1
|
Rajagopal K, Naseri N, Parastesh F, Ghassemi F, Jafari S. Phase synchronization analysis of EEG functional connectivity in Parkinson's disease. J Theor Biol 2025; 598:111997. [PMID: 39571647 DOI: 10.1016/j.jtbi.2024.111997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 10/04/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024]
Abstract
There is a growing need for research on Parkinson's disease (PD), a neurological condition that often affects the elderly. By examining brain network connectivity, researchers are able to discover how different brain regions interact during various cognitive and behavioral tasks. They can also understand how changes in nonlinear connections may be linked to neurological and mental illnesses. In this paper, the synchrony levels of 59 EEG channels from 26 Parkinson's patients and 13 healthy subjects is examined by utilizing Phase-Lag Index (PLI) during a motor task and resting-state conditions. Examining different EEG frequency bands shows that whole-brain synchronization in the delta band is significantly lower in PD patients compared to healthy subjects during the task. PD patients also exhibit a lower clustering coefficient and a higher shortest path length in this band during the task, which shows the higher small-worldness in Parkinson's patients compared to healthy individuals. Moreover, the global efficiency in the delta band is significantly reduced in PD patients during the task. An analysis of local efficiency shows that PD and control groups differ in 57 channels. These results reveal that Parkinson's patients appear to have considerable pathological abnormalities in their delta band connectivity and its characteristic features.
Collapse
Affiliation(s)
- Karthikeyan Rajagopal
- Center for Research, SRM Easwari Engineering College, Chennai, India; Center for Research, SRM Institute of Science and Technology-Ramapuram, Chennai, India
| | - Nafise Naseri
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Iran
| | - Fatemeh Parastesh
- Center for Research, SRM Easwari Engineering College, Chennai, India; Center for Research, SRM Institute of Science and Technology-Ramapuram, Chennai, India.
| | - Farnaz Ghassemi
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Iran
| | - Sajad Jafari
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Iran; Health Technology Research Institute, Amirkabir University of Technology (Tehran Polytechnic), Iran
| |
Collapse
|
2
|
Tustumi F, Ho V, Payne SC, Carra RB. Editorial: Gastrointestinal autonomic disorders. Front Neurol 2024; 15:1492409. [PMID: 39403269 PMCID: PMC11471610 DOI: 10.3389/fneur.2024.1492409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 09/17/2024] [Indexed: 01/04/2025] Open
Affiliation(s)
- Francisco Tustumi
- Department of Gastroenterology, Instituto do cancer do Estado de São Paulo, São Paulo, SP, Brazil
- Department of Health Sciences, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Vincent Ho
- Department of Gastroenterology, Western Sydney University, Penrith, NSW, Australia
| | - Sophie Clementine Payne
- Department of Medical Bionics, University of Melbourne, Melbourne, VIC, Australia
- Bionics Institute, Melbourne, VIC, Australia
| | | |
Collapse
|
3
|
Mitchell CL, Kurouski D. Novel strategies in Parkinson's disease treatment: a review. Front Mol Neurosci 2024; 17:1431079. [PMID: 39183754 PMCID: PMC11341544 DOI: 10.3389/fnmol.2024.1431079] [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: 05/11/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024] Open
Abstract
An unprecedented extension of life expectancy observed during the past century drastically increased the number of patients diagnosed with Parkinson's diseases (PD) worldwide. Estimated costs of PD alone reached $52 billion per year, making effective neuroprotective treatments an urgent and unmet need. Current treatments of both AD and PD focus on mitigating the symptoms associated with these pathologies and are not neuroprotective. In this review, we discuss the most advanced therapeutic strategies that can be used to treat PD. We also critically review the shift of the therapeutic paradigm from a small molecule-based inhibition of protein aggregation to the utilization of natural degradation pathways and immune cells that are capable of degrading toxic amyloid deposits in the brain of PD patients.
Collapse
Affiliation(s)
- Charles L. Mitchell
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, United States
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| | - Dmitry Kurouski
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, TX, United States
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States
| |
Collapse
|
4
|
Grotewold N, Albin RL. Update: Protective and risk factors for Parkinson disease. Parkinsonism Relat Disord 2024; 125:107026. [PMID: 38879999 DOI: 10.1016/j.parkreldis.2024.107026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 06/02/2024] [Accepted: 06/04/2024] [Indexed: 06/18/2024]
Abstract
We review the epidemiologic literature on potential protective and risk factors in Parkinson's Disease (PD). Prior research identified numerous possible protective and risk factors. Potential protective factors include tobacco abuse, physical activity, urate levels, NSAID use, calcium channel blocker use, statin use, and use of some α1-adrenergic antagonists. Some potential protective factors could be products of reverse causation, including increased serum urate, tobacco abuse, and coffee-tea-caffeine consumption. Potential risk factors include traumatic brain injury, pesticide exposure, organic solvent exposure, lead exposure, air pollution, Type 2 Diabetes, some dairy products, cardiovascular disease, and some infections including Hepatitis C, H. pylori, and COVID-19. Potential non-environmental risk factors include bipolar disorder, essential tremor, bullous pemphigoid, and inflammatory bowel disease. There is an inverse relationship with PD and risk of most cancers. Though many potential protective and risk factors for PD were identified, research has not yet led to unique, rigorous prevention trials or successful disease-modifying interventions. While efforts to reduce exposure to some industrial toxicants are well justified, PD incidence might be most effectively reduced by mitigation of risks, such as Type 2 Diabetes, air pollution, traumatic brain injury, or physical inactivity, that are general public health intervention targets.
Collapse
Affiliation(s)
- Nikolas Grotewold
- Dept. of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Roger L Albin
- Dept. of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA; GRECC & Neurology Service, VAAAHS, Ann Arbor, MI, 48105, USA; University of Michigan Morris K. Udall Center of Excellence for Parkinson's Disease Research, Ann Arbor, MI, 48109, USA; University of Michigan Parkinson's Foundation Research Center of Excellence, USA.
| |
Collapse
|
5
|
Homolak J, Joja M, Grabaric G, Schiatti E, Virag D, Babic Perhoc A, Knezovic A, Osmanovic Barilar J, Salkovic-Petrisic M. The Absence of Gastrointestinal Redox Dyshomeostasis in the Brain-First Rat Model of Parkinson's Disease Induced by Bilateral Intrastriatal 6-Hydroxydopamine. Mol Neurobiol 2024; 61:5481-5493. [PMID: 38200352 PMCID: PMC11249596 DOI: 10.1007/s12035-023-03906-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024]
Abstract
The gut-brain axis plays an important role in Parkinson's disease (PD) by acting as a route for vagal propagation of aggregated α-synuclein in the gut-first endophenotype and as a mediator of gastrointestinal dyshomeostasis via the nigro-vagal pathway in the brain-first endophenotype of the disease. One important mechanism by which the gut-brain axis may promote PD is by regulating gastrointestinal redox homeostasis as overwhelming evidence suggests that oxidative stress plays a key role in the etiopathogenesis and progression of PD and the gastrointestinal tract maintains redox homeostasis of the organism by acting as a critical barrier to environmental and microbiological electrophilic challenges. The present aim was to utilize the bilateral intrastriatal 6-hydroxydopamine (6-OHDA) brain-first PD model to study the effects of isolated central pathology on redox homeostasis of the gastrointestinal tract. Three-month-old male Wistar rats were either not treated (intact controls; CTR) or treated bilaterally intrastriatally with vehicle (CIS) or 6-OHDA (6-OHDA). Motor deficits were assessed with the rotarod performance test, and the duodenum, ileum, and colon were dissected for biochemical analyses 12 weeks after the treatment. Lipid peroxidation, total antioxidant capacity, low-molecular-weight thiols, and protein sulfhydryls, the activity of total and Mn/Fe superoxide dismutases, and total and azide-insensitive catalase/peroxidase were measured. Both univariate and multivariate models analyzing redox biomarkers indicate that significant disturbances in gastrointestinal redox balance are not present. The findings demonstrate that motor impairment observed in the brain-first 6-OHDA model of PD can occur without concurrent redox imbalances in the gastrointestinal system.
Collapse
Affiliation(s)
- Jan Homolak
- Department of Pharmacology & Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia.
