1
|
Roodveldt C, Bernardino L, Oztop-Cakmak O, Dragic M, Fladmark KE, Ertan S, Aktas B, Pita C, Ciglar L, Garraux G, Williams-Gray C, Pacheco R, Romero-Ramos M. The immune system in Parkinson's disease: what we know so far. Brain 2024; 147:3306-3324. [PMID: 38833182 PMCID: PMC11449148 DOI: 10.1093/brain/awae177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/02/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024] Open
Abstract
Parkinson's disease is characterized neuropathologically by the degeneration of dopaminergic neurons in the ventral midbrain, the accumulation of α-synuclein (α-syn) aggregates in neurons and chronic neuroinflammation. In the past two decades, in vitro, ex vivo and in vivo studies have consistently shown the involvement of inflammatory responses mediated by microglia and astrocytes, which may be elicited by pathological α-syn or signals from affected neurons and other cell types, and are directly linked to neurodegeneration and disease development. Apart from the prominent immune alterations seen in the CNS, including the infiltration of T cells into the brain, more recent studies have demonstrated important changes in the peripheral immune profile within both the innate and adaptive compartments, particularly involving monocytes, CD4+ and CD8+ T cells. This review aims to integrate the consolidated understanding of immune-related processes underlying the pathogenesis of Parkinson's disease, focusing on both central and peripheral immune cells, neuron-glia crosstalk as well as the central-peripheral immune interaction during the development of Parkinson's disease. Our analysis seeks to provide a comprehensive view of the emerging knowledge of the mechanisms of immunity in Parkinson's disease and the implications of this for better understanding the overall pathogenesis of this disease.
Collapse
Affiliation(s)
- Cintia Roodveldt
- Centre for Molecular Biology and Regenerative Medicine-CABIMER, University of Seville-CSIC, Seville 41092, Spain
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville 41009, Spain
| | - Liliana Bernardino
- Health Sciences Research Center (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, 6200-506, Covilhã, Portugal
| | - Ozgur Oztop-Cakmak
- Department of Neurology, Faculty of Medicine, Koç University, Istanbul 34010, Turkey
| | - Milorad Dragic
- Laboratory for Neurobiology, Department of General Physiology and Biophysics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
- Department of Molecular Biology and Endocrinology, ‘VINČA’ Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| | - Kari E Fladmark
- Department of Biological Science, University of Bergen, 5006 Bergen, Norway
| | - Sibel Ertan
- Department of Neurology, Faculty of Medicine, Koç University, Istanbul 34010, Turkey
| | - Busra Aktas
- Department of Molecular Biology and Genetics, Burdur Mehmet Akif Ersoy University, Burdur 15200, Turkey
| | - Carlos Pita
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal
| | - Lucia Ciglar
- Center Health & Bioresources, Competence Unit Molecular Diagnostics, AIT Austrian Institute of Technology GmbH, 1210 Vienna, Austria
| | - Gaetan Garraux
- Movere Group, Faculty of Medicine, GIGA Institute, University of Liège, Liège 4000, Belgium
| | | | - Rodrigo Pacheco
- Laboratorio de Neuroinmunología, Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba 8580702, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia 7510156, Santiago, Chile
| | - Marina Romero-Ramos
- Department of Biomedicine & The Danish Research Institute of Translational Neuroscience—DANDRITE, Aarhus University, DK-8000 Aarhus C, Denmark
| |
Collapse
|
2
|
Qin X, Li B, Hu B, Huang J, Tian X, Zhang X, Wang Y, Huang W. Rhein inhibits M1 polarization of BV2 microglia through MAPK/IκB signalling pathway and reduces neurotoxicity caused by neuroinflammation. Int J Dev Neurosci 2024; 84:533-545. [PMID: 38858813 DOI: 10.1002/jdn.10352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/29/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND Rhein is an anthraquinone compound with anti-inflammatory pharmacological activity. It has been found to play a neuroprotective role in neurological diseases, but the neuroprotective mechanism of rhein remains unclear. METHODS SH-SY5Y cells serving as neuron-like cells and BV2 microglia were used. The toxicity of rhein on BV2 microglia and the viability of SH-SY5Y cells were measured by CCK-8 assay. The mRNA expression and secretion of pro-inflammatory cytokines were detected by qPCR and ELISA. Iba1, CD86 and pathway signalling protein in BV2 microglia were assessed by Western blot and immunofluorescence. Apoptosis of SH-SY5Y cells exposed to neuroinflammation was analysed through flow cytometry. RESULTS Rhein inhibited MAPK/IκB signalling pathways. Further studies revealed that rhein inhibited the production of pro-inflammatory cytokines TNF-α, IL-6, IL-1β and iNOS in BV2 cells and also inhibited the expression of M1 polarization markers Iba1 and CD86 in BV2 cells. Furthermore, rhein reduced the apoptotic rate and restored cell viability of SH-SY5Y cells exposed to neuroinflammation. CONCLUSIONS Our study demonstrated that rhein inhibited microglia M1 polarization via MAPK/IκB signalling pathway and protected nerve cells through suppressing neuroinflammation.
Collapse
Affiliation(s)
- Xin Qin
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Yichang Central People's Hospital, Yichang, China
| | - Bowen Li
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Binbin Hu
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Juan Huang
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xingfu Tian
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xinyue Zhang
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ye Wang
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wei Huang
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
3
|
Qin X, Wang S, Huang J, Hu B, Yang X, Liang L, Zhou R, Huang W. Rhein alleviates MPTP-induced Parkinson's disease by suppressing neuroinflammation via MAPK/IκB pathway. Front Neurosci 2024; 18:1396345. [PMID: 38933815 PMCID: PMC11202316 DOI: 10.3389/fnins.2024.1396345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Background Parkinson's disease (PD) is a common neurodegenerative disease with a rapid increase in incidence in recent years. Existing treatments cannot slow or stop the progression of PD. It was proposed that neuroinflammation leads to neuronal death, making targeting neuroinflammation a promising therapeutic strategy. Our previous studies have demonstrated that rhein protects neurons in vitro by inhibiting neuroinflammation, and it has been found to exhibit neuroprotective effects in Alzheimer's disease and epilepsy, but its neuroprotective mechanisms and effects on PD are still unclear. Methods PD animal model was induced by 1-methyl-4-phenyl-1,2,3, 6-tetrahydropyridine (MPTP). ELISA, RT-qPCR, western blot and Immunofluorescence were used to detect the levels of inflammatory cytokines and M1 polarization markers. The protein expression levels of signaling pathways were measured by western blot. Hematoxylin-eosin (HE) staining showed that rhein did not damage the liver and kidney. Two behavioral tests, pole test and rotarod test, were used to evaluate the improvement effect of rhein on movement disorders. The number of neurons in the substantia nigra was evaluated by Nissl staining. Immunohistochemistry and western blot were used to detect tyrosine hydroxylase (TH) and α-synuclein. Results Rhein inhibited the activation of MAPK/IκB signaling pathway and reduced the levels of pro-inflammatory cytokines (IL-1β, IL-6 and TNF-α) and M1 polarization markers of microglia in vivo. In a mouse model of PD, rhein ameliorated movement disorders, reduced dopaminergic neuron damage and α-synuclein deposition. Conclusion Rhein inhibits neuroinflammation through MAPK/IκB signaling pathway, thereby reducing neurodegeneration, α-synuclein deposition, and improving movement disorders in Parkinson's disease.
Collapse
Affiliation(s)
- Xin Qin
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Neurology, Yichang Central People’s Hospital, Yichang, China
- Jiangxi Province Key Laboratory of Molecular Medicine, Nanchang, China
| | - Shu Wang
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Juan Huang
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Binbin Hu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xingyan Yang
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Liying Liang
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rui Zhou
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wei Huang
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
4
|
Xie S, Peng P, Dong X, Yuan J, Liang J. Novel gene signatures predicting and immune infiltration analysis in Parkinson's disease: based on combining random forest with artificial neural network. Neurol Sci 2024; 45:2681-2696. [PMID: 38265536 DOI: 10.1007/s10072-023-07299-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/29/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND Parkinson's disease (PD) ranks as the second most prevalent neurodegenerative disorder globally, and its incidence is rapidly rising. The diagnosis of PD relies on clinical characteristics. Although current treatments aim to alleviate symptoms, they do not effectively halt the disease's progression. Early detection and intervention hold immense importance. This study aimed to establish a new PD diagnostic model. METHODS Data from a public database were adopted for the construction and validation of a PD diagnostic model with random forest and artificial neural network models. The CIBERSORT platform was applied for the evaluation of immune cell infiltration in PD. Quantitative real-time PCR was performed to verify the accuracy and reliability of the bioinformatics analysis results. RESULTS Leveraging existing gene expression data from the Gene Expression Omnibus (GEO) database, we sifted through differentially expressed genes (DEGs) in PD and identified 30 crucial genes through a random forest classifier. Furthermore, we successfully designed a novel PD diagnostic model using an artificial neural network and verified its diagnostic efficacy using publicly available datasets. Our research also suggests that mast cells may play a significant role in the onset and progression of PD. CONCLUSION This work developed a new PD diagnostic model with machine learning techniques and suggested the immune cells as a potential target for PD therapy.
Collapse
Affiliation(s)
- Shucai Xie
- Department of Critical Care Medicine, National Clinical Research Center for Genetic Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Pei Peng
- Department of Medicine Oncology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde, China
| | - Xingcheng Dong
- Department of Orthopedics, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde, China
| | - Junxing Yuan
- Department of Neurology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), No. 818 Renmin Road, Changde, 415000, Hunan, China
| | - Ji Liang
- Department of Neurology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), No. 818 Renmin Road, Changde, 415000, Hunan, China.
| |
Collapse
|
5
|
Navarro E, Efthymiou AG, Parks M, Riboldi GM, Vialle RA, Udine E, Muller BZ, Humphrey J, Allan A, Argyrou CC, Lopes KDP, Münch A, Raymond D, Sachdev R, Shanker VL, Miravite J, Katsnelson V, Leaver K, Frucht S, Bressman SB, Marcora E, Saunders-Pullman R, Goate A, Raj T. LRRK2 G2019S variant is associated with transcriptional changes in Parkinson's disease human myeloid cells under proinflammatory environment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.27.594821. [PMID: 38854101 PMCID: PMC11160623 DOI: 10.1101/2024.05.27.594821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The G2019S mutation in the leucine-rich repeat kinase 2 (LRRK2) gene is a major risk factor for the development of Parkinson's disease (PD). LRRK2, although ubiquitously expressed, is highly abundant in cells of the innate immune system. Given the importance of central and peripheral immune cells in the development of PD, we sought to investigate the consequences of the G2019S mutation on microglial and monocyte transcriptome and function. We have generated large-scale transcriptomic profiles of isogenic human induced microglial cells (iMGLs) and patient derived monocytes carrying the G2019S mutation under baseline culture conditions and following exposure to the proinflammatory factors IFNγ and LPS. We demonstrate that the G2019S mutation exerts a profound impact on the transcriptomic profile of these myeloid cells, and describe corresponding functional differences in iMGLs. The G2019S mutation led to an upregulation in lipid metabolism and phagolysosomal pathway genes in untreated and LPS/IFNγ stimulated iMGLs, which was accompanied by an increased phagocytic capacity of myelin debris. We also identified dysregulation of cell cycle genes, with a downregulation of the E2F4 regulon. Transcriptomic characterization of human-derived monocytes carrying the G2019S mutation confirmed alteration in lipid metabolism associated genes. Altogether, these findings reveal the influence of G2019S on the dysregulation of the myeloid cell transcriptome under proinflammatory conditions.
