1
|
Maekawa T, Motokawa R, Kawashima R, Tamaki S, Hara Y, Kawakami F, Ichikawa T. Biphenotypic Cells and α-Synuclein Accumulation in Enteric Neurons of Leucine-Rich Repeat Kinase 2 Knockout Mice. Dig Dis Sci 2024; 69:2828-2840. [PMID: 38849592 DOI: 10.1007/s10620-024-08494-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 05/09/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Leucine-rich repeat kinase 2 is a molecule that is responsible for familial Parkinson's disease. Our previous findings revealed that leucine-rich repeat kinase 2 is expressed in the enteric nervous system. However, which cells in the enteric nervous system express leucine-rich repeat kinase 2 and whether leucine-rich repeat kinase 2 is associated with the structure of the enteric nervous system remain unclear. The enteric nervous system is remarkable because some patients with Parkinson's disease experience gastrointestinal symptoms before developing motor symptoms. AIMS We established a leucine-rich repeat kinase 2 reporter mouse model and performed immunostaining in leucine-rich repeat kinase 2 knockout mice. METHODS Longitudinal muscle containing the myenteric plexus prepared from leucine-rich repeat kinase 2 reporter mice was analyzed by immunostaining using anti-green fluorescent protein (GFP) antibody. Immunostaining using several combinations of antibodies characterizing enteric neurons and glial cells was performed on intestinal preparations from leucine-rich repeat kinase 2 knockout mice. RESULTS GFP expression in the reporter mice was predominantly in enteric glial cells rather than in enteric neurons. Immunostaining revealed that differences in the structure and proportion of major immunophenotypic cells were not apparent in the knockout mice. Interestingly, the number of biphenotypic cells expressing the neuronal and glial cell markers increased in the leucine-rich repeat kinase 2 knockout mice. Moreover, there was accumulation of α-synuclein in the knockout mice. CONCLUSIONS Our present findings suggest that leucine-rich repeat kinase 2 is a newly recognized molecule that potentially regulates the integrity of enteric nervous system and enteric α-synuclein accumulation.
Collapse
Affiliation(s)
- Tatsunori Maekawa
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, 1-15-1 Kitasato, Minami-Ku, Sagamihara, Kanagawa, 252-0373, Japan.
- Department of Biochemistry, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa, Japan.
- Research Facility of Regenerative Medicine and Cell Design, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa, Japan.
| | - Ryuichi Motokawa
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, 1-15-1 Kitasato, Minami-Ku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Rei Kawashima
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, 1-15-1 Kitasato, Minami-Ku, Sagamihara, Kanagawa, 252-0373, Japan
- Department of Biochemistry, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
- Research Facility of Regenerative Medicine and Cell Design, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Shun Tamaki
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, 1-15-1 Kitasato, Minami-Ku, Sagamihara, Kanagawa, 252-0373, Japan
- Department of Biochemistry, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
- Research Facility of Regenerative Medicine and Cell Design, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Yusuke Hara
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, 1-15-1 Kitasato, Minami-Ku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Fumitaka Kawakami
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, 1-15-1 Kitasato, Minami-Ku, Sagamihara, Kanagawa, 252-0373, Japan
- Department of Health Administration, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
- Research Facility of Regenerative Medicine and Cell Design, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
| | - Takafumi Ichikawa
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, 1-15-1 Kitasato, Minami-Ku, Sagamihara, Kanagawa, 252-0373, Japan
- Department of Biochemistry, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
- Research Facility of Regenerative Medicine and Cell Design, School of Allied Health Sciences, Kitasato University, Sagamihara, Kanagawa, Japan
| |
Collapse
|
2
|
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
|
3
|
de Guilhem de Lataillade A, Pellegrini C, Neunlist M, Rolli-Derkinderen M, Derkinderen P. Are LRRK2 mysteries lurking in the gut? Am J Physiol Gastrointest Liver Physiol 2023; 325:G429-G435. [PMID: 37643021 DOI: 10.1152/ajpgi.00162.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/23/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023]
Abstract
Gut-brain axis and inflammation are two hot topics in Parkinson's disease (PD). In this setting, the leucine-rich repeat kinase 2 (LRRK2) gene, which encodes the eponym protein, has attracted much attention. LRRK2 is not only the gene most commonly associated with Parkinson's disease but also a susceptibility gene for Crohn's disease (CD), thereby suggesting that it may sit at the crossroads of gastrointestinal inflammation, Parkinson's, and Crohn's disease. In contrast to the accumulated data on LRRK2 in the central nervous system (CNS), research on LRRK2 in the digestive tract is still in its infancy, and the scope of the present review article is therefore to review existing studies on LRRK2 in the gastrointestinal tract in both physiological and pathological conditions. In light of current data on LRRK2 in the gastrointestinal tract, we discuss if LRRK2 could be or not regarded as a molecular link between gut inflammation, Parkinson's disease, and Crohn's disease, and we suggest directions for future research.