- Interfaculty Institute of Microbiology and Infection Medicine & Cluster of Excellence "Controlling Microbes to Fight Infections,", University of Tübingen, Tübingen, Germany.
| | - Mihovil Joja
- Department of Pharmacology & Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Gracia Grabaric
- Department of Pharmacology & Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia
| | - Emiliano Schiatti
- Faculty of Medicine and Surgery, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Davor Virag
- Department of Pharmacology & Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia
| | - Ana Babic Perhoc
- Department of Pharmacology & Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia
| | - Ana Knezovic
- Department of Pharmacology & Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia
| | - Jelena Osmanovic Barilar
- Department of Pharmacology & Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia
| | - Melita Salkovic-Petrisic
- Department of Pharmacology & Croatian Institute for Brain Research, University of Zagreb School of Medicine, Salata 11, 10 000, Zagreb, Croatia
| |
Collapse
|
6
|
Sancho-Alonso M, Sarriés-Serrano U, Miquel-Rio L, Yanes Castilla C, Paz V, Meana JJ, Perello M, Bortolozzi A. New insights into the effects of serotonin on Parkinson's disease and depression through its role in the gastrointestinal tract. SPANISH JOURNAL OF PSYCHIATRY AND MENTAL HEALTH 2024:S2950-2853(24)00039-5. [PMID: 38992345 DOI: 10.1016/j.sjpmh.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/12/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024]
Abstract
Neuropsychiatric and neurodegenerative disorders are frequently associated with gastrointestinal (GI) co-pathologies. Although the central and enteric nervous systems (CNS and ENS, respectively) have been studied separately, there is increasing interest in factors that may contribute to conditions affecting both systems. There is compelling evidence that serotonin (5-HT) may play an important role in several gut-brain disorders. It is well known that 5-HT is essential for the development and functioning of the CNS. However, most of the body's 5-HT is produced in the GI tract. A deeper understanding of the specific effects of enteric 5-HT on gut-brain disorders may provide the basis for the development of new therapeutic targets. This review summarizes current data focusing on the important role of 5-HT in ENS development and motility, with particular emphasis on novel aspects of 5-HT signaling in conditions where CNS and ENS comorbidities are common, such as Parkinson's disease and depressive disorders.
Collapse
Affiliation(s)
- María Sancho-Alonso
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; Anatomy and Human Embryology Department, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Unai Sarriés-Serrano
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; University of the Basque Country UPV/EHU, E-48940 Leioa, Bizkaia, Spain
| | - Lluis Miquel-Rio
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Claudia Yanes Castilla
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain
| | - Verónica Paz
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - José Javier Meana
- Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain; University of the Basque Country UPV/EHU, E-48940 Leioa, Bizkaia, Spain; Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
| | - Mario Perello
- Grupo de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) y Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC-PBA), La Plata, Argentina
| | - Analia Bortolozzi
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; Systems Neuropharmacology Research Group, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; Biomedical Research Networking Center for Mental Health (CIBERSAM), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain.
| |
Collapse
|
7
|
Recinto SJ, Premachandran S, Mukherjee S, Allot A, MacDonald A, Yaqubi M, Gruenheid S, Trudeau LE, Stratton JA. Characterizing enteric neurons in dopamine transporter (DAT)-Cre reporter mice reveals dopaminergic subtypes with dual-transmitter content. Eur J Neurosci 2024; 59:2465-2482. [PMID: 38487941 DOI: 10.1111/ejn.16307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/08/2024] [Accepted: 02/17/2024] [Indexed: 05/22/2024]
Abstract
The enteric nervous system (ENS) comprises a complex network of neurons whereby a subset appears to be dopaminergic although the characteristics, roles, and implications in disease are less understood. Most investigations relating to enteric dopamine (DA) neurons rely on immunoreactivity to tyrosine hydroxylase (TH)-the rate-limiting enzyme in the production of DA. However, TH immunoreactivity is likely to provide an incomplete picture. This study herein provides a comprehensive characterization of DA neurons in the gut using a reporter mouse line, expressing a fluorescent protein (tdTomato) under control of the DA transporter (DAT) promoter. Our findings confirm a unique localization of DA neurons in the gut and unveil the discrete subtypes of DA neurons in this organ, which we characterized using both immunofluorescence and single-cell transcriptomics, as well as validated using in situ hybridization. We observed distinct subtypes of DAT-tdTomato neurons expressing co-transmitters and modulators across both plexuses; some of them likely co-releasing acetylcholine, while others were positive for a slew of canonical DAergic markers (TH, VMAT2 and GIRK2). Interestingly, we uncovered a seemingly novel population of DA neurons unique to the ENS which was ChAT/DAT-tdTomato-immunoreactive and expressed Grp, Calcb, and Sst. Given the clear heterogeneity of DAergic gut neurons, further investigation is warranted to define their functional signatures and decipher their implication in disease.
Collapse
Affiliation(s)
- Sherilyn Junelle Recinto
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Shobina Premachandran
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Sriparna Mukherjee
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Pharmacology and Physiology, Department of Neurosciences, Université de Montreal, Faculty of Medicine, SNC and CIRCA Research Groups, Montreal, Quebec, Canada
| | - Alexis Allot
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
| | - Adam MacDonald
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Moein Yaqubi
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Samantha Gruenheid
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Louis-Eric Trudeau
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
- Department of Pharmacology and Physiology, Department of Neurosciences, Université de Montreal, Faculty of Medicine, SNC and CIRCA Research Groups, Montreal, Quebec, Canada
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery Montreal Neurological Institute-Hospital, McGill University, Montreal, Quebec, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| |
Collapse
|
8
|
Al‐kuraishy HM, Al‐Gareeb AI, Albuhadily AK, Elewa YHA, AL‐Farga A, Aqlan F, Zahran MH, Batiha GE. Sleep disorders cause Parkinson's disease or the reverse is true: Good GABA good night. CNS Neurosci Ther 2024; 30:e14521. [PMID: 38491789 PMCID: PMC10943276 DOI: 10.1111/cns.14521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/03/2023] [Accepted: 10/23/2023] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurodegenerative brain disease due to degeneration of dopaminergic neurons (DNs) presented with motor and non-motor symptoms. PD symptoms are developed in response to the disturbance of diverse neurotransmitters including γ-aminobutyric acid (GABA). GABA has a neuroprotective effect against PD neuropathology by protecting DNs in the substantia nigra pars compacta (SNpc). It has been shown that the degeneration of GABAergic neurons is linked with the degeneration of DNs and the progression of motor and non-motor PD symptoms. GABA neurotransmission is a necessary pathway for normal sleep patterns, thus deregulation of GABAergic neurotransmission in PD could be the potential cause of sleep disorders in PD. AIM Sleep disorders affect GABA neurotransmission leading to memory and cognitive dysfunction in PD. For example, insomnia and short sleep duration are associated with a reduction of brain GABA levels. Moreover, PD-related disorders including rigidity and nocturia influence sleep patterns leading to fragmented sleep which may also affect PD neuropathology. However, the mechanistic role of GABA in PD neuropathology regarding motor and non-motor symptoms is not fully elucidated. Therefore, this narrative review aims to clarify the mechanistic role of GABA in PD neuropathology mainly in sleep disorders, and how good GABA improves PD. In addition, this review of published articles tries to elucidate how sleep disorders such as insomnia and REM sleep behavior disorder (RBD) affect PD neuropathology and severity. The present review has many limitations including the paucity of prospective studies and most findings are taken from observational and preclinical studies. GABA involvement in the pathogenesis of PD has been recently discussed by recent studies. Therefore, future prospective studies regarding the use of GABA agonists in the management of PD are suggested to observe their distinct effects on motor and non-motor symptoms. CONCLUSION There is a bidirectional relationship between the pathogenesis of PD and sleep disorders which might be due to GABA deregulation.
Collapse
Affiliation(s)
- Hayder M. Al‐kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Ali K. Albuhadily
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Yaser Hosny Ali Elewa
- Department of Histology and Cytology, Faculty of Veterinary MedicineZagazig UniversityZagazigEgypt
- Faculty of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Ammar AL‐Farga
- Biochemistry Department, College of SciencesUniversity of JeddahJeddahSaudia Arbia
| | - Faisal Aqlan
- Department of Chemistry, College of SciencesIbb UniversityIbb GovernorateYemen
| | | | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhur UniversityDamanhurEgypt
| |
Collapse
|
9
|
Bicknell B, Liebert A, Herkes G. Parkinson's Disease and Photobiomodulation: Potential for Treatment. J Pers Med 2024; 14:112. [PMID: 38276234 PMCID: PMC10819946 DOI: 10.3390/jpm14010112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/07/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024] Open
Abstract
Parkinson's disease is the second most common neurodegenerative disease and is increasing in incidence. The combination of motor and non-motor symptoms makes this a devastating disease for people with Parkinson's disease and their care givers. Parkinson's disease is characterised by mitochondrial dysfunction and neuronal death in the substantia nigra, a reduction in dopamine, accumulation of α-synuclein aggregates and neuroinflammation. The microbiome-gut-brain axis is also important in Parkinson's disease, involved in the spread of inflammation and aggregated α-synuclein. The mainstay of Parkinson's disease treatment is dopamine replacement therapy, which can reduce some of the motor signs. There is a need for additional treatment options to supplement available medications. Photobiomodulation (PBM) is a form of light therapy that has been shown to have multiple clinical benefits due to its enhancement of the mitochondrial electron transport chain and the subsequent increase in mitochondrial membrane potential and ATP production. PBM also modulates cellular signalling and has been shown to reduce inflammation. Clinically, PBM has been used for decades to improve wound healing, treat pain, reduce swelling and heal deep tissues. Pre-clinical experiments have indicated that PBM has the potential to improve the clinical signs of Parkinson's disease and to provide neuroprotection. This effect is seen whether the PBM is directed to the head of the animal or to other parts of the body (remotely). A small number of clinical trials has given weight to the possibility that using PBM can improve both motor and non-motor clinical signs and symptoms of Parkinson's disease and may potentially slow its progression.
