1
|
Xing C, Chen H, Bi W, Lei T, Hang Z, Du H. Targeting 5-HT Is a Potential Therapeutic Strategy for Neurodegenerative Diseases. Int J Mol Sci 2024; 25:13446. [PMID: 39769209 PMCID: PMC11679250 DOI: 10.3390/ijms252413446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/07/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
There is increasing interest in the potential therapeutic role of 5-HT (serotonin) in the treatment of neurodegenerative diseases, which are characterized by the progressive degeneration and death of nerve cells. 5-HT is a vital neurotransmitter that plays a central role in regulating mood, cognition, and various physiological processes in the body. Disruptions in the 5-HT system have been linked to several neurological and psychiatric disorders, making it an attractive target for therapeutic intervention. Although the exact causes of neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS) are not fully understood, researchers believe that regulating the 5-HT system could help alleviate symptoms and potentially slow the progression of these diseases. Here, we delve into the potential of harnessing 5-HT as a therapeutic target for the treatment of neurodegenerative diseases. It is important to note that the current clinical drugs targeting 5-HT are still limited in the treatment of these complex diseases. Therefore, further research and clinical trials are needed to evaluate the feasibility and effectiveness of its clinical application.
Collapse
Affiliation(s)
- Cencan Xing
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Hongyu Chen
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Wangyu Bi
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Tong Lei
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Zhongci Hang
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| | - Hongwu Du
- Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China; (C.X.); (H.C.); (W.B.); (Z.H.)
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China;
| |
Collapse
|
2
|
Tanaka M, Vécsei L. Revolutionizing our understanding of Parkinson's disease: Dr. Heinz Reichmann's pioneering research and future research direction. J Neural Transm (Vienna) 2024; 131:1367-1387. [PMID: 39110245 PMCID: PMC11608389 DOI: 10.1007/s00702-024-02812-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/22/2024] [Indexed: 11/17/2024]
Abstract
Millions of individuals around the world are afflicted with Parkinson's disease (PD), a prevalent and incapacitating neurodegenerative disorder. Dr. Reichmann, a distinguished professor and neurologist, has made substantial advancements in the domain of PD research, encompassing both fundamental scientific investigations and practical applications. His research has illuminated the etiology and treatment of PD, as well as the function of energy metabolism and premotor symptoms. As a precursor to a number of neurotransmitters and neuromodulators that are implicated in the pathophysiology of PD, he has also investigated the application of tryptophan (Trp) derivatives in the disease. His principal findings and insights are summarized and synthesized in this narrative review article, which also emphasizes the challenges and implications for future PD research. This narrative review aims to identify and analyze the key contributions of Reichmann to the field of PD research, with the ultimate goal of informing future research directions in the domain. By examining Reichmann's work, the study seeks to provide a comprehensive understanding of his major contributions and how they can be applied to advance the diagnosis and treatment of PD. This paper also explores the potential intersection of Reichmann's findings with emerging avenues, such as the investigation of Trp and its metabolites, particularly kynurenines, which could lead to new insights and potential therapeutic strategies for managing neurodegenerative disorders like PD.
Collapse
Affiliation(s)
- Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, Szeged, H-6725, Hungary.
| | - László Vécsei
- HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Danube Neuroscience Research Laboratory, Tisza Lajos krt. 113, Szeged, H-6725, Hungary
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, Szeged, H-6725, Hungary
| |
Collapse
|
3
|
Chakraborty P, Gamage HKAH, Laird AS. Butyrate as a potential therapeutic agent for neurodegenerative disorders. Neurochem Int 2024; 176:105745. [PMID: 38641025 DOI: 10.1016/j.neuint.2024.105745] [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: 02/16/2024] [Revised: 04/08/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Maintaining an optimum microbial community within the gastrointestinal tract is intricately linked to human metabolic, immune and brain health. Disturbance to these microbial populations perturbs the production of vital bioactive compounds synthesised by the gut microbiome, such as short-chain fatty acids (SCFAs). Of the SCFAs, butyrate is known to be a major source of energy for colonocytes and has valuable effects on the maintenance of intestinal epithelium and blood brain barrier integrity, gut motility and transit, anti-inflammatory effects, and autophagy induction. Inducing endogenous butyrate production is likely to be beneficial for gut-brain homeostasis and for optimal neuronal function. For these reasons, butyrate has gained interest as a potential therapy for not only metabolic and immunological disorders, but also conditions related to the brain, including neurodegenerative diseases. While direct and indirect sources of butyrate, including prebiotics, probiotics, butyrate pro-drugs and glucosidase inhibitors, offer a promising therapeutic avenue, their efficacy and dosage in neurodegenerative conditions remain largely unknown. Here, we review current literature on effects of butyrate relevant to neuronal function, the impact of butyrate in a range of neurodegenerative diseases and related treatments that may have potential for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Prapti Chakraborty
- Macquarie University Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia
| | - Hasinika K A H Gamage
- School of Natural Sciences, Macquarie University, NSW, 2109, Australia; ARC Training Centre for Facilitated Advancement of Australia's Bioactives, Macquarie University, NSW, 2109, Australia
| | - Angela S Laird
- Macquarie University Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia.