Collapse
Affiliation(s)
- Adrien de Guilhem de Lataillade
- The Enteric Nervous System In Gut And Brain Disorders, Nantes Université, Centre Hospitalier Universitaire de Nantes, INSERM, Nantes, France
| | - Carolina Pellegrini
- Unit of Histology and Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Michel Neunlist
- The Enteric Nervous System In Gut And Brain Disorders, Nantes Université, Centre Hospitalier Universitaire de Nantes, INSERM, Nantes, France
| | - Malvyne Rolli-Derkinderen
- The Enteric Nervous System In Gut And Brain Disorders, Nantes Université, Centre Hospitalier Universitaire de Nantes, INSERM, Nantes, France
| | - Pascal Derkinderen
- The Enteric Nervous System In Gut And Brain Disorders, Nantes Université, Centre Hospitalier Universitaire de Nantes, INSERM, Nantes, France
| |
Collapse
|
4
|
de Guilhem de Lataillade A, Caillaud M, Oullier T, Naveilhan P, Pellegrini C, Tolosa E, Neunlist M, Rolli-Derkinderen M, Gelpi E, Derkinderen P. LRRK2 expression in normal and pathologic human gut and in rodent enteric neural cell lines. J Neurochem 2023; 164:193-209. [PMID: 36219522 DOI: 10.1111/jnc.15704] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 09/01/2022] [Accepted: 09/15/2022] [Indexed: 02/04/2023]
Abstract
Leucine-rich repeat kinase 2 (LRRK2) gene, which is the gene most commonly associated with Parkinson's disease (PD), is also a susceptibility gene for Crohn's disease, thereby suggesting that LRRK2 may sit at the crossroads of gastrointestinal inflammation, Parkinson's, and Crohn's disease. LRRK2 protein has been studied intensely in both CNS neurons and in immune cells, but there are only few studies on LRRK2 in the enteric nervous system (ENS). LRRK2 is present in ENS ganglia and the existing studies on LRRK2 expression in colonic biopsies from PD subjects have yielded conflicting results. Herein, we propose to extend these findings by studying in more details LRRK2 expression in the ENS. LRRK2 expression was evaluated in full thickness segments of colon of 16 Lewy body, 12 non-Lewy body disorders cases, and 3 non-neurodegenerative controls and in various enteric neural cell lines. We showed that, in addition to enteric neurons, LRRK2 is constitutively expressed in enteric glial cells in both fetal and adult tissues. LRRK2 immunofluorescence intensity in the myenteric ganglia was not different between Lewy body and non-Lewy body disorders. Additionally, we identified the cAMP pathway as a key signaling pathway involved in the regulation of LRRK2 expression and phosphorylation in the enteric glial cells. Our study is the first detailed characterization of LRRK2 in the ENS and the first to show that enteric glial cells express LRRK2. Our findings provide a basis to unravel the functions of LRRK2 in the ENS and to further investigate the pathological changes in enteric synucleinopathies.