Collapse
Affiliation(s)
- Brian Bicknell
- NICM Health Research Institute, University of Western Sydney, Westmead 2145, Australia;
| | - Ann Liebert
- NICM Health Research Institute, University of Western Sydney, Westmead 2145, Australia;
- Sydney Adventist Hospital, Wahroonga 2076, Australia
- Faculty of medicine and Health, Sydney University, Camperdown 2050, Australia
| | - Geoffrey Herkes
- Neurologist, Sydney Adventist Hospital, Wahroonga 2076, Australia;
- College of Health and Medicine, Australian National University, Canberra 2600, Australia
| |
Collapse
|
10
|
Qamar MA, Tall P, van Wamelen D, Wan YM, Rukavina K, Fieldwalker A, Matthew D, Leta V, Bannister K, Chaudhuri KR. Setting the clinical context to non-motor symptoms reflected by Park-pain, Park-sleep, and Park-autonomic subtypes of Parkinson's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 174:1-58. [PMID: 38341227 DOI: 10.1016/bs.irn.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Non-motor symptoms (NMS) of Parkinson's disease (PD) are well described in both clinical practice and the literature, enabling their management and enhancing our understanding of PD. NMS can dominate the clinical pictures and NMS subtypes have recently been proposed, initially based on clinical observations, and later confirmed in data driven analyses of large datasets and in biomarker-based studies. In this chapter, we provide an update on what is known about three common subtypes of NMS in PD. The pain (Park-pain), sleep dysfunction (Park-sleep), and autonomic dysfunction (Park-autonomic), providing an overview of their individual classification, clinical manifestation, pathophysiology, diagnosis, and potential treatments.
Collapse
Affiliation(s)
- Mubasher A Qamar
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom.
| | - Phoebe Tall
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom
| | - Daniel van Wamelen
- Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom; Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom; Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Centre of Expertise for Parkinson & Movement Disorders, Nijmegen, The Netherlands
| | - Yi Min Wan
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom; Department of Psychiatry, Ng Teng Fong General Hospital, Singapore, Singapore
| | - Katarina Rukavina
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom
| | - Anna Fieldwalker
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Central Modulation of Pain Lab, Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Donna Matthew
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom
| | - Valentina Leta
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom; Department of Clinical Neurosciences, Parkinson, and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Kirsty Bannister
- Central Modulation of Pain Lab, Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - K Ray Chaudhuri
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom
| |
Collapse
|
11
|
Magnuson FS, Christensen P, Krassioukov A, Rodriguez G, Emmanuel A, Kirshblum S, Krogh K. Neurogenic Bowel Dysfunction in Patients with Spinal Cord Injury and Multiple Sclerosis-An Updated and Simplified Treatment Algorithm. J Clin Med 2023; 12:6971. [PMID: 38002586 PMCID: PMC10672578 DOI: 10.3390/jcm12226971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/27/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Neurogenic bowel dysfunction (NBD) is a common condition in individuals with spinal cord injury (SCI) or multiple sclerosis (MS). It usually entails constipation, difficult evacuation of the rectum, and fecal incontinence (FI); often in combination. It is highly burdensome for affected patients and is correlated with poor quality of life. The current treatment algorithm, or treatment pyramid, does not completely correspond to actual clinical practice, and the known and classical pyramid contains both treatments still in their experimental stage as well as several treatments which are not available at all treatment centers. Thus, an updated treatment algorithm is called upon, and the authors of this paper therefore propose a simplified version of the treatment pyramid, aiming to guide clinicians in treating NBD.
Collapse
Affiliation(s)
- Fredrika S. Magnuson
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Peter Christensen
- Department of Surgery, Aarhus University Hospital, 8200 Aarhus, Denmark
| | - Andrei Krassioukov
- International Collaboration of Repair Discoveries (ICORD), Department of Medicine, Division of Physical Medicine and Rehabilitation, The University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Gianna Rodriguez
- Physical Medicine and Rehabilitation, Spinal Cord Injury Medicine, University of Michigan Health, Ann Arbor, MI 48108, USA
| | - Anton Emmanuel
- GI Physiology Unit, University College London Hospital, London WC1E 6DB, UK
| | - Steven Kirshblum
- Kessler Institute for Rehabilitation, West Orange, NJ 07052, USA;
- Department of Physical Medicine and Rehabilitation, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Klaus Krogh
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, 8200 Aarhus, Denmark
| |
Collapse
|
12
|
Thomasi B, Valdetaro L, Ricciardi MC, Gonçalves de Carvalho M, Fialho Tavares I, Tavares-Gomes AL. Enteric glia as a player of gut-brain interactions during Parkinson's disease. Front Neurosci 2023; 17:1281710. [PMID: 38027511 PMCID: PMC10644407 DOI: 10.3389/fnins.2023.1281710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
The enteric glia has been shown as a potential component of neuroimmune interactions that signal in the gut-brain axis during Parkinson's disease (PD). Enteric glia are a peripheral glial type found in the enteric nervous system (ENS) that, associated with enteric neurons, command various gastrointestinal (GI) functions. They are a unique cell type, with distinct phenotypes and distribution in the gut layers, which establish relevant neuroimmune modulation and regulate neuronal function. Comprehension of enteric glial roles during prodromal and symptomatic phases of PD should be a priority in neurogastroenterology research, as the reactive enteric glial profile, gastrointestinal dysfunction, and colonic inflammation have been verified during the prodromal phase of PD-a moment that may be interesting for interventions. In this review, we explore the mechanisms that should govern enteric glial signaling through the gut-brain axis to understand pathological events and verify the possible windows and pathways for therapeutic intervention. Enteric glia directly modulate several functional aspects of the intestine, such as motility, visceral sensory signaling, and immune polarization, key GI processes found deregulated in patients with PD. The search for glial biomarkers, the investigation of temporal-spatial events involving glial reactivity/signaling, and the proposal of enteric glia-based therapies are clearly demanded for innovative and intestine-related management of PD.
Collapse
Affiliation(s)
- Beatriz Thomasi
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Luisa Valdetaro
- Department of Molecular Pathobiology, NYU College of Dentistry, New York, NY, United States
| | - Maria Carolina Ricciardi
- Neuroglial Interaction Lab, Neuroscience Program, Universidade Federal Fluminense, Niterói, Brazil
| | | | - Isabela Fialho Tavares
- Neuroglial Interaction Lab, Neurobiology Department, Universidade Federal Fluminense, Niterói, Brazil
| | - Ana Lucia Tavares-Gomes
- Neuroglial Interaction Lab, Neuroscience Program, Universidade Federal Fluminense, Niterói, Brazil
- Neuroglial Interaction Lab, Neurobiology Department, Universidade Federal Fluminense, Niterói, Brazil
| |
Collapse
|
13
|
Sun J, Ludvigsson JF, Roelstraete B, Pedersen NL, Pawitan Y, Wirdefeldt K, Fang F. Gastrointestinal biopsy of normal mucosa or nonspecific inflammation and risk of neurodegenerative disease: Nationwide matched cohort study. Eur J Neurol 2023; 30:3430-3439. [PMID: 36447380 DOI: 10.1111/ene.15654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 10/02/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND AND PURPOSE Evidence has accumulated to support the early involvement of altered gastrointestinal (GI) function in neurodegenerative disease. However, risk of Alzheimer disease (AD) and Parkinson disease (PD) among individuals with a GI biopsy of normal mucosa or nonspecific inflammation is unknown. METHODS This matched cohort study included all individuals in Sweden with a GI biopsy of normal mucosa (n = 480,346) or nonspecific inflammation (n = 655,937) during 1965-2016 (exposed group) as well as their individually matched population references and unexposed full siblings. A flexible parametric model and stratified Cox model were used to estimate hazard ratio (HR) and its 95% confidence interval (CI). RESULTS Individuals with normal mucosa or nonspecific inflammation had a higher risk of AD and PD during the 20 years after biopsy. Compared with the population references, individuals with normal mucosa had an increased risk of AD (incidence rate [IR] difference = 13.53 per 100,000 person-years, HR [95% CI] = 1.15 [1.11-1.20]) and PD (IR difference = 6.72, HR [95% CI] = 1.16 [1.10-1.23]). Elevated risk was also observed for nonspecific inflammation regarding AD (IR difference = 13.28, HR [95% CI] = 1.11 [1.08-1.14]) and PD (IR difference = 6.83, HR [95% CI] = 1.10 [1.06-1.14]). Similar results were observed in subgroup and sensitivity analyses and when comparing with their unexposed siblings. CONCLUSIONS Individuals with a GI biopsy of normal mucosa or nonspecific inflammation had an increased risk of AD and PD. This adds new evidence of the early involvement of GI dysfunction in neurodegenerative disease.
