurons in the striatum correlated most strongly with dyskinesia severity. Extensive evidence for circuit mechanisms in driving LID symptoms has also been presented. LID is a multisystem disease that affects wide areas of the brain. Brain regions including the striatum, the pallidal–subthalamic network, the motor cortex, the thalamus, and the cerebellum are all involved in the pathophysiology of LID. In addition, although both amantadine and deep brain stimulation help reduce LID, these approaches have complications that limit their wide use, and a novel antidyskinetic drug is strongly needed; these require us to understand the circuit mechanism of LID more deeply.
Collapse
Affiliation(s)
- Kai Yang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Xinyue Zhao
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Changcai Wang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Cheng Zeng
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Yan Luo
- Department of Physiology, School of Basic Medical Science, Ningxia Medical University, Yinchuan, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China.,State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
| |
Collapse
|
22
|
Guo JJ, Yue F, Song DY, Bousset L, Liang X, Tang J, Yuan L, Li W, Melki R, Tang Y, Chan P, Guo C, Li JY. Intranasal administration of α-synuclein preformed fibrils triggers microglial iron deposition in the substantia nigra of Macaca fascicularis. Cell Death Dis 2021; 12:81. [PMID: 33441545 PMCID: PMC7807015 DOI: 10.1038/s41419-020-03369-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022]
Abstract
Iron deposition is present in main lesion areas in the brains of patients with Parkinson's disease (PD) and an abnormal iron content may be associated with dopaminergic neuronal cytotoxicity and degeneration in the substantia nigra of the midbrain. However, the cause of iron deposition and its role in the pathological process of PD are unclear. In the present study, we investigated the effects of the nasal mucosal delivery of synthetic human α-synuclein (α-syn) preformed fibrils (PFFs) on the pathogenesis of PD in Macaca fascicularis. We detected that iron deposition was clearly increased in a time-dependent manner from 1 to 17 months in the substantia nigra and globus pallidus, highly contrasting to other brain regions after treatments with α-syn PFFs. At the cellular level, the iron deposits were specifically localized in microglia but not in dopaminergic neurons, nor in other types of glial cells in the substantia nigra, whereas the expression of transferrin (TF), TF receptor 1 (TFR1), TF receptor 2 (TFR2), and ferroportin (FPn) was increased in dopaminergic neurons. Furthermore, no clear dopaminergic neuron loss was observed in the substantia nigra, but with decreased immunoreactivity of tyrosine hydroxylase (TH) and appearance of axonal swelling in the putamen. The brain region-enriched and cell-type-dependent iron localizations indicate that the intranasal α-syn PFFs treatment-induced iron depositions in microglia in the substantia nigra may appear as an early cellular response that may initiate neuroinflammation in the dopaminergic system before cell death occurs. Our data suggest that the inhibition of iron deposition may be a potential approach for the early prevention and treatment of PD.
Collapse
Affiliation(s)
- Jian-Jun Guo
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
| | - Feng Yue
- Beijing Key Laboratory of Parkinson's Disease, National Clinical Research Center for Geriatric Disorders, Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Dong-Yan Song
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
| | - Luc Bousset
- Laboratory of Neurodegenerative Diseases, CNRS and Institut François Jacob (MIRCen), CEA, Fontenay-aux-Roses, 92260, France
| | - Xin Liang
- Department of Histology, Chongqing Medical University, Chongqing, 400000, China
| | - Jing Tang
- Department of Histology, Chongqing Medical University, Chongqing, 400000, China
| | - Lin Yuan
- Unit of Neurodegenerative Diseases and Repair, Institute of Health Sciences, China Medical University, Shenyang, 110112, China
| | - Wen Li
- Unit of Neurodegenerative Diseases and Repair, Institute of Health Sciences, China Medical University, Shenyang, 110112, China
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Lund University, Lund, 22184, Sweden
| | - Ronald Melki
- Laboratory of Neurodegenerative Diseases, CNRS and Institut François Jacob (MIRCen), CEA, Fontenay-aux-Roses, 92260, France
| | - Yong Tang
- Department of Histology, Chongqing Medical University, Chongqing, 400000, China
| | - Piu Chan
- Beijing Key Laboratory of Parkinson's Disease, National Clinical Research Center for Geriatric Disorders, Department of Neurobiology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Chuang Guo
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China.
| | - Jia-Yi Li
- Institute of Neuroscience, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China.