Collapse
Affiliation(s)
| | - Martial Caillaud
- Nantes Université, CHU Nantes, INSERM, The enteric nervous system in gut and brain disorders, Nantes, France
| | - Thibauld Oullier
- Nantes Université, CHU Nantes, INSERM, The enteric nervous system in gut and brain disorders, Nantes, France
| | - Philippe Naveilhan
- Nantes Université, CHU Nantes, INSERM, The enteric nervous system in gut and brain disorders, Nantes, France
| | - Carolina Pellegrini
- Unit of Histology and Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Eduardo Tolosa
- Parkinson disease and Movement Disorders Unit, Neurology Service, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED: CB06/05/0018-ISCIII), Barcelona, Spain
| | - Michel Neunlist
- Nantes Université, CHU Nantes, INSERM, The enteric nervous system in gut and brain disorders, Nantes, France
| | - Malvyne Rolli-Derkinderen
- Nantes Université, CHU Nantes, INSERM, The enteric nervous system in gut and brain disorders, Nantes, France
| | - Ellen Gelpi
- Neurological Tissue Bank of the Biobank-Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain.,Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Pascal Derkinderen
- Nantes Université, CHU Nantes, INSERM, The enteric nervous system in gut and brain disorders, Nantes, France
| |
Collapse
|
5
|
Peripheral Blood DNA Methylation Profiles Do Not Predict Endoscopic Post-Operative Recurrence in Crohn's Disease Patients. Int J Mol Sci 2022; 23:ijms231810467. [PMID: 36142381 PMCID: PMC9503775 DOI: 10.3390/ijms231810467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/30/2022] [Accepted: 09/04/2022] [Indexed: 11/30/2022] Open
Abstract
Prediction of endoscopic post-operative recurrence (POR) in Crohn’s disease (CD) patients following ileocolonic resection (ICR) using clinical risk factors alone has thus far been inadequate. While peripheral blood leukocyte (PBL) DNA methylation has shown promise as a tool for predicting recurrence in cancer, no data in CD patients exists. Therefore, this study explored the association and predictive value of PBL DNA methylation in CD patients following ICR. From a cohort of 117 CD patients undergoing ICR, epigenome-wide PBL methylation profiles from 25 carefully selected patients presenting either clear endoscopic remission (n = 12) or severe recurrence (n = 13) were assessed using the Illumina MethylationEPIC (850K) array. No statistically significant differentially methylated positions (DMPs) or regions (DMRs) associated with endoscopic POR were identified (FDR p ≤ 0.05), further evidenced by the low accuracy (0.625) following elastic net classification analysis. Nonetheless, interrogating the most significant differences in methylation suggested POR-associated hypermethylation in the MBNL1, RAB29 and LEPR genes, respectively, which are involved in intestinal fibrosis, inflammation and wound healing. Notably, we observed a higher estimated proportion of monocytes in endoscopic POR compared to remission. Altogether, we observed limited differences in the genome-wide DNA methylome among CD patients with and without endoscopic POR. We therefore conclude that PBL DNA methylation is not a feasible predictive tool in post-operative CD.
Collapse
|
6
|
Lee JYS, Ng JH, Saffari SE, Tan EK. Parkinson's disease and cancer: a systematic review and meta-analysis on the influence of lifestyle habits, genetic variants, and gender. Aging (Albany NY) 2022; 14:2148-2173. [PMID: 35247252 PMCID: PMC8954974 DOI: 10.18632/aging.203932] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/15/2022] [Indexed: 12/09/2022]
Abstract
PURPOSE The relationship between Parkinson's disease (PD) and cancer has been debated. Gender and genetic influences on cancer development in PD is unclear. METHODS Using QUOROM guidelines, we conducted a systematic review and meta-analysis on potential clinical and genetic factors influencing the PD and subsequent cancer relationship. English articles published in PubMed, Web of Science, and SCOPUS from 2010 to 30 August 2020 were considered for suitability. RESULTS Of 46 studies identified, fourteen satisfied the inclusion criteria and were further analysed. Unadjusted risk ratios (RR) and 95% confidence intervals were computed to determine the PD and cancer relationship. PD patients have decreased subsequent cancer risks (RR = 0.87, 95% CI = 0.81-0.93), reduced risks of colon, rectal, and colorectal cancer (RR = 0.77, 95% CI = 0.63-0.94), lung cancer (RR = 0.62, 95% CI = 0.48-0.80), and increased brain cancer (R = 1.48, 95% CI = 1.02-2.13) and melanoma risk (R = 1.76, 95% CI = 1.23-2.50). Compared to idiopathic PD, LRRK2-G2019S carriers had increased general cancer risks (RR = 1.26, 95% CI = 1.09-1.46), particularly brain (RR = 2.41, 95% CI = 1.06-5.50), breast (RR = 2.57, 95% CI = 1.19-5.58), colon (RR = 1.83, 95% CI = 1.13-2.99), and haematological cancers (RR = 2.05, 95% CI = 1.07-3.92). Female PD patients have decreased general cancer risks compared to male PD patients in this analysis (RR = 0.83, 95% CI = 0.69-0.98). CONCLUSION PD patients have reduced risks of colon, rectal, colorectal cancer and lung cancers and increased risks of brain cancer and melanoma. LRRK2-G2019S carriers have increased cancer risks, particularly brain, breast, colon and blood cancers. Female gender was associated with reduced risks. The role of ethnicity, comorbidities, and lifestyle habits on PD patients' subsequent cancer risk should be further investigated.