Collapse
Affiliation(s)
- Jiangwei Sun
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Jonas F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
- Department of Pediatrics, Örebro University Hospital, Örebro, Sweden
- Division of Epidemiology and Public Health, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Bjorn Roelstraete
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Yudi Pawitan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Karin Wirdefeldt
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Fang Fang
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
14
|
Konings B, Villatoro L, Van den Eynde J, Barahona G, Burns R, McKnight M, Hui K, Yenokyan G, Tack J, Pasricha PJ. Gastrointestinal syndromes preceding a diagnosis of Parkinson's disease: testing Braak's hypothesis using a nationwide database for comparison with Alzheimer's disease and cerebrovascular diseases. Gut 2023; 72:2103-2111. [PMID: 37620120 DOI: 10.1136/gutjnl-2023-329685] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 07/02/2023] [Indexed: 08/26/2023]
Abstract
OBJECTIVE Braak's hypothesis states that Parkinson's disease (PD) originates in the gastrointestinal (GI) tract, and similar associations have been established for Alzheimer's disease (AD) and cerebrovascular diseases (CVD). We aimed to determine the incidence of GI syndromes and interventions preceding PD compared with negative controls (NCs), AD and CVD. DESIGN We performed a combined case-control and cohort study using TriNetX, a US based nationwide medical record network. Firstly, we compared subjects with new onset idiopathic PD with matched NCs and patients with contemporary diagnoses of AD and CVD, to investigate preceding GI syndromes, appendectomy and vagotomy. Secondly, we compared cohorts with these exposures to matched NCs for the development of PD, AD and CVD within 5 years. RESULTS We identified 24 624 PD patients in the case-control analysis and matched 18 cohorts with each exposure to their NCs. Gastroparesis, dysphagia, irritable bowel syndrome (IBS) without diarrhoea and constipation showed specific associations with PD (vs NCs, AD and CVD) in both the case-control (odds ratios (ORs) vs NCs 4.64, 3.58, 3.53 and 3.32, respectively, all p<0.0001) and cohort analyses (relative risks (RRs) vs NCs 2.43, 2.27, 1.17 and 2.38, respectively, all p<0.05). While functional dyspepsia, IBS with diarrhoea, diarrhoea and faecal incontinence were not PD specific, IBS with constipation and intestinal pseudo-obstruction showed PD specificity in the case-control (OR 4.11) and cohort analysis (RR 1.84), respectively. Appendectomy decreased the risk of PD in the cohort analysis (RR 0.48). Neither inflammatory bowel disease nor vagotomy were associated with PD. CONCLUSION Dysphagia, gastroparesis, IBS without diarrhoea and constipation might specifically predict Parkinson's disease.
Collapse
Affiliation(s)
- Bo Konings
- Translational Research Centre for Gastrointestinal Disorders (TARGID), KU Leuven University Hospitals, Leuven, Belgium
| | - Luisa Villatoro
- Department of Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Jef Van den Eynde
- Department of Cardiology, KU Leuven University Hospitals, Leuven, Belgium
| | | | - Robert Burns
- Department of Gastroenterology, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Megan McKnight
- Department of Gastroenterology, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Ken Hui
- Department of Gastroenterology, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Gayane Yenokyan
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jan Tack
- Translational Research Centre for Gastrointestinal Disorders (TARGID), KU Leuven University Hospitals, Leuven, Belgium
| | | |
Collapse
|
15
|
Vendrik KE, Chernova VO, Kuijper EJ, Terveer EM, van Hilten JJ, Contarino MF. Safety and feasibility of faecal microbiota transplantation for patients with Parkinson's disease: a protocol for a self-controlled interventional donor-FMT pilot study. BMJ Open 2023; 13:e071766. [PMID: 37798034 PMCID: PMC10565159 DOI: 10.1136/bmjopen-2023-071766] [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: 03/14/2023] [Accepted: 08/30/2023] [Indexed: 10/07/2023] Open
Abstract
INTRODUCTION Experimental studies suggest a role of gut microbiota in the pathophysiology of Parkinson's disease (PD) via the gut-brain axis. The gut microbiota can also influence the metabolism of levodopa, which is the mainstay of treatment of PD. Therefore, modifying the gut microbiota by faecal microbiota transplantation (FMT) could be a supportive treatment strategy. METHODS AND ANALYSIS We have developed a study protocol for a single-centre, prospective, self-controlled, interventional, safety and feasibility donor-FMT pilot study with randomisation and double-blinded allocation of donor faeces. The primary objectives are feasibility and safety of FMT in patients with PD. Secondary objectives include exploring whether FMT leads to alterations in motor complications (fluctuations and dyskinesias) and PD motor and non-motor symptoms (including constipation), determining alterations in gut microbiota composition, assessing donor-recipient microbiota similarities and their association with PD symptoms and motor complications, evaluating the ease of the study protocol and examining FMT-related adverse events in patients with PD. The study population will consist of 16 patients with idiopathic PD that use levodopa and experience motor complications. They will receive FMT with faeces from one of two selected healthy human donors. FMT will be administered via a gastroscope into the duodenum, after treatment with oral vancomycin, bowel lavage and domperidone. There will be seven follow-up moments during 12 months. ETHICS AND DISSEMINATION This study was approved by the Medical Ethical Committee Leiden Den Haag Delft (ref. P20.087). Study results will be disseminated through publication in peer-reviewed journals and international conferences. TRIAL REGISTRATION NUMBER International Clinical Trial Registry Platform: NL9438.
Collapse
Affiliation(s)
- Karuna Ew Vendrik
- Department of Medical Microbiology, Centre for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Vlada O Chernova
- Department of Medical Microbiology, Centre for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ed J Kuijper
- Department of Medical Microbiology, Centre for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Elisabeth M Terveer
- Department of Medical Microbiology, Centre for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Jacobus J van Hilten
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maria Fiorella Contarino
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Neurology, Haga Teaching hospital, The Hague, The Netherlands
| |
Collapse
|
16
|
Chang JJ, Gadi SR, Videnovic A, Kuo B, Pasricha TS. Impact of outpatient gastroenterology consult on pharmacotherapy and management of gastrointestinal symptoms in Parkinson's Disease. Clin Park Relat Disord 2023; 9:100215. [PMID: 37700817 PMCID: PMC10493246 DOI: 10.1016/j.prdoa.2023.100215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/14/2023] [Accepted: 08/23/2023] [Indexed: 09/14/2023] Open
Abstract
Background & aims Gastrointestinal (GI) symptoms are common in Parkinson's Disease (PD) patients, and GI dysmotility is thought to induce motor fluctuations, requiring escalation of levodopa therapy. The role of GI consultation in managing such symptoms, however, is unclear. In this study, we investigate the possible association between GI dysmotility symptoms and escalated LEDD therapy, as well as factors associated with GI consultation for PD symptom management. Methods This was a retrospective case-study of 248 PD patients evaluated by outpatient neurology at Massachusetts General Brigham Healthcare from 2018 to 2022. Logistic regression, t-test, and Fisher exact tests were performed to identify factors associated with GI consult, change in LEDD with consult, and association of consultation with GI diagnoses and treatments, respectively. Results Among 248 PD patients, 12.9% received GI consultation despite 96.8% having GI symptoms. Bloating was the primary symptom associated with receiving GI consultation (OR 3.59 [95% CI 1.47-8.88], p = 0.005). GI consultation increased the odds of receiving GI-specific medications (78.2% vs 46.3%, p = 0.001) and specialized GI diagnoses like gastroparesis (9.4% vs 0.46%, p < 0.001) and pelvic floor dysfunction (15.6% vs 0%, p < 0.0001). Interestingly, LEDD tended not to change after GI consultation, and dysmotility symptoms, including bloating, did not predict need for higher LEDD. Conclusions While treating symptoms of dysmotility may not ameliorate levodopa-based motor fluctuations as much as previously thought, GI consultations are underutilized in PD, and patients who receive GI consultation are more likely to have changes in GI diagnosis and treatment.
Collapse
Affiliation(s)
| | - Sanjay R.V. Gadi
- Department of Medicine, Duke University Health System, Durham, NC, United States
- Harvard Medical School, Boston, MA, United States
| | - Aleksandar Videnovic
- Neurological Clinical Research Institute, Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | - Braden Kuo
- Harvard Medical School, Boston, MA, United States
- Center for Neurointestinal Health, Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Trisha S. Pasricha
- Harvard Medical School, Boston, MA, United States
- Center for Neurointestinal Health, Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
17
|
Zhou W, Triadafilopoulos G, Gurland B, Halawi H, Becker L, Garcia P, Nguyen L, Miglis M, Muppidi S, Sinn D, Jaradeh S, Neshatian L. Differential Findings on Anorectal Manometry in Patients with Parkinson's Disease and Defecatory Dysfunction. Mov Disord Clin Pract 2023; 10:1074-1081. [PMID: 37476327 PMCID: PMC10354598 DOI: 10.1002/mdc3.13755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/22/2023] [Accepted: 04/15/2023] [Indexed: 07/22/2023] Open
Abstract
Introduction Gastrointestinal dysfunction, particularly constipation, is among the most common non-motor manifestations in Parkinson's Disease (PD). We aimed to identify high-resolution anorectal manometry (HR-ARM) abnormalities in patients with PD using the London Classification. Methods We conducted a retrospective review of all PD patients at our institution who underwent HR-ARM and balloon expulsion test (BET) for evaluation of constipation between 2015 and 2021. Using age and sex-specific normal values, HR-ARM recordings were re-analyzed and abnormalities were reported using the London Classification. A combination of Wilcoxon rank sum and Fisher's exact test were used. Results 36 patients (19 women) with median age 71 (interquartile range [IQR]: 69-74) years, were included. Using the London Classification, 7 (19%) patients had anal hypotension, 17 (47%) had anal hypocontractility, and 3 women had combined hypotension and hypocontractility. Anal hypocontractility was significantly more common in women compared to men. Abnormal BET and dyssynergia were noted in 22 (61%) patients, while abnormal BET and poor propulsion were only seen in 2 (5%). Men had significantly more paradoxical anal contraction and higher residual anal pressures during simulated defecation, resulting in more negative recto-anal pressure gradients. Rectal hyposensitivity was seen in nearly one third of PD patients and comparable among men and women. Conclusion Our data affirms the high prevalence of anorectal disorders in PD. Using the London Classification, abnormal expulsion and dyssynergia and anal hypocontractility were the most common findings in PD. Whether the high prevalence of anal hypocontractility in females is directly related to PD or other confounding factors will require further research.