Collapse
Affiliation(s)
- Elisa Navarro
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Genetics and Genomic Sciences & Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Instituto Universitario de Investigacion en Neuroquimica, Departamento de Bioquimica y Biologia Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
- Instituto Ramon y Cajal de Investigacion Sanitaria (IRYCIS), Madrid, Spain
| | - Anastasia G. Efthymiou
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Genetics and Genomic Sciences & Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Madison Parks
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Genetics and Genomic Sciences & Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Giulietta M Riboldi
- The Marlene and Paolo Fresco Institute for Parkinson’s Disease and Movement Disorders, New York University Langone Health, New York, NY, USA
| | - Ricardo A. Vialle
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Genetics and Genomic Sciences & Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Rush Alzheimer’s Disease Center, Rush University Medical Center; Chicago, IL, 60612, USA
- Department of Neurological Sciences, Rush University Medical Center; Chicago, IL, 60612, USA
| | - Evan Udine
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Genetics and Genomic Sciences & Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Benjamin Z. Muller
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Genetics and Genomic Sciences & Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Jack Humphrey
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Genetics and Genomic Sciences & Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Amanda Allan
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Genetics and Genomic Sciences & Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Charlie Charalambos Argyrou
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Genetics and Genomic Sciences & Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Katia de Paiva Lopes
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Genetics and Genomic Sciences & Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Rush Alzheimer’s Disease Center, Rush University Medical Center; Chicago, IL, 60612, USA
- Department of Neurological Sciences, Rush University Medical Center; Chicago, IL, 60612, USA
| | - Alexandra Münch
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Genetics and Genomic Sciences & Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Deborah Raymond
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rivka Sachdev
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vicki L. Shanker
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joan Miravite
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Viktoryia Katsnelson
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine Leaver
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steve Frucht
- The Marlene and Paolo Fresco Institute for Parkinson’s Disease and Movement Disorders, New York University Langone Health, New York, NY, USA
| | - Susan B Bressman
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edoardo Marcora
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Genetics and Genomic Sciences & Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Rachel Saunders-Pullman
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Neurology, Mount Sinai Beth Israel, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison Goate
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Genetics and Genomic Sciences & Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Towfique Raj
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Ronald M. Loeb Center for Alzheimer’s Disease, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Genetics and Genomic Sciences & Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| |
Collapse
|
6
|
Li D, Yu SF, Lin L, Guo JR, Huang SM, Wu XL, You HL, Cheng XJ, Zhang QY, Zeng YQ, Pan XD. Deficiency of leucine-rich repeat kinase 2 aggravates thioacetamide-induced acute liver failure and hepatic encephalopathy in mice. J Neuroinflammation 2024; 21:123. [PMID: 38725082 PMCID: PMC11084037 DOI: 10.1186/s12974-024-03125-4] [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: 08/14/2023] [Accepted: 05/05/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Hepatic encephalopathy (HE) is closely associated with inflammatory responses. However, as a crucial regulator of the immune and inflammatory responses, the role of leucine-rich repeat kinase 2 (LRRK2) in the pathogenesis of HE remains unraveled. Herein, we investigated this issue in thioacetamide (TAA)-induced HE following acute liver failure (ALF). METHODS TAA-induced HE mouse models of LRRK2 wild type (WT), LRRK2 G2019S mutation (Lrrk2G2019S) and LRRK2 knockout (Lrrk2-/-) were established. A battery of neurobehavioral experiments was conducted. The biochemical indexes and pro-inflammatory cytokines were detected. The prefrontal cortex (PFC), striatum (STR), hippocampus (HIP), and liver were examined by pathology and electron microscopy. The changes of autophagy-lysosomal pathway and activity of critical Rab GTPases were analyzed. RESULTS The Lrrk2-/--HE model reported a significantly lower survival rate than the other two models (24% vs. 48%, respectively, p < 0.05), with no difference found between the WT-HE and Lrrk2G2019S-HE groups. Compared with the other groups, after the TAA injection, the Lrrk2-/- group displayed a significant increase in ammonium and pro-inflammatory cytokines, aggravated hepatic inflammation/necrosis, decreased autophagy, and abnormal phosphorylation of lysosomal Rab10. All three models reported microglial activation, neuronal loss, disordered vesicle transmission, and damaged myelin structure. The Lrrk2-/--HE mice presented no severer neuronal injury than the other genotypes. CONCLUSIONS LRRK2 deficiency may exacerbate TAA-induced ALF and HE in mice, in which inflammatory response is evident in the brain and aggravated in the liver. These novel findings indicate a need of sufficient clinical awareness of the adverse effects of LRRK2 inhibitors on the liver.
Collapse
Affiliation(s)
- Dan Li
- Department of Gastroenterology, Fujian Medical University Union Hospital, 29, Xinquan Road, Fujian, 350001, China.
- Fujian Clinical Research Center for Digestive System Tumors and Upper Gastrointestinal Diseases, Fujian, 350001, China.
| | - Shu-Fang Yu
- Department of Gastroenterology, Fujian Medical University Union Hospital, 29, Xinquan Road, Fujian, 350001, China
| | - Lin Lin
- Department of Neurology, Fujian Institute of Geriatrics, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fuzhou, 350001, Fujian, China
| | - Jie-Ru Guo
- Department of Gastroenterology, Fujian Medical University Union Hospital, 29, Xinquan Road, Fujian, 350001, China
| | - Si-Mei Huang
- Department of Gastroenterology, Fujian Medical University Union Hospital, 29, Xinquan Road, Fujian, 350001, China
| | - Xi-Lin Wu
- Department of Neurology, Fujian Institute of Geriatrics, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China
- Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China
| | - Han-Lin You
- Department of Neurology, Fujian Institute of Geriatrics, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China
| | - Xiao-Juan Cheng
- Department of Neurology, Fujian Institute of Geriatrics, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China
| | - Qiu-Yang Zhang
- Department of Neurology, Fujian Institute of Geriatrics, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
| | - Yu-Qi Zeng
- Department of Neurology, Fujian Institute of Geriatrics, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China
| | - Xiao-Dong Pan
- Department of Neurology, Fujian Institute of Geriatrics, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, China.
- Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China.
- Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fuzhou, 350001, Fujian, China.
- Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou, 350001, China.
- Clinical Research Center for Precision Diagnosis and Treatment of Neurological Diseases of Fujian Province, Fuzhou, 350001, China.
| |
Collapse
|
7
|
El Otmani H, Daghi M, Tahiri Jouti N, Lesage S. An overview of the worldwide distribution of LRRK2 mutations in Parkinson's disease. Neurodegener Dis Manag 2023; 13:335-350. [PMID: 38305913 DOI: 10.2217/nmt-2023-0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder with significant genetic influence. The LRRK2 gene is a major genetic contributor, particularly the Gly2019Ser mutation. This focused review investigates the global distribution of LRRK2 mutations, with emphasis on Gly2019Ser and other pathogenic variants. Prevalence rates of Gly2019Ser are highest in North Africa and the Ashkenazi-Jewish population, indicating a potential common ancestor and founder effect. Other LRRK2 mutations, including Asn1437His, Arg1441Gly/Cys/His, Tyr1699Cys and Ile2020Thr, exhibit varying global prevalences. Understanding these distributions enhances our knowledge of PD genetics and aids personalized medicine. Further research is crucial to unravel clinical implications and develop targeted therapies for LRRK2 mutation carriers.
Collapse
Affiliation(s)
- Hicham El Otmani
- Laboratory of Medical Genetics & Molecular Pathology. Faculty of Medicine and Pharmacy, Hassan II University, 20250, Casablanca, Morocco
- Laboratory of Cellular and Molecular Inflammatory, Degenerative & Oncologic Pathophysiology. Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, 20250, Morocco
- Department of Neurology. Ibn Rochd University Hospital, Casablanca, 20360, Morocco
| | - Mohamed Daghi
- Research Laboratory of Nervous System Diseases, Neurosensory Disorders & Disability. Faculty of Medicine & Pharmacy, Hassan II University, Casablanca, 20250, Morocco
| | - Nadia Tahiri Jouti
- Laboratory of Cellular and Molecular Inflammatory, Degenerative & Oncologic Pathophysiology. Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, 20250, Morocco
| | - Suzanne Lesage
- Sorbonne University, Institut du Cerveau-Paris Brain Institute, ICM, INSERM, CNRS, Assistance Publique-Hôpitaux de Paris, Paris, 75013, France
| |
Collapse
|
8
|
Yadavalli N, Ferguson SM. LRRK2 suppresses lysosome degradative activity in macrophages and microglia through MiT-TFE transcription factor inhibition. Proc Natl Acad Sci U S A 2023; 120:e2303789120. [PMID: 37487100 PMCID: PMC10400961 DOI: 10.1073/pnas.2303789120] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/12/2023] [Indexed: 07/26/2023] Open
Abstract
Cells maintain optimal levels of lysosome degradative activity to protect against pathogens, clear waste, and generate nutrients. Here, we show that LRRK2, a protein that is tightly linked to Parkinson's disease, negatively regulates lysosome degradative activity in macrophages and microglia via a transcriptional mechanism. Depletion of LRRK2 and inhibition of LRRK2 kinase activity enhanced lysosomal proteolytic activity and increased the expression of multiple lysosomal hydrolases. Conversely, the kinase hyperactive LRRK2 G2019S Parkinson's disease mutant suppressed lysosomal degradative activity and gene expression. We identified MiT-TFE transcription factors (TFE3, TFEB, and MITF) as mediators of LRRK2-dependent control of lysosomal gene expression. LRRK2 negatively regulated the abundance and nuclear localization of these transcription factors and their depletion prevented LRRK2-dependent changes in lysosome protein levels. These observations define a role for LRRK2 in controlling lysosome degradative activity and support a model wherein LRRK2 hyperactivity may increase Parkinson's disease risk by suppressing lysosome degradative activity.
Collapse
Affiliation(s)
- Narayana Yadavalli
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT06510
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT06510
- Wu Tsai Institute, Yale University School of Medicine, New Haven, CT06510
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
| | - Shawn M. Ferguson
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT06510
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT06510
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT06510
- Wu Tsai Institute, Yale University School of Medicine, New Haven, CT06510
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD20815
- Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT06510
| |
Collapse
|
9
|
Miller GK, Kuruvilla S, Jacob B, LaFranco-Scheuch L, Bakthavatchalu V, Flor J, Flor K, Ziegler J, Reichard C, Manfre P, Firner S, McNutt T, Quay D, Bellum S, Doto G, Ciaccio PJ, Pearson K, Valentine J, Fuller P, Fell M, Tsuchiya T, Williamson T, Wollenberg G. Effects of LRRK2 Inhibitors in Nonhuman Primates. Toxicol Pathol 2023; 51:232-245. [PMID: 37916535 DOI: 10.1177/01926233231205895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Toxicology studies in nonhuman primates were conducted to evaluate selective, brain penetrant inhibitors of LRRK2. GNE 7915 was limited to 7-day administration in cynomolgus monkeys at 65 mg/kg/day or limited to 14 days in rhesus at 22.5 mg/kg b.i.d. due to physical signs. Compound 25 demonstrated acceptable tolerability at 50 and 225 mg/kg b.i.d. for 7 days in rhesus monkeys. MK-1468 was tolerated during 7-day administration at 100, 200 or 800 mg/kg/day or for 30-day administration at 30, 100, or 500 mg/kg b.i.d. in rhesus monkeys. The lungs revealed hypertrophy of type 2 pneumocytes, with accumulation of intra-alveolar macrophages. Transmission electron microscopy confirmed increased lamellar structures within hypertrophic type 2 pneumocytes. Hypertrophy and hyperplasia of type 2 pneumocytes with accumulation of intra-alveolar macrophages admixed with neutrophils were prominent at peripheral lungs of animals receiving compound 25 or MK-1468. Affected type 2 pneumocytes were immuno-positive for pro-surfactant C, but negative for CD11c, a marker for intra-alveolar macrophages. Accumulation of collagen within alveolar walls, confirmed by histochemical trichrome stain, accompanied changes described for compound 25 and MK-1468. Following a 12-week treatment-free interval, animals previously receiving MK-1468 for 30 days exhibited remodeling of alveolar structure and interstitial components that did not demonstrate reversibility.
Collapse
Affiliation(s)
| | | | | | | | | | - Jason Flor
- Merck & Co., Inc., Rahway, New Jersey, USA
| | | | | | | | | | | | | | - Diane Quay
- Merck & Co., Inc., Rahway, New Jersey, USA
| | | | - Greg Doto
- Merck & Co., Inc., Rahway, New Jersey, USA
| | | | | | | | | | - Matt Fell
- Merck & Co., Inc., Rahway, New Jersey, USA
| | | | | | | |
Collapse
|
10
|
Berriat F, Lobsiger CS, Boillée S. The contribution of the peripheral immune system to neurodegeneration. Nat Neurosci 2023:10.1038/s41593-023-01323-6. [PMID: 37231108 DOI: 10.1038/s41593-023-01323-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 04/05/2023] [Indexed: 05/27/2023]
Abstract
Microglial cells are the major immune cells of the central nervous system (CNS), and directly react to neurodegeneration, but other immune cell types are also able to react to pathology and can modify the course of neurodegenerative processes. These mainly include monocytes/macrophages and lymphocytes. While these peripheral immune cells were initially considered to act only after infiltrating the CNS, recent evidence suggests that some of them can also act directly from the periphery. We will review the existing and emerging evidence for a role of peripheral immune cells in neurodegenerative diseases, both with and without CNS infiltration. Our focus will be on amyotrophic lateral sclerosis, but we will also compare to Alzheimer's disease and Parkinson's disease to highlight similarities or differences. Peripheral immune cells are easily accessible, and therefore may be an attractive therapeutic target for neurodegenerative diseases. Thus, understanding how these peripheral immune cells communicate with the CNS deserves deeper investigation.
Collapse
Affiliation(s)
- Félix Berriat
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Christian S Lobsiger
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Séverine Boillée
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
11
|
Peter I, Strober W. Immunological Features of LRRK2 Function and Its Role in the Gut-Brain Axis Governing Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2023; 13:279-296. [PMID: 37066923 PMCID: PMC10200211 DOI: 10.3233/jpd-230021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/20/2023] [Indexed: 04/18/2023]
Abstract
Emerging evidence implicates intestinal involvement in the onset and/or progression on the selective degeneration of dopaminergic neurons characterizing Parkinson's disease (PD). On the one hand, there are studies supporting the Braak hypothesis that holds that pathologic α-synuclein, a hallmark of PD, is secreted by enteric nerves into intestinal tissue and finds its way to the central nervous system (CNS) via retrograde movement in the vagus nerve. On the other hand, there is data showing that cells bearing leucine-rich repeat kinase 2 (LRRK2), a signaling molecule with genetic variants associated with both PD and with inflammatory bowel disease, can be activated in intestinal tissue and contribute locally to intestinal inflammation, or peripherally to PD pathogenesis via cell trafficking to the CNS. Importantly, these gut-centered factors affecting PD development are not necessarily independent of one another: they may interact and enhance their respective pathologic functions. In this review, we discuss this possibility by analysis of studies conducted in recent years focusing on the ability of LRRK2 to shape immunologic responses and the role of α-synuclein in influencing this ability.