Collapse
Affiliation(s)
- Joon Yan Selene Lee
- Department of Neuroscience and Behavioural Disorders Programme, Duke-NUS Medical School, Singapore
| | - Jing Han Ng
- Department of Neurology, National Neuroscience Institute, Singapore
| | - Seyed Ehsan Saffari
- Department of Neuroscience and Behavioural Disorders Programme, Duke-NUS Medical School, Singapore.,Department of Neurology, National Neuroscience Institute, Singapore
| | - Eng-King Tan
- Department of Neuroscience and Behavioural Disorders Programme, Duke-NUS Medical School, Singapore.,Department of Neurology, National Neuroscience Institute, Singapore
| |
Collapse
|
7
|
Hemmings SMJ, Swart P, Womersely JS, Ovenden ES, van den Heuvel LL, McGregor NW, Meier S, Bardien S, Abrahams S, Tromp G, Emsley R, Carr J, Seedat S. RNA-seq analysis of gene expression profiles in posttraumatic stress disorder, Parkinson's disease and schizophrenia identifies roles for common and distinct biological pathways. DISCOVER MENTAL HEALTH 2022; 2:6. [PMID: 37861850 PMCID: PMC10501040 DOI: 10.1007/s44192-022-00009-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/14/2022] [Indexed: 10/21/2023]
Abstract
Evidence suggests that shared pathophysiological mechanisms in neuropsychiatric disorders (NPDs) may contribute to risk and resilience. We used single-gene and network-level transcriptomic approaches to investigate shared and disorder-specific processes underlying posttraumatic stress disorder (PTSD), Parkinson's disease (PD) and schizophrenia in a South African sample. RNA-seq was performed on blood obtained from cases and controls from each cohort. Gene expression and weighted gene correlation network analyses (WGCNA) were performed using DESeq2 and CEMiTool, respectively. Significant differences in gene expression were limited to the PTSD cohort. However, WGCNA implicated, amongst others, ribosomal expression, inflammation and ubiquitination as key players in the NPDs under investigation. Differential expression in ribosomal-related pathways was observed in the PTSD and PD cohorts, and focal adhesion and extracellular matrix pathways were implicated in PD and schizophrenia. We propose that, despite different phenotypic presentations, core transdiagnostic mechanisms may play important roles in the molecular aetiology of NPDs.
Collapse
Affiliation(s)
- Sian M J Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa.
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa.