Collapse
Affiliation(s)
- Wendy Zhou
- Division of Gastroenterology and HepatologyStanford University School of MedicineStanfordCAUSA
| | - George Triadafilopoulos
- Division of Gastroenterology and HepatologyStanford University School of MedicineStanfordCAUSA
| | - Brooke Gurland
- Division of Gastroenterology and HepatologyStanford University School of MedicineStanfordCAUSA
| | - Houssam Halawi
- Division of Gastroenterology and HepatologyStanford University School of MedicineStanfordCAUSA
| | - Laren Becker
- Division of Gastroenterology and HepatologyStanford University School of MedicineStanfordCAUSA
| | - Patricia Garcia
- Division of Gastroenterology and HepatologyStanford University School of MedicineStanfordCAUSA
| | - Linda Nguyen
- Division of Gastroenterology and HepatologyStanford University School of MedicineStanfordCAUSA
| | - Mitchell Miglis
- Stanford University, Department of Neurology and Autonomic DisordersStanford Neuroscience Health CenterStanfordCAUSA
| | - Srikanth Muppidi
- Stanford University, Department of Neurology and Autonomic DisordersStanford Neuroscience Health CenterStanfordCAUSA
| | - Dong‐In Sinn
- Stanford University, Department of Neurology and Autonomic DisordersStanford Neuroscience Health CenterStanfordCAUSA
| | - Safwan Jaradeh
- Stanford University, Department of Neurology and Autonomic DisordersStanford Neuroscience Health CenterStanfordCAUSA
| | - Leila Neshatian
- Division of Gastroenterology and HepatologyStanford University School of MedicineStanfordCAUSA
| |
Collapse
|
18
|
Matheson JAT, Holsinger RMD. The Role of Fecal Microbiota Transplantation in the Treatment of Neurodegenerative Diseases: A Review. Int J Mol Sci 2023; 24:1001. [PMID: 36674517 PMCID: PMC9864694 DOI: 10.3390/ijms24021001] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/28/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
Neurodegenerative diseases are highly prevalent but poorly understood, and with few treatment options despite decades of intense research, attention has recently shifted toward other mediators of neurological disease that may present future targets for therapeutic research. One such mediator is the gut microbiome, which communicates with the brain through the gut-brain axis and has been implicated in various neurological disorders. Alterations in the gut microbiome have been associated with numerous neurological and other diseases, and restoration of the dysbiotic gut has been shown to improve disease conditions. One method of restoring a dysbiotic gut is via fecal microbiota transplantation (FMT), recolonizing the "diseased" gut with normal microbiome. Fecal microbiota transplantation is a treatment method traditionally used for Clostridium difficile infections, but it has recently been used in neurodegenerative disease research as a potential treatment method. This review aims to present a summary of neurodegenerative research that has used FMT, whether as a treatment or to investigate how the microbiome influences pathogenesis.
Collapse
Affiliation(s)
- Julie-Anne T. Matheson
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - R. M. Damian Holsinger
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
19
|
Lefter R, Balmus I, Ciobica A, Antioch I, Ababei D, Bild W, Hritcu L, Musteata M, Timofte D, Hogas S. CENTRAL AND PERIPHERAL EFFECT OF MPTP VIA DOSE-DEPENDENT MAGNESIUM MODULATION. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2023; 19:36-48. [PMID: 37601708 PMCID: PMC10439331 DOI: 10.4183/aeb.2023.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Background Recent studies suggested that MPTP could cause gastrointestinal motility deficits additionally to its nonconclusive and controverted effects on the CNS (behavior and brain oxidative stress) in rats. A possible interaction between MPTP typical impairments and magnesium modulatory potential was previously suggested, as magnesium role was described in neuroprotection, gastrointestinal function, and oxidative stress. Aim To investigate the possible modulatory effect of several magnesium intake formulations (via drinking water) in MPTP neurotoxicity and functional gastrointestinal impairment induction. Materials and Methods Adult male Wistar rats were subjected to 3-week magnesium intake-controlled diets (magnesium depleted food and magnesium enriched drinking water) previously to acute subcutaneous MPTP treatment (30 mg/ kg body weight). Gastrointestinal motility (one hour stool collection test), and behavioral patterns (Y maze task, elevated plus maze test, open field test, forced swim test) were evaluated. Followingly, brain and bowel samples were collected, and oxidative stress was evaluated (glutathione peroxidase activity, malondial-dehyde concentrations). Results MPTP could lead to magnesium intake-dependent constipation-like gastrointestinal motility impairments, anxiety- and depressive-like affective behavior changes, and mild pain tolerance defects. Also, we found similar brain and intestinal patterns in magnesium-dependent oxidative stress. Conclusion While the MPTP effects in normal magnesium intake could be regarded as not fully relevant in rat models and limited to the current experimental conditions, the abnormalities observed in the affective behavior, gastrointestinal status, pain tolerance, peripheric and central oxidative status could be indicative of the extent of the systemic effects of MPTP that are not restricted to the CNS level, but also to gastro-intestinal system.
Collapse
Affiliation(s)
- R. Lefter
- Centre of Biomedical Research, Romanian Academy, Iasi, Romania
| | - I.M. Balmus
- “Alexandru Ioan Cuza” University of Iasi, Department of Exact Sciences and Natural Sciences, Institute of Interdisciplinary Research, Iasi, Romania
| | - A. Ciobica
- Centre of Biomedical Research, Romanian Academy, Iasi, Romania
- “Alexandru Ioan Cuza” University, Department of Biology, Faculty of Biology, Iasi, Romania
- Academy of Romanian Scientists, Bucharest, Iasi, Romania
| | - Iulia Antioch
- “Alexandru Ioan Cuza” University, Department of Biology, Faculty of Biology, Iasi, Romania
| | - D.C. Ababei
- “Grigore T. Popa” University of Medicine and Pharmacy, Department of Pharmacodynamics and Clinical Pharmac Iasi, Romania
| | - W. Bild
- Centre of Biomedical Research, Romanian Academy, Iasi, Romania
- “Grigore T. Popa” University of Medicine and Pharmacy, Department of Physiology Iasi, Romania
| | - L.D. Hritcu
- “Ion Ionescu de la Brad” University of Agricultural Science and Veterinary Medicine, Internal Medicine Clinic, Faculty of Veterinary Medicine Iasi, Romania
| | - M. Musteata
- “Ion Ionescu de la Brad” University of Agricultural Science and Veterinary Medicine, Internal Medicine Clinic, Faculty of Veterinary Medicine Iasi, Romania
| | - D. Timofte
- “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| | - S. Hogas
- “Grigore T. Popa” University of Medicine and Pharmacy, Iasi, Romania
| |
Collapse
|
20
|
Pilipovich AA, Vorob'eva OV, Makarov SA, Kuchuk AV. [Lower gastrointestinal dysfunction in patients with Parkinson's disease]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:42-49. [PMID: 38147381 DOI: 10.17116/jnevro202312312142] [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: 12/27/2023]
Abstract
OBJECTIVE To assess the lower gastrointestinal tract dysfunction in patients with Parkinson's disease (PD) and to reveal its relationships with motor and non-motor symptoms. MATERIAL AND METHODS One hundred and eighteen patients with PD of I-III Hoehn and Yahr (H&Y) stages were studied using UPDRSI-IV, Sch&En, PDQ-39, MMSE, BDI, STAI-S and STAI-T, PFS-16, NMSQ, GSRS, BSFS, AUA. Body mass index and saliva amount and lacrimation (Schirmer's test) were assessed. RESULTS Constipation from mild to moderate intensity was present in 71.2% of the patients; predominantly mild diarrhea occurred in 27.9%; alternations of diarrhea with the difficulty in intestine emptying were observed in 25.4%. We found significant correlations of constipation with the following parameters: Sch&En scales (rS=-0.291) and PDQ-39 (rS=0.478), patient's age (rS=0.275), H&Y stage (rS=0.2604), UPDRS (rS=0.254), axial motor symptoms of parkinsonism, and a number of affective and autonomic disorders, most of which were partly dopamine-resistant. Diarrhea did not affect the quality of patient's life, or depend on age, PD stage, main digital and non-motor symptoms, but directly correlated with the severity of constipation (rS=0.263) and other gastrointestinal disorders. There were no effects of dopaminergic therapy, including levodopa, dopamine-receptor-agonists, and amantadine, on the lower gastrointestinal tract dysfunction. CONCLUSION Dysfunction of the lower gastrointestinal tract (predominantly from mild to moderate intensity) was detected in most PD patients of I-III stages. Our data indicate a complex pathogenesis of the PD impaired bowel emptying, involving degeneration of non-dopaminergic structures, and the predominant influence of concomitant diseases and inadequate laxative therapy on the formation of diarrhea syndrome in PD.