Collapse
Affiliation(s)
- Inga Peter
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Warren Strober
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
12
|
Kim B, Suh YH, Joe E. LRRK2 decreases microglial actin dynamics by filamentous actin depolymerization and Rac1 inhibition. Anim Cells Syst (Seoul) 2022; 26:380-387. [PMID: 36605588 PMCID: PMC9809388 DOI: 10.1080/19768354.2022.2158219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
An active actin dynamic is a crucial feature of brain microglia. Here we report that LRRK2, a primary familial Parkinson's disease-associated gene, negatively regulates microglia's actin dynamics. LRRK2 depolymerized filamentous actin (F-actin) by directly binding to it or inhibiting microglia's Rac-PAK signaling. LRRK2 knockdown resulted in a reduced ruffle and enhanced lamellipodia formation of ADP-activated microglia, altering the microglia's physiological activity to vigorous migration toward damaged cells. These results suggest that LRRK2 is a negative regulator for the controlled actin dynamics in microglia.
Collapse
Affiliation(s)
- Beomsue Kim
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, Republic of Korea, Beomsue Kim Neural Circuit Research Group, Korea Brain Research Institute, Daegu41062, Republic of Korea; Eunhye Joe Department of Pharmacology; Neuroscience Graduate Program, Department of Biomedical Sciences; Center for Convergence Research of Neurological Disorders, Ajou University Schoo lof Medicine, Suwon16499, Republic of Korea
| | - Young Ho Suh
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eunhye Joe
- Department of Pharmacology, Ajou University School of Medicine, Suwon, Republic of Korea,Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Republic of Korea,Center for Convergence Research of Neurological Disorders, Ajou University Schoo lof Medicine, Suwon, Republic of Korea, Beomsue Kim Neural Circuit Research Group, Korea Brain Research Institute, Daegu41062, Republic of Korea; Eunhye Joe Department of Pharmacology; Neuroscience Graduate Program, Department of Biomedical Sciences; Center for Convergence Research of Neurological Disorders, Ajou University Schoo lof Medicine, Suwon16499, Republic of Korea
| |
Collapse
|
13
|
Kim J, Daadi EW, Oh T, Daadi ES, Daadi MM. Human Induced Pluripotent Stem Cell Phenotyping and Preclinical Modeling of Familial Parkinson's Disease. Genes (Basel) 2022; 13:1937. [PMID: 36360174 PMCID: PMC9689743 DOI: 10.3390/genes13111937] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 12/05/2022] Open
Abstract
Parkinson's disease (PD) is primarily idiopathic and a highly heterogenous neurodegenerative disease with patients experiencing a wide array of motor and non-motor symptoms. A major challenge for understanding susceptibility to PD is to determine the genetic and environmental factors that influence the mechanisms underlying the variations in disease-associated traits. The pathological hallmark of PD is the degeneration of dopaminergic neurons in the substantia nigra pars compacta region of the brain and post-mortem Lewy pathology, which leads to the loss of projecting axons innervating the striatum and to impaired motor and cognitive functions. While the cause of PD is still largely unknown, genome-wide association studies provide evidence that numerous polymorphic variants in various genes contribute to sporadic PD, and 10 to 15% of all cases are linked to some form of hereditary mutations, either autosomal dominant or recessive. Among the most common mutations observed in PD patients are in the genes LRRK2, SNCA, GBA1, PINK1, PRKN, and PARK7/DJ-1. In this review, we cover these PD-related mutations, the use of induced pluripotent stem cells as a disease in a dish model, and genetic animal models to better understand the diversity in the pathogenesis and long-term outcomes seen in PD patients.
Collapse
Affiliation(s)
- Jeffrey Kim
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
- Cell Systems and Anatomy, San Antonio, TX 78229, USA
| | - Etienne W. Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Thomas Oh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Elyas S. Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Marcel M. Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
- Cell Systems and Anatomy, San Antonio, TX 78229, USA
- Department of Radiology, Long School of Medicine, University of Texas Health at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
14
|
Suhail M, Sohrab SS, Kamal M, Azhar EI. Role of hepatitis c virus in hepatocellular carcinoma and neurological disorders: an overview. Front Oncol 2022; 12:913231. [PMID: 35965577 PMCID: PMC9372299 DOI: 10.3389/fonc.2022.913231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
The hepatitis C virus (HCV) causes serious issues, affecting 71 million people globally. The most common manifestations range from chronic hepatitis to liver cirrhosis, leading to hepatocellular carcinoma. Many mechanisms are known to play an important role in HCV-induced HCC. The interaction of viral proteins with host cells results in oxidative stress damage, liver inflammation, and irregularities in signaling pathways. These results in the activation of oncogenes and metabolic disturbances, liver fibrosis, and angiogenesis. Additionally, some non-coding RNAs (ncRNAs) and toll-like receptors have been identified and play a significant role in HCC development. This virus is also associated with impairment of the central nervous system, resulting in acute or sub-acute encephalopathy and inflammatory disorders. Neurological disorders are associated with the inflammatory responses of many cells, including microglia and astrocytes. Additionally, there are many other extrahepatic manifestations, including neurological disorders such as depression and fatigue, in 50% of infected patients. These manifestations include neuro-invasion, immune-mediated damage, neurotransmitter alterations, sensory-motor polyneuropathy, sensitivity loss, weakness of the leg, and cryoglobulinemia, which significantly results in a reduced quality of life. HCV infection may be improved using an appropriate diagnosis and direct antiviral therapy for sustained virological response. However, the success of therapy depends on the symptoms and organ damage, diagnosis, and therapeutic strategies applied. Some published reports have discussed that HCV is associated with both HCC and neurological disorders. Additionally, it has also been observed that individuals with HCC also develop neurological disorders compared with individuals with HCV alone. This review aims to provide an overview of the latest information about the relationship between HCV-induced HCC and their role in neurological disorders. Additionally, we have also discussed the progress made in the diagnosis, physio-pathological mechanisms, and strong antiviral therapies developed for HCV infection and HCC, as well as the latest advancements made in the study of the neurological disorders associated with HCV infection.
Collapse
Affiliation(s)
- Mohd Suhail
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sayed Sartaj Sohrab
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- *Correspondence: Sayed Sartaj Sohrab,
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- West China School of Nursing/Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Enzymoics Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | - Esam Ibraheem Azhar
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
15
|
Zhang M, Li C, Ren J, Wang H, Yi F, Wu J, Tang Y. The Double-Faceted Role of Leucine-Rich Repeat Kinase 2 in the Immunopathogenesis of Parkinson's Disease. Front Aging Neurosci 2022; 14:909303. [PMID: 35645775 PMCID: PMC9131027 DOI: 10.3389/fnagi.2022.909303] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/20/2022] [Indexed: 12/17/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is one of the most common causative genes in Parkinson's disease (PD). The complex structure of this multiple domains' protein determines its versatile functions in multiple physiological processes, including migration, autophagy, phagocytosis, and mitochondrial function, among others. Mounting studies have also demonstrated the role of LRRK2 in mediating neuroinflammation, the prominent hallmark of PD, and intricate functions in immune cells, such as microglia, macrophages, and astrocytes. Of those, microglia were extensively studied in PD, which serves as the resident immune cell of the central nervous system that is rapidly activated upon neuronal injury and pathogenic insult. Moreover, the activation and function of immune cells can be achieved by modulating their intracellular metabolic profiles, in which LRRK2 plays an emerging role. Here, we provide an updated review focusing on the double-faceted role of LRRK2 in regulating various cellular physiology and immune functions especially in microglia. Moreover, we will summarize the latest discovery of the three-dimensional structure of LRRK2, as well as the function and dysfunction of LRRK2 in immune cell-related pathways.
Collapse
Affiliation(s)
- Mengfei Zhang
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- Aging Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chaoyi Li
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- Aging Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Ren
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- Aging Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Huakun Wang
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- Aging Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Fang Yi
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- Aging Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Junjiao Wu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Tang
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- Aging Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
16
|
Russo I, Bubacco L, Greggio E. LRRK2 as a target for modulating immune system responses. Neurobiol Dis 2022; 169:105724. [DOI: 10.1016/j.nbd.2022.105724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/07/2022] [Accepted: 04/08/2022] [Indexed: 01/08/2023] Open
|
17
|
Shao F, Wang X, Wu H, Wu Q, Zhang J. Microglia and Neuroinflammation: Crucial Pathological Mechanisms in Traumatic Brain Injury-Induced Neurodegeneration. Front Aging Neurosci 2022; 14:825086. [PMID: 35401152 PMCID: PMC8990307 DOI: 10.3389/fnagi.2022.825086] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/21/2022] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the most common diseases in the central nervous system (CNS) with high mortality and morbidity. Patients with TBI usually suffer many sequelae in the life time post injury, including neurodegenerative disorders such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). However, the pathological mechanisms connecting these two processes have not yet been fully elucidated. It is important to further investigate the pathophysiological mechanisms underlying TBI and TBI-induced neurodegeneration, which will promote the development of precise treatment target for these notorious neurodegenerative consequences after TBI. A growing body of evidence shows that neuroinflammation is a pivotal pathological process underlying chronic neurodegeneration following TBI. Microglia, as the immune cells in the CNS, play crucial roles in neuroinflammation and many other CNS diseases. Of interest, microglial activation and functional alteration has been proposed as key mediators in the evolution of chronic neurodegenerative pathology following TBI. Here, we review the updated studies involving phenotypical and functional alterations of microglia in neurodegeneration after injury, survey key molecules regulating the activities and functional responses of microglia in TBI pathology, and explore their potential implications to chronic neurodegeneration after injury. The work will give us a comprehensive understanding of mechanisms driving TBI-related neurodegeneration and offer novel ideas of developing corresponding prevention and treatment strategies for this disease.
Collapse
Affiliation(s)
- Fangjie Shao
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haijian Wu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Qun Wu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- *Correspondence: Qun Wu,
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Brain Research Institute, Zhejiang University, Hangzhou, China
- Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, China
- Jianmin Zhang,
| |
Collapse
|
18
|
Does the Expression and Epigenetics of Genes Involved in Monogenic Forms of Parkinson’s Disease Influence Sporadic Forms? Genes (Basel) 2022; 13:genes13030479. [PMID: 35328033 PMCID: PMC8951612 DOI: 10.3390/genes13030479] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 12/25/2022] Open
Abstract
Parkinson’s disease (PD) is a disorder characterized by a triad of motor symptoms (akinesia, rigidity, resting tremor) related to loss of dopaminergic neurons mainly in the Substantia nigra pars compacta. Diagnosis is often made after a substantial loss of neurons has already occurred, and while dopamine replacement therapies improve symptoms, they do not modify the course of the disease. Although some biological mechanisms involved in the disease have been identified, such as oxidative stress and accumulation of misfolded proteins, they do not explain entirely PD pathophysiology, and a need for a better understanding remains. Neurodegenerative diseases, including PD, appear to be the result of complex interactions between genetic and environmental factors. The latter can alter gene expression by causing epigenetic changes, such as DNA methylation, post-translational modification of histones and non-coding RNAs. Regulation of genes responsible for monogenic forms of PD may be involved in sporadic PD. This review will focus on the epigenetic mechanisms regulating their expression, since these are the genes for which we currently have the most information available. Despite technical challenges, epigenetic epidemiology offers new insights on revealing altered biological pathways and identifying predictive biomarkers for the onset and progression of PD.
Collapse
|
19
|
Li KL, Huang HY, Ren H, Yang XL. Role of exosomes in the pathogenesis of inflammation in Parkinson's disease. Neural Regen Res 2022; 17:1898-1906. [PMID: 35142665 PMCID: PMC8848593 DOI: 10.4103/1673-5374.335143] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Inflammatory responses, including glial cell activation and peripheral immune cell infiltration, are involved in the pathogenesis of Parkinson’s disease (PD). These inflammatory responses appear to be closely related to the release of extracellular vesicles, such as exosomes. However, the relationships among different forms of glial cell activation, synuclein dysregulation, mitochondrial dysfunction, and exosomes are complicated. This review discusses the multiple roles played by exosomes in PD-associated inflammation and concludes that exosomes can transport toxic α-synuclein oligomers to immature neurons and into the extracellular environment, inducing the oligomerization of α-synuclein in normal neurons. Misfolded α-synuclein causes microglia and astrocytes to activate and secrete exosomes. Glial cell-derived exosomes participate in communications between glial cells and neurons, triggering anti-stress and anti-inflammatory responses, in addition to axon growth. The production and release of mitochondrial vesicles and exosomes establish a new mechanism for linking mitochondrial dysfunction to systemic inflammation associated with PD. Given the relevance of exosomes as mediators of neuron-glia communication in neuroinflammation and neuropathogenesis, new targeted treatment strategies are currently being developed that use these types of extracellular vesicles as drug carriers. Exosome-mediated inflammation may be a promising target for intervention in PD patients.