| | - Patricia Swart
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
| | - Jacqueline S Womersely
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
| | - Ellen S Ovenden
- Systems Genetics Working Group, Department of Genetics, Stellenbosch University, Stellenbosch, South Africa
| | - Leigh L van den Heuvel
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
| | - Nathaniel W McGregor
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
- Systems Genetics Working Group, Department of Genetics, Stellenbosch University, Stellenbosch, South Africa
| | - Stuart Meier
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Tuberculosis Bioinformatics Initiative, Stellenbosch University, Cape Town, South Africa
| | - Soraya Bardien
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
| | - Shameemah Abrahams
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
| | - Gerard Tromp
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Tuberculosis Bioinformatics Initiative, Stellenbosch University, Cape Town, South Africa
- Centre for Bioinformatics and Computational Biology, Stellenbosch University, Stellenbosch, South Africa
| | - Robin Emsley
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
| | - Jonathan Carr
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Soraya Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
8
|
Derkinderen P, de Guilhem de Lataillade A, Neunlist M, Rolli‐Derkinderen M. Mild Chronic Colitis Triggers Parkinsonism in
LRRK2
Mutant Mice through Activating
TNF
‐α Pathway. Mov Disord 2022; 37:664-665. [DOI: 10.1002/mds.28948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/03/2022] [Indexed: 11/12/2022] Open
Affiliation(s)
- Pascal Derkinderen
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD Nantes France
- Department of Neurology CHU Nantes Nantes France
| | - Adrien de Guilhem de Lataillade
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD Nantes France
- Department of Neurology CHU Nantes Nantes France
| | - Michel Neunlist
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD Nantes France
| | - Malvyne Rolli‐Derkinderen
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD Nantes France
| |
Collapse
|
9
|
Lin CH, Lin HY, Ho EP, Ke YC, Cheng MF, Shiue CY, Wu CH, Liao PH, Hsu AYH, Chu LA, Liu YD, Lin YH, Tai YC, Shun CT, Chiu HM, Wu MS. Reply to: "Letter to the Editor by Derkinderen and Colleagues". Mov Disord 2022; 37:665-666. [PMID: 35092086 PMCID: PMC9306538 DOI: 10.1002/mds.28947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 01/16/2022] [Indexed: 11/28/2022] Open
Affiliation(s)
- Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Han-Yi Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - En-Pong Ho
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Ci Ke
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Mei-Fang Cheng
- Department of Nuclear Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chyng-Yann Shiue
- Department of Nuclear Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chi-Han Wu
- Department of Nuclear Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | - Li-An Chu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.,Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Ya-Ding Liu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.,Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Ya-Hui Lin
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.,Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Cheng Tai
- Department of Neurology, E-Da Hospital, Kaohsiung, Taiwan
| | - Chia-Tung Shun
- Department of Pathology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Han-Mo Chiu
- Department of Integrated Diagnostics & Therapeutics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Shiang Wu
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
10
|
de Guilhem de Lataillade A, Verchere J, Oullier T, Prigent A, Durand T, Pellegrini C, Neunlist M, Baron T, Rolli-Derkinderen M, Derkinderen P. LRRK2 is reduced in Parkinson's disease gut. Acta Neuropathol 2021; 142:601-603. [PMID: 34091743 DOI: 10.1007/s00401-021-02334-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 11/26/2022]
Affiliation(s)
- Adrien de Guilhem de Lataillade
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
- Department of Neurology, CHU Nantes, Inserm U1235 Nantes, 1 rue Gaston Veil, 44035, Nantes, France
| | - Jérémy Verchere
- French Agency for Food, Environmental, and Occupational Health and Safety (ANSES), University of Lyon, Lyon, France
| | - Thibauld Oullier
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Alice Prigent
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Tony Durand
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | | | - Michel Neunlist
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Thierry Baron
- French Agency for Food, Environmental, and Occupational Health and Safety (ANSES), University of Lyon, Lyon, France
| | - Malvyne Rolli-Derkinderen
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Pascal Derkinderen
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France.
- Department of Neurology, CHU Nantes, Inserm U1235 Nantes, 1 rue Gaston Veil, 44035, Nantes, France.
| |
Collapse
|
11
|
Lorente-Picón M, Laguna A. New Avenues for Parkinson's Disease Therapeutics: Disease-Modifying Strategies Based on the Gut Microbiota. Biomolecules 2021; 11:433. [PMID: 33804226 PMCID: PMC7998286 DOI: 10.3390/biom11030433] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is a multifactorial neurodegenerative disorder that currently affects 1% of the population over the age of 60 years, and for which no disease-modifying treatments exist. Neurodegeneration and neuropathology in different brain areas are manifested as both motor and non-motor symptoms in patients. Recent interest in the gut-brain axis has led to increasing research into the gut microbiota changes in PD patients and their impact on disease pathophysiology. As evidence is piling up on the effects of gut microbiota in disease development and progression, another front of action has opened up in relation to the potential usage of microbiota-based therapeutic strategies in treating gastrointestinal alterations and possibly also motor symptoms in PD. This review provides status on the different strategies that are in the front line (i.e., antibiotics; probiotics; prebiotics; synbiotics; dietary interventions; fecal microbiota transplantation, live biotherapeutic products), and discusses the opportunities and challenges the field of microbiome research in PD is facing.