Collapse
Affiliation(s)
- A A Pilipovich
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - O V Vorob'eva
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - S A Makarov
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - A V Kuchuk
- Peoples' Friendship University of Russia, Moscow, Russia
| |
Collapse
|
21
|
Schneider KM, Kim J, Bahnsen K, Heuckeroth RO, Thaiss CA. Environmental perception and control of gastrointestinal immunity by the enteric nervous system. Trends Mol Med 2022; 28:989-1005. [PMID: 36208986 DOI: 10.1016/j.molmed.2022.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/25/2022] [Accepted: 09/07/2022] [Indexed: 12/12/2022]
Abstract
The enteric nervous system (ENS) forms a versatile sensory system along the gastrointestinal tract that interacts with most cell types in the bowel. Herein, we portray host-environment interactions at the intestinal mucosal surface through the lens of the enteric nervous system. We describe local cellular interactions as well as long-range circuits between the enteric, central, and peripheral nervous systems. Additionally, we discuss recently discovered mechanisms by which enteric neurons and glia respond to biotic and abiotic environmental changes and how they regulate intestinal immunity and inflammation. The enteric nervous system emerges as an integrative sensory system with manifold immunoregulatory functions under both homeostatic and pathophysiological conditions.
Collapse
Affiliation(s)
- Kai Markus Schneider
- Microbiology Department, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, PA, USA; Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | - Jihee Kim
- Microbiology Department, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, PA, USA; Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | - Klaas Bahnsen
- Microbiology Department, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, PA, USA; Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, PA, USA
| | - Robert O Heuckeroth
- Department of Pediatrics, Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christoph A Thaiss
- Microbiology Department, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, PA, USA; Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, PA, USA.
| |
Collapse
|
22
|
Knecht L, Folke J, Dodel R, Ross JA, Albus A. Alpha-synuclein Immunization Strategies for Synucleinopathies in Clinical Studies: A Biological Perspective. Neurotherapeutics 2022; 19:1489-1502. [PMID: 36083395 PMCID: PMC9606184 DOI: 10.1007/s13311-022-01288-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2022] [Indexed: 11/29/2022] Open
Abstract
The therapeutic strategies currently available for neurodegenerative diseases such as Parkinson's disease target only the symptoms of the disease. Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy can be summarized as synucleinopathies, as they are all characterized by the aggregation and accumulation of alpha-synuclein (α-syn) in the brain. Targeting α-syn by its formation and progression opens a new and promising disease-modifying therapeutic strategy. Thus, several distinct immunotherapeutic approaches are currently being evaluated in clinical trials. The objective of this article is to review, from a biological perspective, the most important properties of these passive and active immunotherapies to point out their relevance and suitability for the treatment of synucleinopathies.
Collapse
Affiliation(s)
- Luisa Knecht
- Chair of Geriatric Medicine, University Duisburg-Essen, Essen, Germany
| | - Jonas Folke
- Chair of Geriatric Medicine, University Duisburg-Essen, Essen, Germany
- Centre for Neuroscience & Stereology, Department of Neurology, Bispebjerg-Frederiksberg Hospital, University Hospital of Copenhagen, 2400, Copenhagen, Denmark
| | - Richard Dodel
- Chair of Geriatric Medicine, University Duisburg-Essen, Essen, Germany.
| | - J Alexander Ross
- Chair of Geriatric Medicine, University Duisburg-Essen, Essen, Germany
| | - Alexandra Albus
- Chair of Geriatric Medicine, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
23
|
Klæstrup IH, Just MK, Holm KL, Alstrup AKO, Romero-Ramos M, Borghammer P, Van Den Berge N. Impact of aging on animal models of Parkinson's disease. Front Aging Neurosci 2022; 14:909273. [PMID: 35966779 PMCID: PMC9366194 DOI: 10.3389/fnagi.2022.909273] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/28/2022] [Indexed: 11/23/2022] Open
Abstract
Aging is the biggest risk factor for developing Parkinson's disease (PD), the second most common neurodegenerative disorder. Several animal models have been developed to explore the pathophysiology underlying neurodegeneration and the initiation and spread of alpha-synuclein-related PD pathology, and to investigate biomarkers and therapeutic strategies. However, bench-to-bedside translation of preclinical findings remains suboptimal and successful disease-modifying treatments remain to be discovered. Despite aging being the main risk factor for developing idiopathic PD, most studies employ young animals in their experimental set-up, hereby ignoring age-related cellular and molecular mechanisms at play. Consequently, studies in young animals may not be an accurate reflection of human PD, limiting translational outcomes. Recently, it has been shown that aged animals in PD research demonstrate a higher susceptibility to developing pathology and neurodegeneration, and present with a more disseminated and accelerated disease course, compared to young animals. Here we review recent advances in the investigation of the role of aging in preclinical PD research, including challenges related to aged animal models that are limiting widespread use. Overall, current findings indicate that the use of aged animals may be required to account for age-related interactions in PD pathophysiology. Thus, although the use of older animals has disadvantages, a model that better represents clinical disease within the elderly would be more beneficial in the long run, as it will increase translational value and minimize the risk of therapies failing during clinical studies. Furthermore, we provide recommendations to manage the challenges related to aged animal models.
Collapse
Affiliation(s)
- Ida Hyllen Klæstrup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- DANDRITE-Danish Research Institute of Translational Neuroscience, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Mie Kristine Just
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | | | - Aage Kristian Olsen Alstrup
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | - Marina Romero-Ramos
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- DANDRITE-Danish Research Institute of Translational Neuroscience, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Per Borghammer
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | - Nathalie Van Den Berge
- Institute for Clinical Medicine, Aarhus University, Aarhus, Denmark
- Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
24
|
Intestinal Transit in Early Moderate Parkinson’s Disease Correlates with Probable RBD: Subclinical Esophageal Dysmotility Does Not Correlate. PARKINSON'S DISEASE 2022; 2022:4108401. [PMID: 35873702 PMCID: PMC9307413 DOI: 10.1155/2022/4108401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 06/15/2022] [Indexed: 11/18/2022]
Abstract
Background Nonmotor symptoms, including constipation and dysphagia, are very common in Parkinson's disease (PD) and Lewy pathology is widespread in the gastrointestinal tract, particularly in the lower esophagus. Constipation and REM sleep behavior disorder (RBD) may present prior to clinical diagnosis. Yet, little is known about esophageal dysfunction and its connection to constipation in early PD. Objective This study aimed to investigate esophageal and colonic transit in early moderate PD and to study correlations between symptoms and objective measures. Methods Thirty early moderate PD patients and 28 healthy controls (HC) were included in this cross-sectional study. Esophageal transit times were determined by esophageal scintigraphy and colonic transit times by CT after radio-opaque marker ingestion. Olfaction tests, clinical evaluation, and nonmotor questionnaires were also performed. Results Distal esophageal transit times and colonic transit times were both significantly prolonged in the PD group compared to HC (p < 0.05 andp < 0.01, respectively) and a moderate-strong positive correlation was found between colonic transit time (CTT) and RBDSQ score (r = 0.61,p < 0.001). Significant correlations were also found between CTT and SCOPA-AUT scores as well as between CTT and ROME III functional constipation scores. Conclusion Colonic transit correlates with probable RBD and is more severely prolonged in early moderate PD than is the distal esophageal transit time.
Collapse
|
25
|
Gastrointestinal Dysfunction Impact on Life Quality in a Cohort of Russian Patients with Parkinson’s Disease I-III H&Y Stage. PARKINSON'S DISEASE 2022; 2022:1571801. [PMID: 35529475 PMCID: PMC9072049 DOI: 10.1155/2022/1571801] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 03/27/2022] [Accepted: 04/09/2022] [Indexed: 11/18/2022]
Abstract
Background There are still no clearly proven methods to slow down or stop the progression of Parkinson's disease (PD). Thus, improving the quality of life (QoL) of patients with PD becomes of primary importance. Autonomic dysfunction and its symptoms are known to worsen the quality of life in PD, but the degree of this influence is underinvestigated. Particularly, impacts of the separate significant gastrointestinal symptoms, such as dyspepsia, constipation, and abdominal pain, in PD should be more precisely evaluated with the help of specific scales. Objective To assess the impacts of gastrointestinal dysfunction and its symptoms on PD patient's QoL using PDQ-39. Methods 111 PD patients in the I-III Hoehn and Yahr (H&Y) stage were enrolled in the study. The following scales were applied: UPDRS III, PDQ-39, GSRS, GDSS, MMSE, BDI, STAI-S, and STAI-T. Results The linear regression model showed that the PDQ-39 SI depended on summary assessments GSRS-SI (β = 0.333, p < 0.001), BDI (β = 0.463, p < 0.001), and UPDRS III (β = 0.163, p < 0.05). The use of the stepwise method, adding GSRS-SI and UPDRS III scores to the BDI predictor, improved the model (R2 increased from 0.454 to 0.574). The investigation of GSRS domain's influence revealed that PDQ-39 SI had a significant correlation with almost all of them, but the regression analysis showed significant QoL impacts of only two factors: constipation and abdominal pain (β = 0.288, p < 0.01 and β = 0.243, p < 0.05 accordingly). Conclusions Our results suggest a considerable negative influence of depression and gastrointestinal dysfunction (especially constipation and abdominal pain) on QoL of patients with PD. Their impact on QoL in patients with I-III H&Y stages of PD is more significant than that of motor symptoms. Therefore, the correction of depression and gastrointestinal dysfunction should be prioritized in PD therapy.