- Unit of Neurodegenerative Diseases and Repair, Institute of Health Sciences, China Medical University, Shenyang, 110112, China.
- Neural Plasticity and Repair Unit, Wallenberg Neuroscience Center, Lund University, Lund, 22184, Sweden.
| |
Collapse
|
23
|
Singh A, Papa SM. Striatal Oscillations in Parkinsonian Non-Human Primates. Neuroscience 2020; 449:116-122. [PMID: 32905842 DOI: 10.1016/j.neuroscience.2020.09.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023]
Abstract
Dopamine loss in Parkinson's disease (PD) is associated with abnormal oscillatory activity in the cortico-basal ganglia network. However, the oscillatory pattern of striatal neurons in PD remains poorly defined. Here, we analyzed the local field potentials in one untreated and five MPTP-treated non-human primates (NHP) with chronic, advanced parkinsonism. Oscillatory activities in the alpha (8-13 Hz) and low-beta (13-20 Hz) frequency bands were found in the striatum similarly to the motor cortex and globus pallidus of the NHP model of PD. Both alpha and low-beta frequency band oscillations of the striatum were highly coherent with the cortical and pallidal oscillations, confirming the presence of abnormal 8-20 Hz oscillatory activity in the cortico-basal ganglia network in parkinsonian NHPs. The reversal of parkinsonism induced by acute levodopa administration was associated with reduced 8-20 Hz oscillations in the striatum. These findings indicate that pathological oscillations at alpha and low-beta bands are also present in the striatum concordant with basal ganglia network changes in the primate model of PD.
Collapse
Affiliation(s)
- Arun Singh
- Yerkes National Primate Research Center, Emory University Atlanta, GA, United States; Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States.
| | - Stella M Papa
- Yerkes National Primate Research Center, Emory University Atlanta, GA, United States; Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
24
|
Olanow CW, Calabresi P, Obeso JA. Continuous Dopaminergic Stimulation as a Treatment for Parkinson's Disease: Current Status and Future Opportunities. Mov Disord 2020; 35:1731-1744. [DOI: 10.1002/mds.28215] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/05/2020] [Accepted: 06/15/2020] [Indexed: 12/21/2022] Open
Affiliation(s)
- C. Warren Olanow
- Department of Neurology and Department of Neuroscience Mount Sinai School of Medicine New York New York USA
- Clintrex Research Corporation Sarasota Florida USA
| | - Paolo Calabresi
- Neurology Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy
- Dipartimento Neuroscienze Università Cattolica del Sacro Cuore Rome Italy
| | - Jose A. Obeso
- CINAC, Hospital Universitario HM Puerta del Sur, Universidad CEU‐San Pablo Móstoles Madrid Spain
- CIBERNED, Instituto de Salud Carlos III Madrid Spain
| |
Collapse
|
25
|
Leta V, Jenner P, Chaudhuri KR, Antonini A. Can therapeutic strategies prevent and manage dyskinesia in Parkinson’s disease? An update. Expert Opin Drug Saf 2019; 18:1203-1218. [DOI: 10.1080/14740338.2019.1681966] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Valentina Leta
- King’s College London, Department of Neurosciences, Institute of Psychiatry, Psychology & Neuroscience, London, UK
- Parkinson’s Foundation Centre of Excellence, King’s College Hospital, London, UK
| | - Peter Jenner
- Neurodegenerative Diseases Research Group, School of Cancer and Pharmaceutical Sciences, Faculty of Life Science and Medicine, King’s College London, London, UK
| | - K. Ray Chaudhuri
- King’s College London, Department of Neurosciences, Institute of Psychiatry, Psychology & Neuroscience, London, UK
- Parkinson’s Foundation Centre of Excellence, King’s College Hospital, London, UK
| | - Angelo Antonini
- Department of Neuroscience, University of Padova, Padua, Italy
| |
Collapse
|