| |
Collapse
|
4
|
Xie Z, Zhang M, Luo Y, Jin D, Guo X, Yang W, Zheng J, Zhang H, Zhang L, Deng C, Zheng W, Tan EK, Jin K, Zhu S, Wang Q. Healthy Human Fecal Microbiota Transplantation into Mice Attenuates MPTP-Induced Neurotoxicity via AMPK/SOD2 Pathway. Aging Dis 2023; 14:2193-2214. [PMID: 37199590 PMCID: PMC10676800 DOI: 10.14336/ad.2023.0309] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/09/2023] [Indexed: 05/19/2023] Open
Abstract
Increasing evidence has shown that gut dysbacteriosis may play a crucial role in neuroinflammation in Parkinson's disease (PD). However, the specific mechanisms that link gut microbiota to PD remain unexplored. Given the critical roles of blood-brain barrier (BBB) dysfunction and mitochondrial dysfunction in the development of PD, we aimed to evaluate the interactions among the gut microbiota, BBB, and mitochondrial resistance to oxidation and inflammation in PD. We investigated the effects of fecal microbiota transplantation (FMT) on the physiopathology of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice. The aim was to explore the role of fecal microbiota from PD patients and healthy human controls in neuroinflammation, BBB components, and mitochondrial antioxidative capacity via the AMPK/SOD2 pathway. Compared to control mice, MPTP-treated mice exhibited elevated levels of Desulfovibrio, whereas mice given FMT from PD patients exhibited enriched levels of Akkermansia and mice given FMT from healthy humans showed no significant alterations in gut microbiota. Strikingly, FMT from PD patients to MPTP-treated mice significantly aggravated motor impairments, dopaminergic neurodegeneration, nigrostriatal glial activation and colonic inflammation, and inhibited the AMPK/SOD2 signaling pathway. However, FMT from healthy human controls greatly improved the aforementioned MPTP-caused effects. Surprisingly, the MPTP-treated mice displayed a significant loss in nigrostriatal pericytes, which was restored by FMT from healthy human controls. Our findings demonstrate that FMT from healthy human controls can correct gut dysbacteriosis and ameliorate neurodegeneration in the MPTP-induced PD mouse model by suppressing microgliosis and astrogliosis, ameliorating mitochondrial impairments via the AMPK/SOD2 pathway, and restoring the loss of nigrostriatal pericytes and BBB integrity. These findings raise the possibility that the alteration in the human gut microbiota may be a risk factor for PD and provide evidence for potential application of FMT in PD preclinical treatment.
Collapse
Affiliation(s)
- Zhenchao Xie
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| | - Mahui Zhang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| | - Yuqi Luo
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| | - Dana Jin
- College of Biological Science, University of California, Davis, CA 95616, USA.
| | - Xingfang Guo
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| | - Wanlin Yang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| | - Jialing Zheng
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| | - Hongfei Zhang
- Department of Anaesthesiology, Zhujiang Hospital of Southern Medical University, Guangdong, China.
| | - Lu Zhang
- Key Laboratory of Functional Proteomics of Guangdong Province, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Southern Medical University, Guangdong, China.
| | - Chao Deng
- School of Medical, Indigenous and Health Sciences, and Molecular Horizons, University of Wollongong, Wollongong, Australia.
| | - Wenhua Zheng
- Centre of Reproduction, Development & Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Macau, China.
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore.