Collapse
Affiliation(s)
- Marina Lorente-Picón
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Ariadna Laguna
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| |
Collapse
|
12
|
Lee HS, Lobbestael E, Vermeire S, Sabino J, Cleynen I. Inflammatory bowel disease and Parkinson's disease: common pathophysiological links. Gut 2021; 70:408-417. [PMID: 33067333 DOI: 10.1136/gutjnl-2020-322429] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/19/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022]
Abstract
Inflammatory bowel disease and Parkinson's disease are chronic progressive disorders that mainly affect different organs: the gut and brain, respectively. Accumulating evidence has suggested a bidirectional link between gastrointestinal inflammation and neurodegeneration, in accordance with the concept of the 'gut-brain axis'. Moreover, recent population-based studies have shown that inflammatory bowel disease might increase the risk of Parkinson's disease. Although the precise mechanisms underlying gut-brain interactions remain elusive, some of the latest findings have begun to explain the link. Several genetic loci are shared between both disorders with a similar direction of effect on the risk of both diseases. The most interesting example is LRRK2 (leucine-rich repeat kinase 2), initially identified as a causal gene in Parkinson's disease, and recently also implicated in Crohn's disease. In this review, we highlight recent findings on the link between these seemingly unrelated diseases with shared genetic susceptibility. We discuss supporting and conflicting data obtained from epidemiological and genetic studies along with remaining questions and concerns. In addition, we discuss possible biological links including the gut-brain axis, microbiota, autoimmunity, mitochondrial function and autophagy.
Collapse
Affiliation(s)
- Ho-Su Lee
- Department of Human Genetics, KU Leuven, Leuven, Belgium.,Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Evy Lobbestael
- Laboratory for Neurobiology and Gene Therapy, KU Leuven, Leuven, Belgium
| | - Séverine Vermeire
- Department of Chronic diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium.,Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - João Sabino
- Department of Chronic diseases, Metabolism and Ageing, KU Leuven, Leuven, Belgium.,Department of Gastroenterology and Hepatology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
13
|
Menozzi E, Macnaughtan J, Schapira AHV. LRRK2 Parkinsonism: Does the Response to Gut Bacteria Mitigate the Neurological Picture? Mov Disord 2020; 36:71-75. [PMID: 33107648 DOI: 10.1002/mds.28347] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/02/2020] [Accepted: 09/23/2020] [Indexed: 12/23/2022] Open
Affiliation(s)
- Elisa Menozzi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - Jane Macnaughtan
- Institute for Liver and Digestive Health, University College London, London, UK
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
14
|
Rideout HJ, Chartier-Harlin MC, Fell MJ, Hirst WD, Huntwork-Rodriguez S, Leyns CEG, Mabrouk OS, Taymans JM. The Current State-of-the Art of LRRK2-Based Biomarker Assay Development in Parkinson's Disease. Front Neurosci 2020; 14:865. [PMID: 33013290 PMCID: PMC7461933 DOI: 10.3389/fnins.2020.00865] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/24/2020] [Indexed: 12/22/2022] Open
Abstract
Evidence is mounting that LRRK2 function, particularly its kinase activity, is elevated in multiple forms of Parkinson's disease, both idiopathic as well as familial forms linked to mutations in the LRRK2 gene. However, sensitive quantitative markers of LRRK2 activation in clinical samples remain at the early stages of development. There are several measures of LRRK2 activity that could potentially be used in longitudinal studies of disease progression, as inclusion/exclusion criteria for clinical trials, to predict response to therapy, or as markers of target engagement. Among these are levels of LRRK2, phosphorylation of LRRK2 itself, either by other kinases or via auto-phosphorylation, its in vitro kinase activity, or phosphorylation of downstream substrates. This is advantageous on many levels, in that multiple indices of elevated kinase activity clearly strengthen the rationale for targeting this kinase with novel therapeutic candidates, and provide alternate markers of activation in certain tissues or biofluids for which specific measures are not detectable. However, this can also complicate interpretation of findings from different studies using disparate measures. In this review we discuss the current state of LRRK2-focused biomarkers, the advantages and disadvantages of the current pallet of outcome measures, the gaps that need to be addressed, and the priorities that the field has defined.