Collapse
|
26
|
Multiomics implicate gut microbiota in altered lipid and energy metabolism in Parkinson's disease. NPJ Parkinsons Dis 2022; 8:39. [PMID: 35411052 PMCID: PMC9001728 DOI: 10.1038/s41531-022-00300-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 03/04/2022] [Indexed: 12/19/2022] Open
Abstract
We aimed to investigate the link between serum metabolites, gut bacterial community composition, and clinical variables in Parkinson’s disease (PD) and healthy control subjects (HC). A total of 124 subjects were part of the study (63 PD patients and 61 HC subjects). 139 metabolite features were found to be predictive between the PD and Control groups. No associations were found between metabolite features and within-PD clinical variables. The results suggest alterations in serum metabolite profiles in PD, and the results of correlation analysis between metabolite features and microbiota suggest that several bacterial taxa are associated with altered lipid and energy metabolism in PD.
Collapse
|
27
|
Roe K. An Alternative Explanation for Alzheimer's Disease and Parkinson's Disease Initiation from Specific Antibiotics, Gut Microbiota Dysbiosis and Neurotoxins. Neurochem Res 2022; 47:517-530. [PMID: 34669122 DOI: 10.1007/s11064-021-03467-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/09/2021] [Accepted: 10/11/2021] [Indexed: 01/03/2023]
Abstract
The late onset neuropathologies, including Alzheimer's disease and Parkinson's disease, have become increasingly prevalent. Their causation has been linked to genetics, gut microbiota dysbiosis (gut dysbiosis), autoimmune diseases, pathogens and exposures to neurotoxins. An alternative explanatory hypothesis is provided for their pathogenesis. Virtually everyone has pervasive daily exposures to neurotoxins, through inhalation, skin contact, direct blood transmission and through the gastrointestinal tract by ingestion. As a result, every individual has substantial and fluctuating neurotoxin blood levels. Two major barriers to neurotoxin entry into the central nervous system are the blood-brain barrier and the intestinal wall, in the absence of gut dysbiosis. Inflammation from gut dysbiosis, induced by antibiotic usage, can increase the intestinal wall permeability for neurotoxins to reach the bloodstream, and also increase the blood-brain barrier permeability to neurotoxins. Gut dysbiosis, including gut dysbiosis caused by antibiotic treatments, is an especially high risk for neurotoxin entry into the brain to cause late onset neuropathologies. Gut dysbiosis has far-reaching immune system and central nervous system effects, and even a transient gut dysbiosis can act in combination with neurotoxins, such as aluminum, mercury, lead, arsenic, cadmium, selenium, manganese, organophosphate pesticides and organochlorines, to reach neurotoxin blood levels that can initiate a late onset neuropathology, depending on an individual's age and genetic vulnerability.
Collapse
|
28
|
Voluntary Wheel Running in Old C57BL/6 Mice Reduces Age-Related Inflammation in the Colon but Not in the Brain. Cells 2022; 11:cells11030566. [PMID: 35159375 PMCID: PMC8834481 DOI: 10.3390/cells11030566] [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: 10/05/2021] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 12/04/2022] Open
Abstract
Inflammation is considered a possible cause of cognitive decline during aging. This study investigates the influence of physical activity and social isolation in old mice on their cognitive functions and inflammation. The Barnes maze task was performed to assess spatial learning and memory in 3, 9, 15, 24, and 28 months old male C57BL/6 mice as well as following voluntary wheel running (VWR) and social isolation (SI) in 20 months old mice. Inflammatory gene expression was analyzed in hippocampal and colonic samples by qPCR. Cognitive decline occurs in mice between 15 and 24 months of age. VWR improved cognitive functions while SI had negative effects. Expression of inflammatory markers changed during aging in the hippocampus (Il1a/Il6/S100b/Iba1/Adgre1/Cd68/Itgam) and colon (Tnf/Il6/Il1ra/P2rx7). VWR attenuates inflammaging specifically in the colon (Ifng/Il10/Ccl2/S100b/Iba1), while SI regulates intestinal Il1b and Gfap. Inflammatory markers in the hippocampus were not altered following VWR and SI. The main finding of our study is that both the hippocampus and colon exhibit an increase in inflammatory markers during aging, and that voluntary wheel running in old age exclusively attenuates intestinal inflammation. Based on the existence of the gut-brain axis, our results extend therapeutic approaches preserving cognitive functions in the elderly to the colon.
Collapse
|
29
|
Van Den Berge N, Ulusoy A. Animal models of brain-first and body-first Parkinson's disease. Neurobiol Dis 2022; 163:105599. [DOI: 10.1016/j.nbd.2021.105599] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/14/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022] Open
|
30
|
Passive Immunization in Alpha-Synuclein Preclinical Animal Models. Biomolecules 2022; 12:biom12020168. [PMID: 35204668 PMCID: PMC8961624 DOI: 10.3390/biom12020168] [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: 12/02/2021] [Revised: 01/07/2022] [Accepted: 01/15/2022] [Indexed: 12/20/2022] Open
Abstract
Alpha-synucleinopathies include Parkinson’s disease, dementia with Lewy bodies, pure autonomic failure and multiple system atrophy. These are all progressive neurodegenerative diseases that are characterized by pathological misfolding and accumulation of the protein alpha-synuclein (αsyn) in neurons, axons or glial cells in the brain, but also in other organs. The abnormal accumulation and propagation of pathogenic αsyn across the autonomic connectome is associated with progressive loss of neurons in the brain and peripheral organs, resulting in motor and non-motor symptoms. To date, no cure is available for synucleinopathies, and therapy is limited to symptomatic treatment of motor and non-motor symptoms upon diagnosis. Recent advances using passive immunization that target different αsyn structures show great potential to block disease progression in rodent studies of synucleinopathies. However, passive immunotherapy in clinical trials has been proven safe but less effective than in preclinical conditions. Here we review current achievements of passive immunotherapy in animal models of synucleinopathies. Furthermore, we propose new research strategies to increase translational outcome in patient studies, (1) by using antibodies against immature conformations of pathogenic αsyn (monomers, post-translationally modified monomers, oligomers and protofibrils) and (2) by focusing treatment on body-first synucleinopathies where damage in the brain is still limited and effective immunization could potentially stop disease progression by blocking the spread of pathogenic αsyn from peripheral organs to the brain.
Collapse
|
31
|
Pilipovich A, Vorob’eva O. Upper gastrointestinal tract dysfunction and its correction by dopamine agonists for patients with Parkinson’s disease of I—III stage. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:86-93. [DOI: 10.17116/jnevro202212211186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
32
|
O’Day C, Finkelstein DI, Diwakarla S, McQuade RM. A Critical Analysis of Intestinal Enteric Neuron Loss and Constipation in Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:1841-1861. [PMID: 35848035 PMCID: PMC9535602 DOI: 10.3233/jpd-223262] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/26/2022] [Indexed: 06/06/2023]
Abstract
Constipation afflicts many patients with Parkinson's disease (PD) and significantly impacts on patient quality of life. PD-related constipation is caused by intestinal dysfunction, but the etiology of this dysfunction in patients is unknown. One possible cause is neuron loss within the enteric nervous system (ENS) of the intestine. This review aims to 1) Critically evaluate the evidence for and against intestinal enteric neuron loss in PD patients, 2) Justify why PD-related constipation must be objectively measured, 3) Explore the potential link between loss of enteric neurons in the intestine and constipation in PD, 4) Provide potential explanations for disparities in the literature, and 5) Outline data and study design considerations to improve future research. Before the connection between intestinal enteric neuron loss and PD-related constipation can be confidently described, future research must use sufficiently large samples representative of the patient population (majority diagnosed with idiopathic PD for at least 5 years), implement a consistent neuronal quantification method and study design, including standardized patient recruitment criteria, objectively quantify intestinal dysfunctions, publish with a high degree of data transparency and account for potential PD heterogeneity. Further investigation into other potential influencers of PD-related constipation is also required, including changes in the function, connectivity, mitochondria and/or α-synuclein proteins of enteric neurons and their extrinsic innervation. The connection between enteric neuron loss and other PD-related gastrointestinal (GI) issues, including gastroparesis and dysphagia, as well as changes in nutrient absorption and the microbiome, should be explored in future research.