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Shuzhen Zhu
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
5
|
Hirayama M, Nishiwaki H, Hamaguchi T, Ohno K. Gastrointestinal disorders in Parkinson's disease and other Lewy body diseases. NPJ Parkinsons Dis 2023; 9:71. [PMID: 37147392 PMCID: PMC10160728 DOI: 10.1038/s41531-023-00511-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/20/2023] [Indexed: 05/07/2023] Open
Abstract
Parkinson's disease (PD) is pathologically characterized by the abnormal accumulation of α-synuclein fibrils (Lewy bodies) in the substantia nigra and other brain regions, although the role of Lewy bodies remains elusive. Constipation usually precedes the motor symptoms in PD, which is in accordance with the notion that α-synuclein fibrils start from the intestinal neural plexus and ascend to the brain in at least half of PD patients. The gut microbiota is likely to be involved in intestinal and brain pathologies. Analyses of the gut microbiota in PD, rapid-eye-movement sleep behavior disorder, and dementia with Lewy bodies suggest three pathological pathways. First, Akkermansia, which is increased in PD, degrades the intestinal mucus layer and increases intestinal permeability, which triggers inflammation and oxidative stress in the intestinal neural plexus. Second, decreased short-chain fatty acids (SCFAs)-producing bacteria in PD reduce the number of regulatory T cells. Third, SCFAs also aggravate microglial activation with an unelucidated pathway. In addition, in dementia with Lewy bodies (DLB), which is another form of α-synucleinopathies, increased genera, Ruminococcus torques and Collinsella, may mitigate neuroinflammation in the substantia nigra by increasing secondary bile acids. Interventions for the gut microbiota and their metabolites may potentially delay or mitigate the development and progression of PD and other Lewy body diseases.
Collapse
Affiliation(s)
- Masaaki Hirayama
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Hiroshi Nishiwaki
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomonari Hamaguchi
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
6
|
Fan L, Zhang S, Li X, Hu Z, Yang J, Zhang S, Zheng H, Su Y, Luo H, Liu X, Fan Y, Sun H, Zhang Z, Miao J, Song B, Xia Z, Shi C, Mao C, Xu Y. CHCHD2 p.Thr61Ile knock-in mice exhibit motor defects and neuropathological features of Parkinson's disease. Brain Pathol 2023; 33:e13124. [PMID: 36322611 PMCID: PMC10154378 DOI: 10.1111/bpa.13124] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/07/2022] [Indexed: 05/04/2023] Open
Abstract
The p.Thr61Ile (p.T61I) mutation in coiled-coil-helix-coiled-coil-helix domain containing 2 (CHCHD2) was deemed a causative factor in Parkinson's disease (PD). However, the pathomechanism of the CHCHD2 p.T61I mutation in PD remains unclear. Few existing mouse models of CHCHD2-related PD completely reproduce the features of PD, and no transgenic or knock-in (KI) mouse models of CHCHD2 mutations have been reported. In the present study, we generated a novel CHCHD2 p.T61I KI mouse model, which exhibited accelerated mortality, progressive motor deficits, and dopaminergic (DA) neurons loss with age, accompanied by the accumulation and aggregation of α-synuclein and p-α-synuclein in the brains of the mutant mice. The mitochondria of mouse brains and induced pluripotent stem cells (iPSCs)-derived DA neurons carrying the CHCHD2 p.T61I mutation exhibited aberrant morphology and impaired function. Mechanistically, proteomic and RNA sequencing analysis revealed that p.T61I mutation induced mitochondrial dysfunction in aged mice likely through repressed insulin-degrading enzyme (IDE) expression, resulting in the degeneration of the nervous system. Overall, this CHCHD2 p.T61I KI mouse model recapitulated the crucial clinical and neuropathological aspects of patients with PD and provided a novel tool for understanding the pathogenic mechanism and therapeutic interventions of CHCHD2-related PD.