Collapse
Affiliation(s)
- Ke-Lu Li
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Hong-Yan Huang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hui Ren
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Xing-Long Yang
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| |
Collapse
|
20
|
Li J, Wu M, Gong Y, Tang J, Shen J, Xu L, Dang B, Chen G. Inhibition of LRRK2-Rab10 Pathway Improves Secondary Brain Injury After Surgical Brain Injury in Rats. Front Surg 2022; 8:749310. [PMID: 35071308 PMCID: PMC8766807 DOI: 10.3389/fsurg.2021.749310] [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: 07/29/2021] [Accepted: 12/13/2021] [Indexed: 11/28/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is considered as a potential target for the treatment of Parkinson's disease. This protein is expressed in the brain and has been associated with various diseases and lysosomal maintenance. Rab10 is a member of the Rab protein GTPase family that has been recently shown to be a kinase substrate of LRRK2. In addition, LRRK2 and its kinase substrate Rab10 constitute a key stress response pathway during lysosomal overload stress. This study aimed to investigate the potential role and mechanism underlying LRRK2 and its kinase substrate Rab10 involving surgical brain injury (SBI). One hundred and forty-four male Sprague-Dawley rats were examined using an SBI model, and some had received the LRRK2-specific inhibitor PF-06447475. Thereafter, western blotting, immunofluorescence, brain water content analysis, neuronal apoptosis assay, and neurological score analysis were conducted. The results showed that after SBI, LRRK2 and phosphorylated Rab10 (p-Rab10) expression in neuronal cells were upregulated, and administration of PF-06447475 significantly reduced neuronal apoptosis, neuroinflammation, and brain water content 12 h after SBI and improved neurological deficit 72 h after SBI, which is related to the decreased expression of LRRK2 and p-Rab10, and the lessening of lysosomal overload stress. Our research suggests that the inhibition of LRRK2 can effectively interfere with the role of p-Rab10 in promoting the secretion of lysosomal hydrolase in lysosomal overload stress after SBI, thereby reducing neuronal apoptosis and inflammation after SBI and playing a major role in brain protection.
Collapse
Affiliation(s)
- Jie Li
- Department of Intensive Care Unit, Zhangjiagang Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Suzhou, China
| | - Muyao Wu
- Department of Rehabilitation, Zhangjiagang Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Suzhou, China
| | - Yating Gong
- Department of Rehabilitation, Zhangjiagang Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Suzhou, China
| | - Jiafeng Tang
- Department of Rehabilitation, Zhangjiagang Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Suzhou, China
| | - Jinchao Shen
- Department of Anesthesiology, Zhangjiagang Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Suzhou, China
| | - Li Xu
- Department of Intensive Care Unit, Zhangjiagang Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Suzhou, China
- *Correspondence: Li Xu
| | - Baoqi Dang
- Department of Rehabilitation, Zhangjiagang Traditional Chinese Medicine Hospital, Nanjing University of Chinese Medicine, Suzhou, China
- Baoqi Dang
| | - Gang Chen
- Department of Neurosurgery and Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
21
|
Nandi SK, Singh D, Upadhay J, Gupta N, Dhiman N, Mittal SK, Mahindroo N. Identification of tear-based protein and non-protein biomarkers: Its application in diagnosis of human diseases using biosensors. Int J Biol Macromol 2021; 193:838-846. [PMID: 34728300 DOI: 10.1016/j.ijbiomac.2021.10.198] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/30/2021] [Accepted: 10/26/2021] [Indexed: 12/27/2022]
Abstract
Discovery of robust, selective and specific biomarkers are important for early diagnosis and monitor progression of human diseases. Eye being a common target for several human diseases, vision impediment and complications are often associated with systemic and ocular diseases. Tears are bodily fluids that are closest to eye and are rich in protein content and other metabolites. As a biomarker repository, it advantages over other bodily fluids due to the ability to collect it non-invasively. In this review, we highlight some recent advancements in identification of tear-based protein biomarkers like lacryglobin and cystatin SA for cancer; interleukin-6 and immunoglobulin-A antibody for COVID-19; tau, amyloid-β-42 and lysozyme-C for Alzheimer's disease; peroxiredoxin-6 and α-synuclein for Parkinson's disease; kallikrein, angiotensin converting enzyme and lipocalin-1 for glaucoma; lactotransferrin and lipophilin-A for diabetic retinopathy and zinc-alpha-2 glycoprotein-1, prolactin and calcium binding protein-A4 for eye thyroid disease. We also discussed identification of tear based non-protein biomarkers like lysophospholipids and acetylcarnitine for glaucoma, 8-hydroxy-2'-deoxyquanosine and malondialdehyde for thyroid eye disease. We elucidate technological advancement in developing tear-based biosensors for diagnosis and monitoring diseases such as diabetes, diabetic retinopathy and Alzheimer's disease. Altogether, the study of tears as potential biomarkers for early diagnosis of human diseases is promising.
Collapse
Affiliation(s)
- Sandip K Nandi
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand 248007, India.
| | - Deepanmol Singh
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand 248007, India
| | - Jyoti Upadhay
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand 248007, India
| | - Neeti Gupta
- Department of Ophthalmology, All India Institute of Medical Sciences, Rishikesh 249203, India
| | - Nayan Dhiman
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand 248007, India
| | - Sanjeev Kumar Mittal
- Department of Ophthalmology, All India Institute of Medical Sciences, Rishikesh 249203, India
| | - Neeraj Mahindroo
- School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand 248007, India.
| |
Collapse
|
22
|
Khan SS, Sobu Y, Dhekne HS, Tonelli F, Berndsen K, Alessi DR, Pfeffer SR. Pathogenic LRRK2 control of primary cilia and Hedgehog signaling in neurons and astrocytes of mouse brain. eLife 2021; 10:67900. [PMID: 34658337 PMCID: PMC8550758 DOI: 10.7554/elife.67900] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 10/17/2021] [Indexed: 12/11/2022] Open
Abstract
Activating LRRK2 mutations cause Parkinson’s disease, and pathogenic LRRK2 kinase interferes with ciliogenesis. Previously, we showed that cholinergic interneurons of the dorsal striatum lose their cilia in R1441C LRRK2 mutant mice (Dhekne et al., 2018). Here, we show that cilia loss is seen as early as 10 weeks of age in these mice and also in two other mouse strains carrying the most common human G2019S LRRK2 mutation. Loss of the PPM1H phosphatase that is specific for LRRK2-phosphorylated Rab GTPases yields the same cilia loss phenotype seen in mice expressing pathogenic LRRK2 kinase, strongly supporting a connection between Rab GTPase phosphorylation and cilia loss. Moreover, astrocytes throughout the striatum show a ciliation defect in all LRRK2 and PPM1H mutant models examined. Hedgehog signaling requires cilia, and loss of cilia in LRRK2 mutant rodents correlates with dysregulation of Hedgehog signaling as monitored by in situ hybridization of Gli1 and Gdnf transcripts. Dopaminergic neurons of the substantia nigra secrete a Hedgehog signal that is sensed in the striatum to trigger neuroprotection; our data support a model in which LRRK2 and PPM1H mutant mice show altered responses to critical Hedgehog signals in the nigrostriatal pathway.
Collapse
Affiliation(s)
- Shahzad S Khan
- Department of Biochemistry, Stanford University School of Medicine, Stanford, United States.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, United States
| | - Yuriko Sobu
- Department of Biochemistry, Stanford University School of Medicine, Stanford, United States.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, United States
| | - Herschel S Dhekne
- Department of Biochemistry, Stanford University School of Medicine, Stanford, United States
| | - Francesca Tonelli
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, United States.,MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, United Kingdom
| | - Kerryn Berndsen
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, United States.,MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, United Kingdom
| | - Dario R Alessi
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, United States.,MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, United Kingdom
| | - Suzanne R Pfeffer
- Department of Biochemistry, Stanford University School of Medicine, Stanford, United States.,Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, United States
| |
Collapse
|
23
|
Naoi M, Maruyama W, Shamoto-Nagai M. Disease-modifying treatment of Parkinson's disease by phytochemicals: targeting multiple pathogenic factors. J Neural Transm (Vienna) 2021; 129:737-753. [PMID: 34654977 DOI: 10.1007/s00702-021-02427-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022]
Abstract
Parkinson's disease is characterized by typical motor symptoms, loss of dopamine neurons in the substantia nigra, and accumulation of Lewy body composed of mutated α-synuclein. However, now it is considered as a generalized disease with multiple pathological features. Present available treatments can ameliorate symptoms at least for a while, but only a few therapies could delay progressive neurodegeneration of dopamine neurons. Lewy body accumulates in peripheral tissues many years before motor dysfunction becomes manifest, suggesting that disease-modifying therapy should start earlier during the premotor stage. Long-termed regulation of lifestyle, diet and supplement of nutraceuticals may be possible ways for the disease-modification. Diet can reduce the incidence of Parkinson's disease and phytochemicals, major bioactive ingredients of herbs and plant food, modulate multiple pathogenic factors and exert neuroprotective effects in preclinical studies. This review presents mechanisms underlying neuroprotection of phytochemicals against neuronal cell death and α-synuclein toxicity in Parkinson's disease. Phytochemicals are antioxidants, maintain mitochondrial function and homeostasis, prevent intrinsic apoptosis and neuroinflammation, activate cellular signal pathways to induce anti-apoptotic and pro-survival genes, such as Bcl-2 protein family and neurotrophic factors, and promote cleavage of damaged mitochondria and α-synuclein aggregates. Phytochemicals prevent α-synuclein oligomerization and aggregation, and dissolve preformed α-synuclein aggregates. Novel neuroprotective agents are expected to develop based on the scaffold of phytochemicals permeable across the blood-brain-barrier, to increase the bioavailability, ameliorate brain dysfunction and prevent neurodegeneration.
Collapse
Affiliation(s)
- Makoto Naoi
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 320-0195, Japan.
| | - Wakako Maruyama
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 320-0195, Japan
| | - Masayo Shamoto-Nagai
- Department of Health and Nutrition, Faculty of Psychological and Physical Science, Aichi Gakuin University, 12 Araike, Iwasaki-cho, Nisshin, Aichi, 320-0195, Japan
| |
Collapse
|
24
|
Rosenbusch KE, Oun A, Sanislav O, Lay ST, Keizer-Gunnink I, Annesley SJ, Fisher PR, Dolga AM, Kortholt A. A Conserved Role for LRRK2 and Roco Proteins in the Regulation of Mitochondrial Activity. Front Cell Dev Biol 2021; 9:734554. [PMID: 34568343 PMCID: PMC8455996 DOI: 10.3389/fcell.2021.734554] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/16/2021] [Indexed: 01/02/2023] Open
Abstract
Parkinson's Disease (PD) is the second most common neurodegenerative disease world-wide. Mutations in the multidomain protein Leucine Rich Repeat Kinase 2 (LRRK2) are the most frequent cause of hereditary PD. Furthermore, recent data suggest that independent of mutations, increased kinase activity of LRRK2 plays an essential role in PD pathogenesis. Isolated mitochondria of tissue samples from PD patients carrying LRRK2 mutations display a significant impairment of mitochondrial function. However, due to the complexity of the mitochondrial signaling network, the role of LRRK2 in mitochondrial metabolism is still not well understood. Previously we have shown that D. discoideum Roco4 is a suitable model to study the activation mechanism of LRRK2 in vivo. To get more insight in the LRRK2 pathways regulating mitochondrial activity we used this Roco4 model system in combination with murine RAW macrophages. Here we show that both Dictyostelium roco4 knockout and cells expressing PD-mutants show behavioral and developmental phenotypes that are characteristic for mitochondrial impairment. Mitochondrial activity measured by Seahorse technology revealed that the basal respiration of D. discoideum roco4- cells is significantly increased compared to the WT strain, while the basal and maximal respiration values of cells overexpressing Roco4 are reduced compared to the WT strain. Consistently, LRRK2 KO RAW 264.7 cells exhibit higher maximal mitochondrial respiration activity compared to the LRRK2 parental RAW264.7 cells. Measurement on isolated mitochondria from LRRK2 KO and parental RAW 264.7 cells revealed no difference in activity compared to the parental cells. Furthermore, neither D. discoideum roco4- nor LRRK2 KO RAW 264.7 showed a difference in either the number or the morphology of mitochondria compared to their respective parental strains. This suggests that the observed effects on the mitochondrial respiratory in cells are indirect and that LRRK2/Roco proteins most likely require other cytosolic cofactors to elicit mitochondrial effects.