Collapse
Affiliation(s)
- Hardy J. Rideout
- Division of Basic Neurosciences, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Marie-Christine Chartier-Harlin
- Univ. Lille, Inserm, CHU Lille, U1172 - Lille Neuroscience & Cognition, Lille, France
- Inserm, UMR-S 1172, Team “Brain Biology and Chemistry”, Lille, France
| | | | | | | | | | | | - Jean-Marc Taymans
- Univ. Lille, Inserm, CHU Lille, U1172 - Lille Neuroscience & Cognition, Lille, France
- Inserm, UMR-S 1172, Team “Brain Biology and Chemistry”, Lille, France
| |
Collapse
|
15
|
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: 90] [Impact Index Per Article: 18.0] [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
|
16
|
Candelario KM, Balaj L, Zheng T, Skog J, Scheffler B, Breakefield X, Schüle B, Steindler DA. Exosome/microvesicle content is altered in leucine-rich repeat kinase 2 mutant induced pluripotent stem cell-derived neural cells. J Comp Neurol 2019; 528:1203-1215. [PMID: 31743443 DOI: 10.1002/cne.24819] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles, including exosomes/microvesicles (EMVs), have been described as sensitive biomarkers that represent disease states and response to therapies. In light of recent reports of disease-mirroring EMV molecular signatures, the present study profiled two EMVs from different Parkinson's disease (PD) tissue sources: (a) neural progenitor cells derived from an endogenous adult stem/progenitor cell, called adult human neural progenitor (AHNP) cells, that we found to be pathological when isolated from postmortem PD patients' substantia nigra; and (b) leucine-rich repeat kinase 2 (LRRK2) gene identified patient induced pluripotent stem cells (iPSCs), which were used to isolate EMVs and begin to characterize their cargoes. Initial characterization of EMVs derived from idiopathic patients (AHNPs) and mutant LRRK2 patients showed differences between both phenotypes and when compared with a sibling control in EMV size and release based on Nanosight analysis. Furthermore, molecular profiling disclosed that neurodegenerative-related gene pathways altered in PD can be reversed using gene-editing approaches. In fact, the EMV cargo genes exhibited normal expression patterns after gene editing. This study shows that EMVs have the potential to serve as sensitive biomarkers of disease state in both idiopathic and gene-identified PD patients and that following gene-editing, EMVs reflect a corrected state. This is relevant for both prodromal and symptomatic patient populations where potential responses to therapies can be monitored via non-invasive liquid biopsies and EMV characterizations.
Collapse
Affiliation(s)
- Kate M Candelario
- Department of Neurological Surgery, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Leonora Balaj
- Massachusetts General Hospital and Harvard University, Boston, Massachusetts
| | - Tong Zheng
- JM USDA Human Nutrition Research Center on Aging, and CTSI of Tufts University, Boston, Massachusetts
| | - Johan Skog
- Exosome Diagnostics, Inc., Cambridge, Massachusetts
| | - Bjorn Scheffler
- DKFZ-Division of Translational Oncology/Neurooncology, German Cancer Consortium (DKTK), Heidelberg & University Hospital Essen, Essen, Germany
| | - Xandra Breakefield
- Massachusetts General Hospital and Harvard University, Boston, Massachusetts
| | - Birgitt Schüle
- Department of Pathology, Stanford University, Stanford, California
| | - Dennis A Steindler
- Department of Neurological Surgery, McKnight Brain Institute, University of Florida, Gainesville, Florida.,JM USDA Human Nutrition Research Center on Aging, and CTSI of Tufts University, Boston, Massachusetts
| |
Collapse
|
17
|
Maekawa T, Tsushima H, Kawakami F, Kawashima R, Kodo M, Imai M, Ichikawa T. Leucine-Rich Repeat Kinase 2 Is Associated With Activation of the Paraventricular Nucleus of the Hypothalamus and Stress-Related Gastrointestinal Dysmotility. Front Neurosci 2019; 13:905. [PMID: 31555076 PMCID: PMC6727664 DOI: 10.3389/fnins.2019.00905] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 08/13/2019] [Indexed: 01/27/2023] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a molecule associated with familial and sporadic Parkinson's disease. It regulates many central neuronal functions, such as cell proliferation, apoptosis, autophagy, and axonal extension. Recently, it has been revealed