Collapse
Affiliation(s)
- Liyuan Fan
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Academy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Shuo Zhang
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Academy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Xinwei Li
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Academy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Zhengwei Hu
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Academy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Jing Yang
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Shuyu Zhang
- Neuro‐Intensive Care UnitThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Huimin Zheng
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Yun Su
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Haiyang Luo
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Xinjing Liu
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Yu Fan
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Academy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Huifang Sun
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Academy of Medical Sciences of Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Zhongxian Zhang
- Sino‐British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Jinxin Miao
- Sino‐British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
- Academy of Chinese Medicine ScienceHenan University of Chinese MedicineZhengzhouChina
| | - Bo Song
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
| | - Zongping Xia
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Clinical Systems Biology LaboratoriesZhengzhou UniversityZhengzhouChina
| | - Changhe Shi
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| | - Chengyuan Mao
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Sino‐British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical SciencesZhengzhou UniversityZhengzhouChina
| | - Yuming Xu
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Henan Key Laboratory of Cerebrovascular DiseasesThe First Affiliated Hospital of Zhengzhou University, Zhengzhou UniversityZhengzhouChina
- Institute of NeuroscienceZhengzhou UniversityZhengzhouChina
| |
Collapse
|
7
|
Hoxhaj P, Shah S, Muyolema Arce VE, Khan W, Sadeghzadegan A, Singh S, Collado GF, Goyal A, Khawaja I, Botlaguduru D, Razzaq W, Abdin ZU, Gupta I. Ampreloxetine Versus Droxidopa in Neurogenic Orthostatic Hypotension: A Comparative Review. Cureus 2023; 15:e38907. [PMID: 37303338 PMCID: PMC10257554 DOI: 10.7759/cureus.38907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/13/2023] Open
Abstract
Neurogenic orthostatic hypotension (nOH) is a disabling problem of autonomic dysfunction in patients with Parkinson's disease, which is associated with poor quality of life and higher mortality rates. The purpose of this literature review was to explore and compare the efficacy and safety of droxidopa (an existing treatment) and ampreloxetine (a newer medication) in the treatment of nOH. We used a mixed-method literature review that addresses the epidemiology, pathophysiology, and pharmacological and non-pharmacological management of nOH in Parkinson's disease in a general way, with a more exploratory approach to droxidopa- and ampreloxetine-controlled trial studies. We included a total of 10 studies of randomized controlled trials with eight studies focused on droxidopa and two studies focused on ampreloxetine. These two drugs were analyzed and compared based on the collected individual study results. Treatment of nOH in Parkinson's disease patients with droxidopa or ampreloxetine showed clinically meaningful and statistically significant improvements relative to placebo on the components of the OHSA (Orthostatic Hypotension Symptom Assessment) composite score and OHDAS (Orthostatic Hypotension Daily Activity Scale composite scores) composite score. Droxidopa had an improved effect on daily activities, with an associated increase in standing systolic blood pressure (BP), but the long-term efficacy of droxidopa has not been documented. Standing systolic BP was maintained by ampreloxetine and worsened after the withdrawal phase. This highlights the importance of conducting further research which will help us to improve the therapeutic approach for patients with nOH and Parkinson's disease.
Collapse
Affiliation(s)
| | - Shruti Shah
- Internal Medicine, Byramjee Jeejeeboy (BJ) Medical College, Pune, IND
| | | | | | | | - Saumya Singh
- Internal Medicine, Gujarat Medical Education & Research Society (GMERS) Medical College and Hospital, Gujarat, IND
| | - Gaudy F Collado
- Internal Medicine, Fleet Medical Unit, Philippine Fleet, Philippine Navy, Cavite City, PHL
| | - Abhishek Goyal
- Internal Medicine, Kasturba Medical College, Manipal, Manipal, IND
| | - Imran Khawaja
- Internal Medicine, Ayub Medical Institute, Abbottabad, PAK
| | | | - Waleed Razzaq
- Internal Medicine, Services Hospital Lahore, Lahore, PAK
| | - Zain U Abdin
- Medicine, District Head Quarters Hospital, Faisalabad, PAK
| | - Ishita Gupta
- Medicine, Dr. Rajendra Prasad Government Medical College, Tanda, IND
| |
Collapse
|
8
|
Pinto-Costa R, Harbachova E, La Vitola P, Di Monte DA. Overexpression-Induced α-Synuclein Brain Spreading. Neurotherapeutics 2023; 20:83-96. [PMID: 36512255 PMCID: PMC10119350 DOI: 10.1007/s13311-022-01332-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2022] [Indexed: 12/15/2022] Open
Abstract
Interneuronal transfer of pathological α-synuclein species is thought to play an important role in the progressive advancement of Lewy pathology and increasing severity of clinical manifestations in Parkinson's and other diseases commonly referred to as synucleinopathies. Pathophysiological conditions and mechanisms triggering this trans-synaptic spreading bear therefore significant pathogenetic implications but have yet to be fully elucidated. In vivo experimental models support the conclusion that increased expression of intraneuronal α-synuclein can itself induce protein spreading throughout the brain as well as from the brain to peripheral tissues. For example, overexpression of α-synuclein targeted to the rodent dorsal medulla oblongata results in its transfer and accumulation into recipient axons innervating this brain region; through these axons, α-synuclein can then travel caudo-rostrally and reach other brain sites in the pons, midbrain, and forebrain. When protein overexpression is induced in the rodent midbrain, long-distance α-synuclein spreading can be followed over time; spreading-induced α-synuclein accumulation affects lower brain regions, including the dorsal motor nucleus of the vagus, proceeds through efferent axons of the vagus nerve, and is ultimately detected within vagal motor nerve endings in the gastric wall. As discussed in this review, animal models featuring α-synuclein overexpression not only support a relationship between α-synuclein burden and protein spreading but have also provided important clues on conditions/mechanisms capable of promoting interneuronal α-synuclein transfer. Intriguing findings include the relationship between neuronal activity and protein spreading and the role of oxidant stress in trans-synaptic α-synuclein mobility.