Collapse
Affiliation(s)
| | - Asmaa Oun
- Department of Cell Biochemistry, University of Groningen, Groningen, Netherlands.,Groningen Research Institute of Pharmacy (GRIP), Molecular Pharmacology XB10, Groningen, Netherlands.,Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Oana Sanislav
- Department of Physiology Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Sui T Lay
- Department of Physiology Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Ineke Keizer-Gunnink
- Department of Physiology Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Sarah J Annesley
- Department of Physiology Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Paul R Fisher
- Department of Physiology Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Amalia M Dolga
- Groningen Research Institute of Pharmacy (GRIP), Molecular Pharmacology XB10, Groningen, Netherlands
| | - Arjan Kortholt
- Department of Cell Biochemistry, University of Groningen, Groningen, Netherlands.,Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
25
|
Fenner BM, Fenner ME, Prowse N, Hayley SP. LRRK2 and WAVE2 regulate microglial-transition through distinct morphological phenotypes to induce neurotoxicity in a novel two-hit in vitro model of neurodegeneration. J Cell Physiol 2021; 237:1013-1032. [PMID: 34543438 DOI: 10.1002/jcp.30588] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 12/16/2022]
Abstract
We report a novel in vitro classification system that tracks microglial activation state and their potential neurotoxicity. Mixed live-cell imaging was used to characterize transition through distinct morphological phenotypes, production of reactive oxygen species (ROS), formation of reactive microglial aggregates, and subsequent cytokine production. Transwell cultures were used to determine microglial migration (control and lipopolysaccharide (LPS) treated) to glutamate pre-stressed or healthy neurons. This two-hit paradigm was developed to model the vast evidence that neurodegenerative conditions, like Parkinson's disease (PD), may stem from the collective impact of multiple environmental stressors. We found that healthy neurons were resistant to microglial-mediated inflammation, whereas glutamate pre-stressed neurons were highly susceptible and in fact, appeared to recruit microglia. The LPS treated microglia progressed through distinct morphological states and expressed high levels of ROS and formed large cellular aggregates. Recent evidence implicates leucine-rich repeat kinase 2 (LRRK2) as an important player in the microglial inflammatory state, as well as in the genesis of PD. We found that inhibition of the LRRK2 signaling pathway using the kinase inhibitor cis-2,6-dimethyl-4-(6-(5-(1-methylcyclopropoxy)-1H-indazol-3-yl)pyrimidin-4-yl)morpholine (MLi2) or inhibition of the actin regulatory protein, Wiskott-Aldrich syndrome family Verprolin-homologous Protein-2 (WAVE2), stunted microglial activation and prevented neurotoxicity. Furthermore, inhibition of LRRK2 kinase activity reduced pro-inflammatory chemokines including MIP-2, CRG-2, and RANTES. These data together support the notion that LRRK2 and WAVE2 are important mediators of cytokine production and cytoskeletal rearrangement necessary for microglial-induced neurotoxicity. Furthermore, our model demonstrated unique microglial phenotypic changes that might be mechanistically important for better understanding neuron-microglial crosstalk.
Collapse
Affiliation(s)
- Barbara M Fenner
- Department of Biology, King's College, Wilkes-Barre, Pennsylvania, USA
| | - Mark E Fenner
- Fenner Training and Consulting, LLC, Kingston, Pennsylvania, USA
| | - Natalie Prowse
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Shawn P Hayley
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
26
|
Nazish I, Arber C, Piers TM, Warner TT, Hardy JA, Lewis PA, Pocock JM, Bandopadhyay R. Abrogation of LRRK2 dependent Rab10 phosphorylation with TLR4 activation and alterations in evoked cytokine release in immune cells. Neurochem Int 2021; 147:105070. [PMID: 34004238 PMCID: PMC7610942 DOI: 10.1016/j.neuint.2021.105070] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/11/2021] [Indexed: 02/08/2023]
Abstract
LRRK2 protein is expressed prominently in immune cells, cell types whose contribution to LRRK2-associated genetic Parkinson's disease (PD) is increasingly being recognised. We investigated the effect of inflammatory stimuli using RAW264.7 murine macrophage cells as model systems. A detailed time course of TLR2 and TLR4 stimulation was investigated through measuring LRRK2 phosphorylation at its specific phospho-sites, and Rab8 and Rab10 phosphorylation together with cytokine release following treatment with LPS and zymosan. LRRK2 phosphorylation at Ser935, Ser955 and Ser973 was increased significantly over untreated conditions at 4-24h in both WT-LRRK2 and T1348N-LRRK2 cell lines to similar extents although levels of Ser910 phosphorylation were maintained at higher levels throughout. Importantly we demonstrate that LPS stimulation significantly decreased phospho-Rab10 but not phospho-Rab8 levels over 4-24h in both WT-LRRK2 and T1348N-LRRK2 cell lines. The dephosphorylation of Rab10 was not attributed to its specific phosphatase, PPM1H as the levels remained unaltered with LPS treatment. MAPK phosphorylation occurred prior to LRRK2 phosphorylation which was validated by blocking TLR4 and TLR2 receptors with TAK242 or Sparstolonin B respectively. A significant decrease in basal level of TNFα release was noted in both T1348N-LRRK2 and KO-LRRK2 cell lines at 48h compared to WT-LRRK2 cell line, however LPS and zymosan treatment did not cause any significant alteration in the TNFα and IL-6 release between the three cell lines. In contrast, LPS and zymosan caused significantly lower IL-10 release in T1348N-LRRK2 and KO-LRRK2 cell lines. A significant decrease in phospho-Rab10 levels was also confirmed in human IPS-derived macrophages with TLR4 activation. Our data demonstrates for the first time that LRRK2-dependent Rab10 phosphorylation is modulated by LPS stimulation, and that cytokine release may be influenced by the status of LRRK2. These data provide further insights into the function of LRRK2 in immune response, and has relevance for understanding cellular dysfunctions when developing LRRK2-based inhibitors for clinical treatment.
Collapse
Affiliation(s)
- Iqra Nazish
- Reta Lila Weston Institute and Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, 1 Wakefield Street, WC1N 1PJ, UK
| | - Charles Arber
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, 1 Wakefield Street, WC1N 1PJ, UK
| | - Thomas M. Piers
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, 1 Wakefield Street, WC1N 1PJ, UK
| | - Thomas T. Warner
- Reta Lila Weston Institute and Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, 1 Wakefield Street, WC1N 1PJ, UK
| | - John A. Hardy
- Reta Lila Weston Institute and Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, 1 Wakefield Street, WC1N 1PJ, UK,Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, 1 Wakefield Street, WC1N 1PJ, UK,Queen Square Genomics, UCL Dementia Research Institute, Wing 1.2 Cruciform Building, Gower Street, London, WC1E 6BT, UK
| | - Patrick A. Lewis
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, 1 Wakefield Street, WC1N 1PJ, UK,Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - Jennifer M. Pocock
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, 1 Wakefield Street, WC1N 1PJ, UK
| | - Rina Bandopadhyay
- Reta Lila Weston Institute and Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, 1 Wakefield Street, WC1N 1PJ, UK.
| |
Collapse
|
27
|
Leucine-rich repeat kinase 2-related functions in GLIA: an update of the last years. Biochem Soc Trans 2021; 49:1375-1384. [PMID: 33960369 DOI: 10.1042/bst20201092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/13/2022]
Abstract
Missense mutations in the leucine-rich repeat kinase-2 (LRRK2) gene represent the most common cause of autosomal dominant Parkinson's disease (PD). In the years LRRK2 has been associated with several organelles and related pathways in cell. However, despite the significant amount of research done in the past decade, the contribution of LRRK2 mutations to PD pathogenesis remains unknown. Growing evidence highlights that LRRK2 controls multiple processes in brain immune cells, microglia and astrocytes, and suggests that deregulated LRRK2 activity in these cells, due to gene mutation, might be directly associated with pathological mechanisms underlying PD. In this brief review, we recapitulate and update the last LRRK2 functions dissected in microglia and astrocytes. Moreover, we discuss how dysfunctions of LRRK2-related pathways may impact glia physiology and their cross-talk with neurons, thus leading to neurodegeneration and progression of PD.
Collapse
|
28
|
Goveas L, Mutez E, Chartier-Harlin MC, Taymans JM. Mind the Gap: LRRK2 Phenotypes in the Clinic vs. in Patient Cells. Cells 2021; 10:981. [PMID: 33922322 PMCID: PMC8145309 DOI: 10.3390/cells10050981] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/14/2022] Open
Abstract
Mutations in the Parkinson's disease (PD) protein Leucine Rich Repeat Kinase 2 (LRRK2) have been under study for more than 15 years and our understanding of the cellular phenotypes for the pathogenic mutant forms of LRRK2 has significantly advanced. In parallel to research on LRRK2 mutations in experimental systems, clinical characterization of patients carrying LRRK2 mutations has advanced, as has the analysis of cells that are derived from these patients, including fibroblasts, blood-derived cells, or cells rendered pluripotent. Under the hypothesis that patient clinical phenotypes are a consequence of a cascade of underlying molecular mechanisms gone astray, we currently have a unique opportunity to compare findings from patients and patient-derived cells to ask the question of whether the clinical phenotype of LRRK2 Parkinson's disease and cellular phenotypes of LRRK2 patient-derived cells may be mutually informative. In this review, we aim to summarize the available information on phenotypes of LRRK2 mutations in the clinic, in patient-derived cells, and in experimental models in order to better understand the relationship between the three at the molecular and cellular levels and identify trends and gaps in correlating the data.
Collapse
Affiliation(s)
- Liesel Goveas
- UMR-S 1172—LilNCog—Lille Neuroscience & Cognition, Université de Lille, Inserm, CHU Lille, F-59000 Lille, France; (L.G.); (E.M.)
| | - Eugénie Mutez
- UMR-S 1172—LilNCog—Lille Neuroscience & Cognition, Université de Lille, Inserm, CHU Lille, F-59000 Lille, France; (L.G.); (E.M.)
- Neurology and Movement Disorders Department, CHU Lille University Hospital, F-59000 Lille, France
| | - Marie-Christine Chartier-Harlin
- UMR-S 1172—LilNCog—Lille Neuroscience & Cognition, Université de Lille, Inserm, CHU Lille, F-59000 Lille, France; (L.G.); (E.M.)
| | - Jean-Marc Taymans
- UMR-S 1172—LilNCog—Lille Neuroscience & Cognition, Université de Lille, Inserm, CHU Lille, F-59000 Lille, France; (L.G.); (E.M.)
| |
Collapse
|
29
|
Prasuhn J, Brüggemann N. Genotype-driven therapeutic developments in Parkinson's disease. Mol Med 2021; 27:42. [PMID: 33874883 PMCID: PMC8056568 DOI: 10.1186/s10020-021-00281-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/12/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Remarkable advances have been reached in the understanding of the genetic basis of Parkinson's disease (PD), with the identification of monogenic causes (mPD) and a plethora of gene loci leading to an increased risk for idiopathic PD. The expanding knowledge and subsequent identification of genetic contributions fosters the understanding of molecular mechanisms leading to disease development and progression. Distinct pathways involved in mitochondrial dysfunction, oxidative stress, and lysosomal function have been identified and open a unique window of opportunity for individualized treatment approaches. These genetic findings have led to an imminent progress towards pathophysiology-targeted clinical trials and potentially disease-modifying treatments in the future. MAIN BODY OF THE MANUSCRIPT In this review article we will summarize known genetic contributors to the pathophysiology of Parkinson's disease, the molecular mechanisms leading to disease development, and discuss challenges and opportunities in clinical trial designs. CONCLUSIONS The future success of clinical trials in PD is mainly dependent on reliable biomarker development and extensive genetic testing to identify genetic cases. Whether genotype-dependent stratification of study participants will extend the potential application of new drugs will be one major challenge in conceptualizing clinical trials. However, the latest developments in genotype-driven treatments will pave the road to individualized pathophysiology-based therapies in the future.
Collapse
Affiliation(s)
- Jannik Prasuhn
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
- Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Norbert Brüggemann
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany.
- Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
30
|
Herrick MK, Tansey MG. Is LRRK2 the missing link between inflammatory bowel disease and Parkinson's disease? NPJ Parkinsons Dis 2021; 7:26. [PMID: 33750819 PMCID: PMC7943592 DOI: 10.1038/s41531-021-00170-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/28/2021] [Indexed: 12/14/2022] Open
Abstract
Links that implicate the gastrointestinal system in Parkinson's disease (PD) pathogenesis and progression have become increasingly common. PD shares several similarities with Crohn's disease (CD). Intestinal inflammation is common in both PD and CD and is hypothesized to contribute to PD neuropathology. Mutations in leucine-rich repeat kinase 2 (LRRK2) are one of the greatest genetic contributors to PD. Variants in LRRK2 have also been associated with increased incidence of CD. Since its discovery, LRRK2 has been studied intensely in neurons, despite multiple lines of evidence showing that LRRK2 is highly expressed in immune cells. Based on the fact that higher levels of LRRK2 are detectable in inflamed colonic tissue from CD patients and in peripheral immune cells from sporadic PD patients relative to matched controls, we posit that LRRK2 regulates inflammatory processes. Therefore, LRRK2 may sit at a crossroads whereby gut inflammation and higher LRRK2 levels in CD may be a biomarker of increased risk for sporadic PD and/or may represent a tractable therapeutic target in inflammatory diseases that increase risk for PD. Here we will focus on reviewing how PD and CD share overlapping phenotypes, particularly in terms of LRRK2 in the context of the immune system, that could be targeted in future therapies.
Collapse
Affiliation(s)
- Mary K Herrick
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease at The University of Florida College of Medicine, Gainesville, FL, USA
| | - Malú G Tansey
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease at The University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
31
|
A LRRK2 GTP Binding Inhibitor, 68, Reduces LPS-Induced Signaling Events and TNF-α Release in Human Lymphoblasts. Cells 2021; 10:cells10020480. [PMID: 33672296 PMCID: PMC7926966 DOI: 10.3390/cells10020480] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/05/2021] [Accepted: 02/16/2021] [Indexed: 12/19/2022] Open
Abstract
Mutations in the leucine-rich repeat kinase-2 (LRRK2) gene cause autosomal-dominant Parkinson’s disease (PD) and contribute to sporadic PD. Common genetic variation in LRRK2 modifies susceptibility to immunological disorders including Crohn’s disease and leprosy. Previous studies have reported that LRRK2 is expressed in B lymphocytes and macrophages, suggesting a role for LRRK2 in immunological functions. In this study, we characterized the LRRK2 protein expression and phosphorylation using human lymphoblasts. Lipopolysaccharide (LPS), a proinflammatory agent, induced the increase of LRRK2 expression and kinase activities in human lymphoblasts in a time-dependent manner. Moreover, LPS activated the Toll-like receptor (TLR) signaling pathway, increased TRAF6/LRRK2 interaction, and elevated the phosphorylation levels of MAPK (JNK1/2, p38, and ERK1/2) and IkBα. Treatment with LRRK2 inhibitor 68 reduced LPS-induced TRAF6/LRRK2 interaction and MAPK and IkBα phosphorylation, thereby reducing TNF-α secretion. These results indicate that LRRK2 is actively involved in proinflammatory responses in human lymphoblasts, and inhibition of GTP binding by 68 results in an anti-inflammation effect against proinflammatory stimuli. These findings not only provide novel insights into the mechanisms of LRRK2-linked immune and inflammatory responses in B-cell-like lymphoblasts, but also suggest that 68 may also have potential therapeutic value for LRRK2-linked immunological disorders.