that LRRK2 is related to anxiety/depression-like behavior, implying an association between LRRK2 and stress. In the present study, we investigated for the first time the stress pathway and its relationship to gastrointestinal motility in LRRK2-knockout (KO) mice. The mice were subjected to acute restraint stress, and analyzed for activation of the paraventricular nucleus of the hypothalamus (PVN) using an immunohistochemical approach. Phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2) was assessed by Western blotting. The KO mice showed a lower number of c-Fos-positive cells and disruption of the ERK signaling pathway in the PVN in the presence of restraint stress. Stress responses in terms of both upper and lower gastrointestinal motility were alleviated in the mice, accompanied by lower c-Fos immunoreactivity in enteric excitatory neurons. Our present findings suggest that LRRK2 is a newly recognized molecule regulating the stress pathway in the PVN, playing a role in stress-related gastrointestinal dysmotility.
Collapse
Affiliation(s)
- Tatsunori Maekawa
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
| | - Hiromichi Tsushima
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan.,Department of Behavioral Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Fumitaka Kawakami
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
| | - Rei Kawashima
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
| | - Masaru Kodo
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
| | - Motoki Imai
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
| | - Takafumi Ichikawa
- Department of Regulation Biochemistry, Graduate School of Medical Sciences, Kitasato University, Sagamihara, Japan
| |
Collapse
|
18
|
Response of Beef Cattle Fecal Microbiota to Grazing on Toxic Tall Fescue. Appl Environ Microbiol 2019; 85:AEM.00032-19. [PMID: 31126949 DOI: 10.1128/aem.00032-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 05/21/2019] [Indexed: 12/20/2022] Open
Abstract
Tall fescue, the predominant southeastern United States cool-season forage grass, frequently becomes infected with an ergot alkaloid-producing toxic endophyte, Epichloë coenophialum Consumption of endophyte-infected fescue results in fescue toxicosis (FT), a condition that lowers beef cow productivity. Limited data on the influence of ergot alkaloids on rumen fermentation profiles or ruminal bacteria that could degrade the ergot alkaloids are available, but how FT influences the grazing bovine fecal microbiota or what role fecal microbiota might play in FT etiology and associated production losses has yet to be investigated. Here, we used 16S rRNA gene sequencing of fecal samples from weaned Angus steers grazing toxic endophyte-infected (E+; n = 6) or nontoxic (Max-Q; n = 6) tall fescue before and 1, 2, 14, and 28 days after pasture assignment. Bacteria in the Firmicutes and Bacteroidetes phyla comprised 90% of the Max-Q and E+ steer fecal microbiota throughout the trial. Early decreases in the Erysipelotrichaceae family and delayed increases of the Ruminococcaceae and Lachnospiraceae families were among the major effects of E+ grazing. E+ also increased abundances within the Planctomycetes, Chloroflexi, and Proteobacteria phyla and the Clostridiaceae family. Multiple operational taxonomic units classified as Ruminococcaceae and Lachnospiraceae were correlated negatively with weight gains (lower in E+) and positively with respiration rates (increased by E+). These data provide insights into how E+ grazing alters the Angus steer microbiota and the relationship of fecal microbiota dynamics with FT.IMPORTANCE Consumption of E+ tall fescue has an estimated annual $1 billion negative impact on the U.S. beef industry, with one driver of these costs being lowered weight gains. As global agricultural demand continues to grow, mitigating production losses resulting from grazing the predominant southeastern United States forage grass is of great value. Our investigation of the effects of E+ grazing on the fecal microbiota furthers our understanding of bovine fescue toxicosis in a real-world grazing production setting and provides a starting point for identifying easy-to-access fecal bacteria that could serve as potential biomarkers of animal productivity and/or FT severity for tall fescue-grazing livestock.
Collapse
|
19
|
|