Collapse
Affiliation(s)
- Rita Pinto-Costa
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, Bonn, 53127, Germany
| | - Eugenia Harbachova
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, Bonn, 53127, Germany
| | - Pietro La Vitola
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, Bonn, 53127, Germany
| | - Donato A Di Monte
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, Bonn, 53127, Germany.
| |
Collapse
|
9
|
Avagliano C, Coretti L, Lama A, Pirozzi C, De Caro C, De Biase D, Turco L, Mollica MP, Paciello O, Calignano A, Meli R, Lembo F, Mattace Raso G. Dual-Hit Model of Parkinson's Disease: Impact of Dysbiosis on 6-Hydroxydopamine-Insulted Mice-Neuroprotective and Anti-Inflammatory Effects of Butyrate. Int J Mol Sci 2022; 23:ijms23126367. [PMID: 35742813 PMCID: PMC9223521 DOI: 10.3390/ijms23126367] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 02/04/2023] Open
Abstract
Recent evidence highlights Parkinson’s disease (PD) initiation in the gut as the prodromal phase of neurodegeneration. Gut impairment due to microbial dysbiosis could affect PD pathogenesis and progression. Here, we propose a two-hit model of PD through ceftriaxone (CFX)-induced dysbiosis and gut inflammation before the 6-hydroxydopamine (6-OHDA) intrastriatal injection to mimic dysfunctional gut-associated mechanisms preceding PD onset. Therefore, we showed that dysbiosis and gut damage amplified PD progression, worsening motor deficits induced by 6-OHDA up to 14 days post intrastriatal injection. This effect was accompanied by a significant increase in neuronal dopaminergic loss (reduced tyrosine hydroxylase expression and increased Bcl-2/Bax ratio). Notably, CFX pretreatment also enhanced systemic and colon inflammation of dual-hit subjected mice. The exacerbated inflammatory response ran in tandem with a worsening of colonic architecture and gut microbiota perturbation. Finally, we demonstrated the beneficial effect of post-biotic sodium butyrate in limiting at once motor deficits, neuroinflammation, and colon damage and re-shaping microbiota composition in this novel dual-hit model of PD. Taken together, the bidirectional communication of the microbiota–gut–brain axis and the recapitulation of PD prodromal/pathogenic features make this new paradigm a useful tool for testing or repurposing new multi-target compounds in the treatment of PD.
Collapse
Affiliation(s)
- Carmen Avagliano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
| | - Lorena Coretti
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
- Task Force on Microbiome Studies, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy;
| | - Adriano Lama
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
- Task Force on Microbiome Studies, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy;
- Correspondence: (A.L.); (G.M.R.); Tel.: +39-081678409 (A.L.); Tel.: +39-081678423 (G.M.R.)
| | - Claudio Pirozzi
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
| | - Carmen De Caro
- Department of Science of Health, School of Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy;
| | - Davide De Biase
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy;
| | - Luigia Turco
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Maria Pina Mollica
- Task Force on Microbiome Studies, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy;
- Department of Biology, University of Naples Federico II, Complesso Universitario di Monte Sant’Angelo, Cupa Nuova Cinthia 21 Edificio, 80126 Naples, Italy
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Via Delpino, 80137 Naples, Italy;
| | - Antonio Calignano
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
| | - Rosaria Meli
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
| | - Francesca Lembo
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
- Task Force on Microbiome Studies, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy;
| | - Giuseppina Mattace Raso
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy; (C.A.); (L.C.); (C.P.); (L.T.); (A.C.); (R.M.); (F.L.)