Collapse
|
32
|
Zhang X, Shao Z, Xu S, Liu Q, Liu C, Luo Y, Jin L, Li S. Immune Profiling of Parkinson's Disease Revealed Its Association With a Subset of Infiltrating Cells and Signature Genes. Front Aging Neurosci 2021; 13:605970. [PMID: 33633562 PMCID: PMC7899990 DOI: 10.3389/fnagi.2021.605970] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/19/2021] [Indexed: 11/25/2022] Open
Abstract
Parkinson’s disease (PD) is an age-related and second most common neurodegenerative disorder. In recent years, increasing evidence revealed that peripheral immune cells might be able to infiltrate into brain tissues, which could arouse neuroinflammation and aggravate neurodegeneration. This study aimed to illuminate the landscape of peripheral immune cells and signature genes associated with immune infiltration in PD. Several transcriptomic datasets of substantia nigra (SN) from the Gene Expression Omnibus (GEO) database were separately collected as training cohort, testing cohort, and external validation cohort. The immunoscore of each sample calculated by single-sample gene set enrichment analysis was used to reflect the peripheral immune cell infiltration and to identify the differential immune cell types between PD and healthy participants. According to receiver operating characteristic (ROC) curve analysis, the immunoscore achieved an overall accuracy of the area under the curve (AUC) = 0.883 in the testing cohort, respectively. The immunoscore displayed good performance in the external validation cohort with an AUC of 0.745. The correlation analysis and logistic regression analysis were used to analyze the correlation between immune cells and PD, and mast cell was identified most associated with the occurrence of PD. Additionally, increased mast cells were also observed in our in vivo PD model. Weighted gene co-expression network analysis (WGCNA) was used to selected module genes related to a mast cell. The least absolute shrinkage and selection operator (LASSO) analysis and random-forest analysis were used to analyze module genes, and two hub genes RBM3 and AGTR1 were identified as associated with mast cells in the training cohort. The expression levels of RBM3 and AGTR1 in these cohorts and PD models revealed that these hub genes were significantly downregulated in PD. Moreover, the expression trend of the aforementioned two genes differed in mast cells and dopaminergic (DA) neurons. In conclusion, this study not only exhibited a landscape of immune infiltrating patterns in PD but also identified mast cells and two hub genes associated with the occurrence of PD, which provided potential therapeutic targets for PD patients (PDs).
Collapse
Affiliation(s)
- Xi Zhang
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Neurology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhihua Shao
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Sutong Xu
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiulu Liu
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chenming Liu
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuping Luo
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China.,Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lingjing Jin
- Department of Neurology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Siguang Li
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Neurology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China.,Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopedic Department of Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Pathological Functions of LRRK2 in Parkinson's Disease. Cells 2020; 9:cells9122565. [PMID: 33266247 PMCID: PMC7759975 DOI: 10.3390/cells9122565] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
Mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) are common genetic risk factors for both familial and sporadic Parkinson’s disease (PD). Pathogenic mutations in LRRK2 have been shown to induce changes in its activity, and abnormal increase in LRRK2 kinase activity is thought to contribute to PD pathology. The precise molecular mechanisms underlying LRRK2-associated PD pathology are far from clear, however the identification of LRRK2 substrates and the elucidation of cellular pathways involved suggest a role of LRRK2 in microtubule dynamics, vesicular trafficking, and synaptic transmission. Moreover, LRRK2 is associated with pathologies of α-synuclein, a major component of Lewy bodies (LBs). Evidence from various cellular and animal models supports a role of LRRK2 in the regulation of aggregation and propagation of α-synuclein. Here, we summarize our current understanding of how pathogenic mutations dysregulate LRRK2 and discuss the possible mechanisms leading to neurodegeneration.
Collapse
|
34
|
Feng YK, Wu QL, Peng YW, Liang FY, You HJ, Feng YW, Li G, Li XJ, Liu SH, Li YC, Zhang Y, Pei Z. Oral P. gingivalis impairs gut permeability and mediates immune responses associated with neurodegeneration in LRRK2 R1441G mice. J Neuroinflammation 2020; 17:347. [PMID: 33213462 PMCID: PMC7677837 DOI: 10.1186/s12974-020-02027-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022] Open
Abstract
Background The R1441G mutation in the leucine-rich repeat kinase 2 (LRRK2) gene results in late-onset Parkinson’s disease (PD). Peripheral inflammation and gut microbiota are closely associated with the pathogenesis of PD. Chronic periodontitis is a common type of peripheral inflammation, which is associated with PD. Porphyromonas gingivalis (Pg), the most common bacterium causing chronic periodontitis, can cause alteration of gut microbiota. It is not known whether Pg-induced dysbiosis plays a role in the pathophysiology of PD. Methods In this study, live Pg were orally administrated to animals, three times a week for 1 month. Pg-derived lipopolysaccharide (LPS) was used to stimulate mononuclear cells in vitro. The effects of oral Pg administration on the gut and brain were evaluated through behaviors, morphology, and cytokine expression. Results Dopaminergic neurons in the substantia nigra were reduced, and activated microglial cells were increased in R1441G mice given oral Pg. In addition, an increase in mRNA expression of tumor necrosis factor (TNF-α) and interleukin-1β (IL-1β) as well as protein level of α-synuclein together with a decrease in zonula occludens-1 (Zo-1) was detected in the colon in Pg-treated R1441G mice. Furthermore, serum interleukin-17A (IL-17A) and brain IL-17 receptor A (IL-17RA) were increased in Pg-treated R1441G mice. Conclusions These findings suggest that oral Pg-induced inflammation may play an important role in the pathophysiology of LRRK2-associated PD. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-020-02027-5.
Collapse
Affiliation(s)
- Yu-Kun Feng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China.,Department of Neurology, Hainan General Hospital; Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, Hainan, China
| | - Qiong-Li Wu
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yan-Wen Peng
- The Biotherapy Center, the Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Feng-Yin Liang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Hua-Jing You
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Yi-Wei Feng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China.,Department of Neurology, Huashan Hospital, Fudan University, Shanghai, 200000, China
| | - Ge Li
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, 510663, Guangdong, China
| | - Xue-Jiao Li
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, 510663, Guangdong, China
| | - Shu-Hua Liu
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, 510663, Guangdong, China
| | - Yong-Chao Li
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, 510663, Guangdong, China
| | - Yu Zhang
- Guangdong Laboratory Animals Monitoring Institute, Guangdong Provincial Key Laboratory of Laboratory Animals, Guangzhou, 510663, Guangdong, China
| | - Zhong Pei
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China.
| |
Collapse
|
35
|
Marogianni C, Sokratous M, Dardiotis E, Hadjigeorgiou GM, Bogdanos D, Xiromerisiou G. Neurodegeneration and Inflammation-An Interesting Interplay in Parkinson's Disease. Int J Mol Sci 2020; 21:E8421. [PMID: 33182554 PMCID: PMC7697354 DOI: 10.3390/ijms21228421] [Citation(s) in RCA: 204] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder, caused by, so far, unknown pathogenetic mechanisms. There is no doubt that pro-inflammatory immune-mediated mechanisms are pivotal to the pathogenicity and progression of the disease. In this review, we highlight the binary role of microglia activation in the pathophysiology of the disorder, both neuroprotective and neuromodulatory. We present how the expression of several cytokines implicated in dopaminergic neurons (DA) degeneration could be used as biomarkers for PD. Viral infections have been studied and correlated to the disease progression, usually operating as trigger factors for the inflammatory process. The gut-brain axis and the possible contribution of the peripheral bowel inflammation to neuronal death, mainly dopaminergic neurons, seems to be a main contributor of brain neuroinflammation. The role of the immune system has also been analyzed implicating a-synuclein in the activation of innate and adaptive immunity. We also discuss therapeutic approaches concerning PD and neuroinflammation, which have been studied in experimental and in vitro models and data stemming from epidemiological studies.
Collapse
Affiliation(s)
- Chrysoula Marogianni
- Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece; (C.M.); (M.S.); (E.D.)
| | - Maria Sokratous
- Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece; (C.M.); (M.S.); (E.D.)
| | - Efthimios Dardiotis
- Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece; (C.M.); (M.S.); (E.D.)
| | | | - Dimitrios Bogdanos
- Department of Internal Medicine, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece;
| | - Georgia Xiromerisiou
- Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece; (C.M.); (M.S.); (E.D.)
| |
Collapse
|
36
|
Marchetti B. Nrf2/Wnt resilience orchestrates rejuvenation of glia-neuron dialogue in Parkinson's disease. Redox Biol 2020; 36:101664. [PMID: 32863224 PMCID: PMC7395594 DOI: 10.1016/j.redox.2020.101664] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/15/2020] [Accepted: 07/27/2020] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress and inflammation have long been recognized to contribute to Parkinson's disease (PD), a common movement disorder characterized by the selective loss of midbrain dopaminergic neurons (mDAn) of the substantia nigra pars compacta (SNpc). The causes and mechanisms still remain elusive, but a complex interplay between several genes and a number of interconnected environmental factors, are chiefly involved in mDAn demise, as they intersect the key cellular functions affected in PD, such as the inflammatory response, mitochondrial, lysosomal, proteosomal and autophagic functions. Nuclear factor erythroid 2 -like 2 (NFE2L2/Nrf2), the master regulator of cellular defense against oxidative stress and inflammation, and Wingless (Wnt)/β-catenin signaling cascade, a vital pathway for mDAn neurogenesis and neuroprotection, emerge as critical intertwinned actors in mDAn physiopathology, as a decline of an Nrf2/Wnt/β-catenin prosurvival axis with age underlying PD mutations and a variety of noxious environmental exposures drive PD neurodegeneration. Unexpectedly, astrocytes, the so-called "star-shaped" cells, harbouring an arsenal of "beneficial" and "harmful" molecules represent the turning point in the physiopathological and therapeutical scenario of PD. Fascinatingly, "astrocyte's fil rouge" brings back to Nrf2/Wnt resilience, as boosting the Nrf2/Wnt resilience program rejuvenates astrocytes, in turn (i) mitigating nigrostriatal degeneration of aged mice, (ii) reactivating neural stem progenitor cell proliferation and neuron differentiation in the brain and (iii) promoting a beneficial immunomodulation via bidirectional communication with mDAns. Then, through resilience of Nrf2/Wnt/β-catenin anti-ageing, prosurvival and proregenerative molecular programs, it seems possible to boost the inherent endogenous self-repair mechanisms. Here, the cellular and molecular aspects as well as the therapeutical options for rejuvenating glia-neuron dialogue will be discussed together with major glial-derived mechanisms and therapies that will be fundamental to the identification of novel diagnostic tools and treatments for neurodegenerative diseases (NDs), to fight ageing and nigrostriatal DAergic degeneration and promote functional recovery.
Collapse
Affiliation(s)
- Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Pharmacology Section, Medical School, University of Catania, Via S. Sofia 65, 95125, Catania, Italy; Oasi Research Institute-IRCCS, Neuropharmacology Section, Via Conte Ruggero 73, 94018, Troina, EN, Italy.
| |
Collapse
|
37
|
Söderbom G. Status and future directions of clinical trials in Parkinson's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:153-188. [PMID: 32739003 DOI: 10.1016/bs.irn.2020.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Novel therapies are needed to treat Parkinson's disease (PD) in which the clinical unmet need is pressing. Currently, no clinically available therapeutic strategy can either retard or reverse PD or repair its pathological consequences. l-DOPA (levodopa) is still the gold standard therapy for motor symptoms yet symptomatic therapies for both motor and non-motor symptoms are improving. Many on-going, intervention trials cover a broad range of targets, including cell replacement and gene therapy approaches, quality of life improving technologies, and disease-modifying strategies (e.g., controlling aberrant α-synuclein accumulation and regulating cellular/neuronal bioenergetics). Notably, the repurposing of glucagon-like peptide-1 analogues with potential disease-modifying effects based on metabolic pathology associated with PD has been promising. Nevertheless, there is a clear need for improved therapeutic and diagnostic options, disease progression tracking and patient stratification capabilities to deliver personalized treatment and optimize trial design. This review discusses some of the risk factors and consequent pathology associated with PD and particularly the metabolic aspects of PD, novel therapies targeting these pathologies (e.g., mitochondrial and lysosomal dysfunction, oxidative stress, and inflammation/neuroinflammation), including the repurposing of metabolic therapies, and unmet needs as potential drivers for future clinical trials and research in PD.