Collapse
Affiliation(s)
- Chelsea O’Day
- Gut-Axis Injury & Repair Laboratory, Department of Medicine - Western Centre for Health Research and Education (WCHRE), The University of Melbourne, Sunshine Hospital, St Albans, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Australian Institute of Musculoskeletal Science (AIMSS), Western Centre for Health Research and Education (WCHRE) Level 3 and 4, Sunshine Hospital, St Albans, VIC, Australia
| | - David Isaac Finkelstein
- Parkinson’s Disease Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Shanti Diwakarla
- Gut-Axis Injury & Repair Laboratory, Department of Medicine - Western Centre for Health Research and Education (WCHRE), The University of Melbourne, Sunshine Hospital, St Albans, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Australian Institute of Musculoskeletal Science (AIMSS), Western Centre for Health Research and Education (WCHRE) Level 3 and 4, Sunshine Hospital, St Albans, VIC, Australia
| | - Rachel Mai McQuade
- Gut-Axis Injury & Repair Laboratory, Department of Medicine - Western Centre for Health Research and Education (WCHRE), The University of Melbourne, Sunshine Hospital, St Albans, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Australian Institute of Musculoskeletal Science (AIMSS), Western Centre for Health Research and Education (WCHRE) Level 3 and 4, Sunshine Hospital, St Albans, VIC, Australia
| |
Collapse
|
33
|
The Impact of SNCA Variations and Its Product Alpha-Synuclein on Non-Motor Features of Parkinson's Disease. Life (Basel) 2021; 11:life11080804. [PMID: 34440548 PMCID: PMC8401994 DOI: 10.3390/life11080804] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s disease (PD) is a common and progressive neurodegenerative disease, caused by the loss of dopaminergic neurons in the substantia nigra pars compacta in the midbrain, which is clinically characterized by a constellation of motor and non-motor manifestations. The latter include hyposmia, constipation, depression, pain and, in later stages, cognitive decline and dysautonomia. The main pathological features of PD are neuronal loss and consequent accumulation of Lewy bodies (LB) in the surviving neurons. Alpha-synuclein (α-syn) is the main component of LB, and α-syn aggregation and accumulation perpetuate neuronal degeneration. Mutations in the α-syn gene (SNCA) were the first genetic cause of PD to be identified. Generally, patients carrying SNCA mutations present early-onset parkinsonism with severe and early non-motor symptoms, including cognitive decline. Several SNCA polymorphisms were also identified, and some of them showed association with non-motor manifestations. The functional role of these polymorphisms is only partially understood. In this review we explore the contribution of SNCA and its product, α-syn, in predisposing to the non-motor manifestations of PD.
Collapse
|
34
|
Franco R, Casanovas B, Camps J, Navarro G, Martínez-Pinilla E. Antixoxidant Supplements versus Health Benefits of Brief/Intermittent Exposure to Potentially Toxic Physical or Chemical Agents. Curr Issues Mol Biol 2021; 43:650-664. [PMID: 34287292 PMCID: PMC8929025 DOI: 10.3390/cimb43020047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/04/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022] Open
Abstract
Although antioxidants can act locally to react with an oxidant, oral administration of "antioxidants" is quite useless in treating oxidative stress in tissues. Furthermore, it does not make sense to consider a vitamin as an antioxidant, but vitamin B3 leads to the in vivo formation of compounds that are essential for reducing this stress. A rigorous treatment of the subject indicates that to deal with oxidative stress, the most direct approach is to enhance the innate antioxidant mechanisms. The question is whether this is possible through daily activities. Diets can contain the necessary components for these mechanisms or may induce the expression of the genes involved in them. Another possibility is that pro-oxidant molecules in food increase the sensitivity and power of the detoxification pathways. This option is based on well-known DNA repair mechanisms after exposure to radiation (even from the Sun), or strong evidence of induction of antioxidant capacity after exposure to powerful pro-oxidants such as H2O2. More experimental work is required to test whether some molecules in food can increase the expression of antioxidant enzymes and/or improve antioxidant mechanisms. Identifying effective molecules to achieve such antioxidant power is critical to the food and nutraceutical industries. The potential of diet-based interventions to combat oxidative stress must be viewed from a new perspective.
Collapse
Affiliation(s)
- Rafael Franco
- Department of Biochemistry and Molecular Biomedicine, School of Chemistry, University of Barcelona, 08028 Barcelona, Spain; (B.C.); (J.C.)
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Correspondence: (R.F.); (E.M.-P.); Tel.: +34-934-021-208 (R.F.)
| | - Berta Casanovas
- Department of Biochemistry and Molecular Biomedicine, School of Chemistry, University of Barcelona, 08028 Barcelona, Spain; (B.C.); (J.C.)
| | - Jordi Camps
- Department of Biochemistry and Molecular Biomedicine, School of Chemistry, University of Barcelona, 08028 Barcelona, Spain; (B.C.); (J.C.)
| | - Gemma Navarro
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona, 02028 Barcelona, Spain
| | - Eva Martínez-Pinilla
- Department of Morphology and Cell Biology, Faculty of Medicine, University of Oviedo, 33006 Oviedo, Spain
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33003 Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Correspondence: (R.F.); (E.M.-P.); Tel.: +34-934-021-208 (R.F.)
| |
Collapse
|
35
|
Li J, Dai X, Zhou L, Li X, Pan D. Edaravone Plays Protective Effects on LPS-Induced Microglia by Switching M1/M2 Phenotypes and Regulating NLRP3 Inflammasome Activation. Front Pharmacol 2021; 12:691773. [PMID: 34135761 PMCID: PMC8201503 DOI: 10.3389/fphar.2021.691773] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/17/2021] [Indexed: 11/17/2022] Open
Abstract
Parkinson’s disease is a neurodegenerative disorder in which activated microglia may appear prior to motor symptoms, but the specific therapeutic mechanisms remain unclear. This study investigated the potential effects of Edaravone (EDA) on M1/M2 polarization of microglia in rats with dopaminergic neurons damage induced by lipopolysaccharide (LPS) and its mechanism. Rats were randomly grouped as the following (n = 10): Control, EDA alone (10 mg/kg), LPS-model (LPS 5 μg), LPS + EDA (5 mg/kg) and LPS + EDA (10 mg/kg). After intragastric administration of EDA once a day for seven consecutive days, LPS was injected into SN pars unilaterally. Rotarod test, pole test, and traction test were used to analyze the intervention effect of EDA on neurobehavioral function in rats. Protein expression levels of TH, TNF-α, Arg-1, Iba-1, NLRP3 and caspase-1 were measured by immunofluorescence staining and western blot. In vitro, BV-2 cells were treated with LPS (100 ng/ml) before adding different doses of EDA. Levels of inflammatory cytokines in culture medium were detected by ELISA. Western blot and immunofluorescence were used to evaluate microglial activation and polarization. First, rotarod test, pole test, and traction test all showed that EDA mitigated motor dysfunction of PD rats. Second, pathological analysis suggested that EDA inhibited LPS-induced microglial activation and remitted declines of dopaminergic neurons. In addition, EDA shifted M1 pro-inflammatory phenotype of microglia to M2 anti-inflammatory state, while decreased expression of M1 markers (TNF-α and IL-1β) and facilitated expression of M2 markers (Arg-1 and IL-10). EDA suppressed inflammatory responses through inhibiting the expression of pro-inflammatory factors (IL-1β, IL-18 and NO), but the neuroprotective effects were invalid while siRNA NLRP3 existed. In conclusion, these results indicated that EDA could improve neurobehavioral functions and play anti-neuroinflammatory roles in PD rats, possibly by inhibiting NLPR3 inflammasome activation and regulating microglia M1/M2 polarization.
Collapse
Affiliation(s)
- Jiping Li
- Department of Neurosurgery, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Xinping Dai
- Department of Emergency, Ningbo Yinzhou No.2 Hospital, Ningbo, China
| | - Liuyi Zhou
- Operating Room, Ningbo Yinzhou No.2 Hospital, Ningbo, China
| | - Xinxiu Li
- Department of Experimental Medical Science, Hwamei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Dongxiao Pan
- Department of Neurosurgery, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| |
Collapse
|
36
|
Treatment Options for Motor and Non-Motor Symptoms of Parkinson's Disease. Biomolecules 2021; 11:biom11040612. [PMID: 33924103 PMCID: PMC8074325 DOI: 10.3390/biom11040612] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/29/2021] [Accepted: 04/15/2021] [Indexed: 12/11/2022] Open
Abstract
Parkinson’s disease (PD) usually presents in older adults and typically has both motor and non-motor dysfunctions. PD is a progressive neurodegenerative disorder resulting from dopaminergic neuronal cell loss in the mid-brain substantia nigra pars compacta region. Outlined here is an integrative medicine and health strategy that highlights five treatment options for people with Parkinson’s (PwP): rehabilitate, therapy, restorative, maintenance, and surgery. Rehabilitating begins following the diagnosis and throughout any additional treatment processes, especially vis-à-vis consulting with physical, occupational, and/or speech pathology therapist(s). Therapy uses daily administration of either the dopamine precursor levodopa (with carbidopa) or a dopamine agonist, compounds that preserve residual dopamine, and other specific motor/non-motor-related compounds. Restorative uses strenuous aerobic exercise programs that can be neuroprotective. Maintenance uses complementary and alternative medicine substances that potentially support and protect the brain microenvironment. Finally, surgery, including deep brain stimulation, is pursued when PwP fail to respond positively to other treatment options. There is currently no cure for PD. In conclusion, the best strategy for treating PD is to hope to slow disorder progression and strive to achieve stability with neuroprotection. The ultimate goal of any management program is to improve the quality-of-life for a person with Parkinson’s disease.
Collapse
|