Collapse
|
38
|
Wijeyekoon RS, Kronenberg-Versteeg D, Scott KM, Hayat S, Kuan WL, Evans JR, Breen DP, Cummins G, Jones JL, Clatworthy MR, Floto RA, Barker RA, Williams-Gray CH. Peripheral innate immune and bacterial signals relate to clinical heterogeneity in Parkinson's disease. Brain Behav Immun 2020; 87:473-488. [PMID: 32006615 PMCID: PMC7613010 DOI: 10.1016/j.bbi.2020.01.018] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/14/2020] [Accepted: 01/28/2020] [Indexed: 01/09/2023] Open
Abstract
The innate immune system is implicated in Parkinson's disease (PD), but peripheral in-vivo clinical evidence of the components and driving mechanisms involved and their relationship with clinical heterogeneity and progression to dementia remain poorly explored. We examined changes in peripheral innate immune-related markers in PD cases (n = 41) stratified according to risk of developing early dementia. 'Higher Risk'(HR) (n = 23) and 'Lower Risk' (LR) (n = 18) groups were defined according to neuropsychological predictors and MAPT H1/H2 genotype, and compared to age, gender and genotype-matched controls. Monocyte subsets and expression of key surface markers were measured using flow cytometry. Serum markers including alpha-synuclein, inflammasome-related caspase-1 and bacterial translocation-related endotoxin were measured using quantitative immuno-based assays. Specific markers were further investigated using monocyte assays and validated in plasma samples from a larger incident PD cohort (n = 95). We found that classical monocyte frequency was elevated in PD cases compared to controls, driven predominantly by the HR group, in whom Toll-Like Receptor (TLR)4+ monocytes and monocyte Triggering Receptor Expressed on Myeloid cells-2 (TREM2) expression were also increased. Monocyte Human Leukocyte Antigen (HLA)-DR expression correlated with clinical variables, with lower levels associated with worse cognitive/motor performance. Notably, monocyte changes were accompanied by elevated serum bacterial endotoxin, again predominantly in the HR group. Serum alpha-synuclein and inflammasome-related caspase-1 were decreased in PD cases compared to controls regardless of group, with decreased monocyte alpha-synuclein secretion in HR cases. Further, alpha-synuclein and caspase-1 correlated positively in serum and monocyte lysates, and in plasma from the larger cohort, though no associations were seen with baseline or 36-month longitudinal clinical data. Principal Components Analysis of all monocyte and significant serum markers indicated 3 major components. Component 1 (alpha-synuclein, caspase-1, TLR2+ monocytes) differentiated PD cases and controls in both groups, while Component 2 (endotoxin, monocyte TREM2, alpha-synuclein) did so predominantly in the HR group. Component 3 (classical monocytes, alpha-synuclein) also differentiated cases and controls overall in both groups. These findings demonstrate that systemic innate immune changes are present in PD and are greatest in those at higher risk of rapid progression to dementia. Markers associated with PD per-se (alpha-synuclein, caspase-1), differ from those related to cognitive progression and clinical heterogeneity (endotoxin, TREM2, TLR4, classical monocytes, HLA-DR), with mechanistic and therapeutic implications. Alpha-synuclein and caspase-1 are associated, suggesting inflammasome involvement common to all PD, while bacterial translocation associated changes may contribute towards progression to Parkinson's dementia. Additionally, HLA-DR-associated variations in antigen presentation/clearance may modulate existing clinical disease.
Collapse
Affiliation(s)
- Ruwani S. Wijeyekoon
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK,Corresponding Author;
| | | | - Kirsten M. Scott
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Shaista Hayat
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Wei-Li Kuan
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Jonathan R. Evans
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK,Nottingham University Hospital NHS Trust, Nottingham, UK
| | - David P. Breen
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor’s Building, 49, Little France Crescent, Edinburgh, EH16 4SB, UK,Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Chancellor’s Building, 49, Little France Crescent, Edinburgh, EH16 4SB, UK,Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, 9, Little France Road, Edinburgh BioQuarter, Edinburgh, EH16 4UX, UK
| | - Gemma Cummins
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| | - Joanne L. Jones
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - R. Andres Floto
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Roger A. Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK,Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Caroline H. Williams-Gray
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, E.D. Adrian Building, Forvie Site, Robinson Way, Cambridge CB2 0PY, UK
| |
Collapse
|
39
|
Rhein Suppresses Neuroinflammation via Multiple Signaling Pathways in LPS-Stimulated BV2 Microglia Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:7210627. [PMID: 32714414 PMCID: PMC7341424 DOI: 10.1155/2020/7210627] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/24/2020] [Accepted: 05/29/2020] [Indexed: 01/09/2023]
Abstract
As a bioactive absorbed compound of rhubarb, Rhein is applied for the treatment of brain injury. However, the underlying pharmacological mechanisms remain unclear. In this study, we aimed to explore antineuroinflammatory functions and underlying mechanisms of Rhein in vitro. BV2 microglia cells were chosen and irritated by LPS. The influence of Rhein on cell viability was determined using MTT assay. We finely gauged the proinflammatory cytokines of TNF-α and IL-1β through tests of immunofluorescence staining, ELISA, RT-qPCR, and western blot. Additionally, mediators including IL-6, IL-12, iNOS, and IL-10 were surveyed by ELISA. Furthermore, protein levels of the underlying signaling pathways (PI3K/Akt, p38, ERK1/2, and TLR4/NF-κB) were tested adopting western blot. We found that Rhein reduced the secretion of pivotal indicators including TNF-α and IL-1β, effectively restraining their mRNA and protein expression in LPS-activated BV2 microglial cells. Besides, Rhein treatment demoted the production of IL-6, IL-12, and iNOS and promoted the excretion of IL-10. Subsequent mechanistic experiments revealed that Rhein obviously downregulated the phosphorylation levels of PI3K, Akt, p38, and ERK1/2 and simultaneously upregulated the PTEN expression. In addition, Rhein antagonized the increase of TLR4, p-IκBα, and NF-κB. In summary, Rhein suppresses neuroinflammation via multiple signaling pathways (PI3K/Akt, p38, ERK1/2, and TLR4/NF-κB) in LPS-stimulated BV2 microglia cells. This study highlights a natural agent for prevention and treatment of neuroinflammation.
Collapse
|
40
|
Madureira M, Connor-Robson N, Wade-Martins R. "LRRK2: Autophagy and Lysosomal Activity". Front Neurosci 2020; 14:498. [PMID: 32523507 PMCID: PMC7262160 DOI: 10.3389/fnins.2020.00498] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/21/2020] [Indexed: 01/07/2023] Open
Abstract
It has been 15 years since the Leucine-rich repeat kinase 2 (LRRK2) gene was identified as the most common genetic cause for Parkinson's disease (PD). The two most common mutations are the LRRK2-G2019S, located in the kinase domain, and the LRRK2-R1441C, located in the ROC-COR domain. While the LRRK2-G2019S mutation is associated with increased kinase activity, the LRRK2-R1441C exhibits a decreased GTPase activity and altered kinase activity. Multiple lines of evidence have linked the LRRK2 protein with a role in the autophagy pathway and with lysosomal activity in neurons. Neurons rely heavily on autophagy to recycle proteins and process cellular waste due to their post-mitotic state. Additionally, lysosomal activity decreases with age which can potentiate the accumulation of α-synuclein, the pathological hallmark of PD, and subsequently lead to the build-up of Lewy bodies (LBs) observed in this disorder. This review provides an up to date summary of the LRRK2 field to understand its physiological role in the autophagy pathway in neurons and related cells. Careful assessment of how LRRK2 participates in the regulation of phagophore and autophagosome formation, autophagosome and lysosome fusion, lysosomal maturation, maintenance of lysosomal pH and calcium levels, and lysosomal protein degradation are addressed. The autophagy pathway is a complex cellular process and due to the variety of LRRK2 models studied in the field, associated phenotypes have been reported to be seemingly conflicting. This review provides an in-depth discussion of different models to assess the normal and disease-associated role of the LRRK2 protein on autophagic function. Given the importance of the autophagy pathway in Parkinson's pathogenesis it is particularly relevant to focus on the role of LRRK2 to discover novel therapeutic approaches that restore lysosomal protein degradation homeostasis.
Collapse
Affiliation(s)
- Marta Madureira
- Department of Physiology, Anatomy and Genetics, Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
- Graduate Program in Areas of Basic and Applied Biology (GABBA), Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Natalie Connor-Robson
- Department of Physiology, Anatomy and Genetics, Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
| | - Richard Wade-Martins
- Department of Physiology, Anatomy and Genetics, Oxford Parkinson’s Disease Centre, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
41
|
Wallings RL, Herrick MK, Tansey MG. LRRK2 at the Interface Between Peripheral and Central Immune Function in Parkinson's. Front Neurosci 2020; 14:443. [PMID: 32508566 PMCID: PMC7253584 DOI: 10.3389/fnins.2020.00443] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/09/2020] [Indexed: 12/20/2022] Open
Abstract
It is becoming increasingly accepted that there is an interplay between the peripheral immune response and neuroinflammation in the pathophysiology of Parkinson's disease (PD). Mutations in the leucine-rich-repeat kinase 2 (LRRK2) gene are associated with familial and sporadic cases of PD but are also found in immune-related disorders, such as inflammatory bowel disease (IBD) and leprosy. Furthermore, LRRK2 has been associated with bacterial infections such as Mycobacterium tuberculosis and Salmonella typhimurium. Recent evidence suggests a role of LRRK2 in the regulation of the immune system and modulation of inflammatory responses, at a systemic level, with LRRK2 functionally implicated in both the immune system of the central nervous system (CNS) and the periphery. It has therefore been suggested that peripheral immune signaling may play an important role in the regulation of neurodegeneration in LRRK2 as well as non-LRRK2-associated PD. This review will discuss the current evidence for this hypothesis and will provide compelling rationale for placing LRRK2 at the interface between peripheral immune responses and neuroinflammation.
Collapse
Affiliation(s)
- Rebecca L. Wallings
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, United States
| | - Mary K. Herrick
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, United States
- Laney Graduate School, Emory University, Atlanta, GA, United States
| | - Malú Gámez Tansey
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
42
|
Lee H, Flynn R, Sharma I, Haberman E, Carling PJ, Nicholls FJ, Stegmann M, Vowles J, Haenseler W, Wade-Martins R, James WS, Cowley SA. LRRK2 Is Recruited to Phagosomes and Co-recruits RAB8 and RAB10 in Human Pluripotent Stem Cell-Derived Macrophages. Stem Cell Reports 2020; 14:940-955. [PMID: 32359446 PMCID: PMC7221108 DOI: 10.1016/j.stemcr.2020.04.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 12/12/2022] Open
Abstract
The Parkinson's disease-associated gene, LRRK2, is also associated with immune disorders and infectious disease and is expressed in immune subsets. Here, we characterize a platform for interrogating the expression and function of endogenous LRRK2 in authentic human phagocytes using human induced pluripotent stem cell-derived macrophages and microglia. Endogenous LRRK2 is expressed and upregulated by interferon-γ in these cells, including a 187-kDa cleavage product. Using LRRK2 knockout and G2019S isogenic repair lines, we find that LRRK2 is not involved in initial phagocytic uptake of bioparticles but is recruited to LAMP1+/RAB9+ "maturing" phagosomes, and LRRK2 kinase inhibition enhances its residency at the phagosome. Importantly, LRRK2 is required for RAB8a and RAB10 recruitment to phagosomes, implying that LRRK2 operates at the intersection between phagosome maturation and recycling pathways in these professional phagocytes.
Collapse
Affiliation(s)
- Heyne Lee
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Rowan Flynn
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Ishta Sharma
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Emma Haberman
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Phillippa J Carling
- Oxford Parkinson's Disease Centre, University of Oxford, South Parks Road, Oxford OX1 3QX, UK; Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Francesca J Nicholls
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
| | - Monika Stegmann
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Jane Vowles
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK; Oxford Parkinson's Disease Centre, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Walther Haenseler
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, University of Oxford, South Parks Road, Oxford OX1 3QX, UK; Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - William S James
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Sally A Cowley
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK; Oxford Parkinson's Disease Centre, University of Oxford, South Parks Road, Oxford OX1 3QX, UK.
| |
Collapse
|
43
|
Hu X, Mao C, Fan L, Luo H, Hu Z, Zhang S, Yang Z, Zheng H, Sun H, Fan Y, Yang J, Shi C, Xu Y. Modeling Parkinson's Disease Using Induced Pluripotent Stem Cells. Stem Cells Int 2020; 2020:1061470. [PMID: 32256606 PMCID: PMC7091557 DOI: 10.1155/2020/1061470] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 02/08/2020] [Accepted: 02/15/2020] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. The molecular mechanisms of PD at the cellular level involve oxidative stress, mitochondrial dysfunction, autophagy, axonal transport, and neuroinflammation. Induced pluripotent stem cells (iPSCs) with patient-specific genetic background are capable of directed differentiation into dopaminergic neurons. Cell models based on iPSCs are powerful tools for studying the molecular mechanisms of PD. The iPSCs used for PD studies were mainly from patients carrying mutations in synuclein alpha (SNCA), leucine-rich repeat kinase 2 (LRRK2), PTEN-induced putative kinase 1 (PINK1), parkin RBR E3 ubiquitin protein ligase (PARK2), cytoplasmic protein sorting 35 (VPS35), and variants in glucosidase beta acid (GBA). In this review, we summarized the advances in molecular mechanisms of Parkinson's disease using iPSC models.
Collapse
Affiliation(s)
- Xinchao Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000 Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, China
| | - Chengyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000 Henan, China
| | - Liyuan Fan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000 Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, China
| | - Haiyang Luo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000 Henan, China
| | - Zhengwei Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000 Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, China
| | - Shuo Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000 Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, China
| | - Zhihua Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000 Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, China
| | - Huimin Zheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000 Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, China
| | - Huifang Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000 Henan, China
| | - Yu Fan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000 Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000 Henan, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000 Henan, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000 Henan, China
| |
Collapse
|
44
|
LRRK2 regulation of immune-pathways and inflammatory disease. Biochem Soc Trans 2020; 47:1581-1595. [PMID: 31769472 PMCID: PMC6925522 DOI: 10.1042/bst20180463] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/13/2022]
Abstract
Mutations in the leucine-rich-repeat kinase 2 (LRRK2) gene are associated with familial and sporadic cases of Parkinson's disease but are also found in immune-related disorders such as inflammatory bowel disease, tuberculosis and leprosy. LRRK2 is highly expressed in immune cells and has been functionally linked to pathways pertinent to immune cell function, such as cytokine release, autophagy and phagocytosis. Here, we examine the current understanding of the role of LRRK2 kinase activity in pathway regulation in immune cells, drawing upon data from multiple diseases associated with LRRK2 to highlight the pleiotropic effects of LRRK2 in different cell types. We discuss the role of the bona fide LRRK2 substrate, Rab GTPases, in LRRK2 pathway regulation as well as downstream events in the autophagy and inflammatory pathways.
Collapse
|
45
|
Cogo S, Manzoni C, Lewis PA, Greggio E. Leucine-rich repeat kinase 2 and lysosomal dyshomeostasis in Parkinson disease. J Neurochem 2020; 152:273-283. [PMID: 31693760 DOI: 10.1111/jnc.14908] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/26/2019] [Accepted: 10/31/2019] [Indexed: 12/24/2022]
Abstract
Over the last two decades, a number of studies have underlined the importance of lysosomal-based degradative pathways in maintaining the homeostasis of post-mitotic cells, and revealed the remarkable contribution of a functional autophagic machinery in the promotion of longevity. In contrast, defects in the clearance of organelles and aberrant protein aggregates have been linked to accelerated neuronal loss and neurological dysfunction. Several neurodegenerative disorders, among which Alzheimer disease (AD), Frontotemporal dementia, and Amyotrophic Lateral Sclerosis to name a few, are associated with alterations of the autophagy and endo-lysosomal pathways. In Parkinson disease (PD), the most prevalent genetic determinant, Leucine-rich repeat kinase 2 (LRRK2), is believed to be involved in the regulation of intracellular vesicle traffic, autophagy and lysosomal function. Here, we review the current understanding of the mechanisms by which LRRK2 regulates lysosomal-based degradative pathways in neuronal and non-neuronal cells and discuss the impact of pathogenic PD mutations in contributing to lysosomal dyshomeostasis.
Collapse
Affiliation(s)
- Susanna Cogo
- Department of Biology, University of Padova, Padova, Italy
| | - Claudia Manzoni
- School of Pharmacy, University of Reading, Reading, UK
- Department of Neurodegenerative Diseases, University College London, London, UK
| | - Patrick A Lewis
- School of Pharmacy, University of Reading, Reading, UK
- Department of Neurodegenerative Diseases, University College London, London, UK
| | - Elisa Greggio
- Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
46
|
Santaella A, Kuiperij HB, van Rumund A, Esselink RAJ, van Gool AJ, Bloem BR, Verbeek MM. Inflammation biomarker discovery in Parkinson's disease and atypical parkinsonisms. BMC Neurol 2020; 20:26. [PMID: 31952511 PMCID: PMC6967088 DOI: 10.1186/s12883-020-1608-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 01/09/2020] [Indexed: 01/09/2023] Open
Abstract
Background Parkinson’s disease (PD) and atypical parkinsonisms (APD) have overlapping symptoms challenging an early diagnosis. Diagnostic accuracy is important because PD and APD have different prognosis and response to treatment. We aimed to identify diagnostic inflammatory biomarkers of PD and APD in cerebrospinal fluid (CSF) using the multiplex proximity extension assay (PEA) technology and to study possible correlations of biomarkers with disease progression. Methods CSF from a longitudinal cohort study consisting of PD and APD patients (PD, n = 44; multiple system atrophy (MSA), n = 14; vascular parkinsonism (VaP), n = 9; and PD with VaP, n = 7) and controls (n = 25) were analyzed. Results Concentrations of CCL28 were elevated in PD compared to controls (p = 0.0001). Five other biomarkers differentiated both MSA and PD from controls (p < 0.05) and 10 biomarkers differentiated MSA from controls, of which two proteins, i.e. beta nerve growth factor (β-NGF) and Delta and Notch like epidermal growth factor-related receptor (DNER), were also present at lower levels in MSA compared to PD (both p = 0.032). Two biomarkers (MCP-1 and MMP-10) positively correlated with PD progression (rho > 0.650; p < 0.01). Conclusions PEA technique identified potential new CSF biomarkers to help to predict the prognosis of PD. Also, we identified new candidate biomarkers to distinguish MSA from PD.
Collapse
Affiliation(s)
- Anna Santaella
- Departments of Neurology, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Laboratory Medicine, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Parkinson Center Nijmegen, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - H Bea Kuiperij
- Departments of Neurology, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Laboratory Medicine, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Anouke van Rumund
- Departments of Neurology, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Rianne A J Esselink
- Departments of Neurology, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Parkinson Center Nijmegen, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Alain J van Gool
- Laboratory Medicine, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Bastiaan R Bloem
- Departments of Neurology, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.,Parkinson Center Nijmegen, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Marcel M Verbeek
- Departments of Neurology, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands. .,Laboratory Medicine, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands. .,Parkinson Center Nijmegen, Radboud University Medical Center, and Donders Institute for Brain, Cognition and Behavior, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
47
|
Diana A, Collu M, Casu MA, Mocci I, Aguilar-Santelises M, Setzu MD. Improvements of Motor Performances in the Drosophila LRRK2 Loss-of-Function Model of Parkinson’s Disease: Effects of Dialyzed Leucocyte Extracts from Human Serum. Brain Sci 2020; 10:brainsci10010045. [PMID: 31947539 PMCID: PMC7017078 DOI: 10.3390/brainsci10010045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/05/2020] [Accepted: 01/09/2020] [Indexed: 11/16/2022] Open
Abstract
Within neurodegenerative syndromes, Parkinson’s disease (PD) is typically associated with its locomotor defects, sleep disturbances and related dopaminergic (DA) neuron loss. The fruit fly, Drosophila melanogaster (D. melanogaster), with leucine-rich repeat kinase 2 mutants (LRRK2) loss-of-function in the WD40 domain, provides mechanistic insights into corresponding human behaviour, possibly disclosing some physiopathologic features of PD in both genetic and sporadic forms. Moreover, several data support the boosting impact of innate and adaptive immunity pathways for driving the progression of PD. In this context, human dialyzable leukocyte extracts (DLE) have been extensively used to transfer antigen-specific information that influences the activity of various immune components, including inflammatory cytokines. Hence, the main goal of our study was to ascertain the therapeutic potential of DLE from male and female donors on D. melanogaster LRRK2 loss-of-function, as compared to D. melanogaster wild-type (WT), in terms of rescuing physiological parameters, such as motor and climbing activities, which are severely compromised in the mutant flies. Finally, in search of the anatomical structures responsible for restored functions in parkinsonian-like mutant flies, we found a topographical correlation between improvement of locomotor performances and an increased number of dopaminergic neurons in selective areas of LRRK2 mutant brains.
Collapse
Affiliation(s)
- Andrea Diana
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy;
- Correspondence: (A.D.); (M.D.S.); Tel.: +39-070-6754054/56 (A.D.); +39-070-6754183 (M.D.S.)
| | - Maria Collu
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy;
| | - Maria Antonietta Casu
- CNR Institute of Translational Pharmacology, 09010 Pula/Cagliari, Italy; (M.A.C.); (I.M.)
| | - Ignazia Mocci
- CNR Institute of Translational Pharmacology, 09010 Pula/Cagliari, Italy; (M.A.C.); (I.M.)
| | | | - Maria Dolores Setzu
- Department of Biomedical Sciences, University of Cagliari, 09042 Monserrato, Italy;
- Correspondence: (A.D.); (M.D.S.); Tel.: +39-070-6754054/56 (A.D.); +39-070-6754183 (M.D.S.)
| |
Collapse
|
48
|
Zaldivar-Diez J, Li L, Garcia AM, Zhao WN, Medina-Menendez C, Haggarty SJ, Gil C, Morales AV, Martinez A. Benzothiazole-Based LRRK2 Inhibitors as Wnt Enhancers and Promoters of Oligodendrocytic Fate. J Med Chem 2019; 63:2638-2655. [DOI: 10.1021/acs.jmedchem.9b01752] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Josefa Zaldivar-Diez
- Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Lingling Li
- Instituto Cajal, CSIC, Av. Doctor Arce, 37, 28002 Madrid, Spain
| | - Ana M. Garcia
- Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Wen-Ning Zhao
- Chemical Neurobiology Lab, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street, Boston, Massachusetts 02114, United States
| | | | - Stephen. J. Haggarty
- Chemical Neurobiology Lab, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Massachusetts General Hospital, Harvard Medical School, 185 Cambridge Street, Boston, Massachusetts 02114, United States
| | - Carmen Gil
- Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, 28031 Madrid, Spain
| | - Aixa V. Morales
- Instituto Cajal, CSIC, Av. Doctor Arce, 37, 28002 Madrid, Spain
| | - Ana Martinez
- Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, 28031 Madrid, Spain
| |
Collapse
|
49
|
Nenasheva VV, Tarantul VZ. Many Faces of TRIM Proteins on the Road from Pluripotency to Neurogenesis. Stem Cells Dev 2019; 29:1-14. [PMID: 31686585 DOI: 10.1089/scd.2019.0152] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tripartite motif (TRIM) proteins participate in numerous biological processes. They are the key players in immune system and are involved in the oncogenesis. Moreover, TRIMs are the highly conserved regulators of developmental pathways in both vertebrates and invertebrates. In particular, numerous data point to the participation of TRIMs in the determination of stem cell fate, as well as in the neurogenesis. TRIMs apply various mechanisms to perform their functions. Their common feature is the ability to ubiquitinate proteins mediated by the Really Interesting New Gene (RING) domain. Different C-terminal domains of TRIMs are involved in DNA and RNA binding, protein/protein interactions, and chromatin-mediated transcriptional regulation. Mutations and alterations of TRIM expression cause significant disturbances in the stem cells' self-renewal and neurogenesis, which result in the various pathologies of the nervous system (neurodegeneration, neuroinflammation, and malignant transformation). This review discusses the diverse molecular mechanisms of participation of TRIMs in stem cell maintenance and self-renewal as well as in neural differentiation processes and neuropathology.
Collapse
Affiliation(s)
- Valentina V Nenasheva
- Department of Viral and Cellular Molecular Genetics, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Vyacheslav Z Tarantul
- Department of Viral and Cellular Molecular Genetics, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
50
|
Subbannayya Y, Pinto SM, Bösl K, Prasad TSK, Kandasamy RK. Dynamics of Dual Specificity Phosphatases and Their Interplay with Protein Kinases in Immune Signaling. Int J Mol Sci 2019; 20:ijms20092086. [PMID: 31035605 PMCID: PMC6539644 DOI: 10.3390/ijms20092086] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/23/2019] [Accepted: 04/25/2019] [Indexed: 12/12/2022] Open
Abstract
Dual specificity phosphatases (DUSPs) have a well-known role as regulators of the immune response through the modulation of mitogen-activated protein kinases (MAPKs). Yet the precise interplay between the various members of the DUSP family with protein kinases is not well understood. Recent multi-omics studies characterizing the transcriptomes and proteomes of immune cells have provided snapshots of molecular mechanisms underlying innate immune response in unprecedented detail. In this study, we focus on deciphering the interplay between members of the DUSP family with protein kinases in immune cells using publicly available omics datasets. Our analysis resulted in the identification of potential DUSP-mediated hub proteins including MAPK7, MAPK8, AURKA, and IGF1R. Furthermore, we analyzed the association of DUSP expression with TLR4 signaling and identified VEGF, FGFR, and SCF-KIT pathway modules to be regulated by the activation of TLR4 signaling. Finally, we identified several important kinases including LRRK2, MAPK8, and cyclin-dependent kinases as potential DUSP-mediated hubs in TLR4 signaling. The findings from this study have the potential to aid in the understanding of DUSP signaling in the context of innate immunity. Further, this will promote the development of therapeutic modalities for disorders with aberrant DUSP signaling.
Collapse
Affiliation(s)
- Yashwanth Subbannayya
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, N-7491 Trondheim, Norway.
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore 575018, India.
| | - Sneha M Pinto
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, N-7491 Trondheim, Norway.
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore 575018, India.
| | - Korbinian Bösl
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, N-7491 Trondheim, Norway.
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore 575018, India.
| | - Richard K Kandasamy
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, N-7491 Trondheim, Norway.
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, N-0349 Oslo, Norway.
| |
Collapse
|