- Task Force on Microbiome Studies, University of Naples Federico II, Via Domenico Montesano, 80131 Naples, Italy;
- Correspondence: (A.L.); (G.M.R.); Tel.: +39-081678409 (A.L.); Tel.: +39-081678423 (G.M.R.)
| |
Collapse
|
10
|
Coping Styles in Patients with Parkinson’s Disease: Consideration in the Co-Designing of Integrated Care Concepts. J Pers Med 2022; 12:jpm12060921. [PMID: 35743706 PMCID: PMC9225444 DOI: 10.3390/jpm12060921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 02/08/2023] Open
Abstract
Integrated care models may help in designing care for Parkinson’s disease (PD) that is more efficient and patient-centered. However, in order to implement such models successfully, it is important to design these models around patients’ needs and preferences. Personality traits and coping styles play a well-studied important role in patients’ disease perception and their utilization of medical and social services to cope with their disease. There is evidence that coping styles remain largely unchanged over the course of PD; coping styles are defined in the early stages of life and extend over the entire lifespan of the patient. Therefore, it seems necessary to consider aspects of the personality traits and coping styles of PD patients in the development and implementation of care models. We postulate that by taking patients’ personality traits and coping styles into account, care models for PD can be designed in a more individualized and, thus, more effective way. This paper, structured in three main sections, attempts to structure the uptake of patients’ coping styles in the co-design of integrated care models. However, further studies are needed to better develop tailored care concepts to the needs of people living with PD and their individual coping styles.
Collapse
|
11
|
Murata H, Barnhill LM, Bronstein JM. Air Pollution and the Risk of Parkinson's Disease: A Review. Mov Disord 2022; 37:894-904. [PMID: 35043999 PMCID: PMC9119911 DOI: 10.1002/mds.28922] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/15/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022] Open
Abstract
Parkinson's disease, as well as other neurodegenerative disorders, are primarily characterized by pathological accumulation of proteins, inflammation, and neuron loss. Although there are some known genetic risk factors, most cases cannot be explained by genetics alone. Therefore, it is important to determine the environmental factors that confer risk and the mechanisms by which they act. Recent epidemiological studies have found that exposure to air pollution is associated with an increased risk for development of Parkinson's disease, although not all results are uniform. The variability between these studies is likely due to differences in what components of air pollution are measured, timing and methods used to determine exposures, and correction for other variables. There are several potential mechanisms by which air pollution could act to increase the risk for development of Parkinson's disease, including direct neuronal toxicity, induction of systemic inflammation leading to central nervous system inflammation, and alterations in gut physiology and the microbiome. Taken together, air pollution is an emerging risk factor in the development of Parkinson's disease. A number of potential mechanisms have been implicated by which it promotes neuropathology providing biological plausibility, and these mechanisms are likely relevant to the development of other neurodegenerative disorders such as Alzheimer's disease. This field is in its early stages, but a better understanding of how environmental exposures influence the pathogenesis of neurodegeneration is essential for reducing the incidence of disease and finding disease-modifying therapies. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | | | - Jeff M. Bronstein
- David Geffen School of Medicine at UCLA, Department of Neurology and Molecular Toxicology, 710 Westwood Plaza, Los Angeles, CA 90095
| |
Collapse
|
12
|
Marchetti B, Giachino C, Tirolo C, Serapide MF. "Reframing" dopamine signaling at the intersection of glial networks in the aged Parkinsonian brain as innate Nrf2/Wnt driver: Therapeutical implications. Aging Cell 2022; 21:e13575. [PMID: 35262262 PMCID: PMC9009237 DOI: 10.1111/acel.13575] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/11/2022] [Accepted: 02/06/2022] [Indexed: 11/30/2022] Open
Abstract
Dopamine (DA) signaling via G protein-coupled receptors is a multifunctional neurotransmitter and neuroendocrine-immune modulator. The DA nigrostriatal pathway, which controls the motor coordination, progressively degenerates in Parkinson's disease (PD), a most common neurodegenerative disorder (ND) characterized by a selective, age-dependent loss of substantia nigra pars compacta (SNpc) neurons, where DA itself is a primary source of oxidative stress and mitochondrial impairment, intersecting astrocyte and microglial inflammatory networks. Importantly, glia acts as a preferential neuroendocrine-immune DA target, in turn, counter-modulating inflammatory processes. With a major focus on DA intersection within the astrocyte-microglial inflammatory network in PD vulnerability, we herein first summarize the characteristics of DA signaling systems, the propensity of DA neurons to oxidative stress, and glial inflammatory triggers dictating the vulnerability to PD. Reciprocally, DA modulation of astrocytes and microglial reactivity, coupled to the synergic impact of gene-environment interactions, then constitute a further level of control regulating midbrain DA neuron (mDAn) survival/death. Not surprisingly, within this circuitry, DA converges to modulate nuclear factor erythroid 2-like 2 (Nrf2), the master regulator of cellular defense against oxidative stress and inflammation, and Wingless (Wnt)/β-catenin signaling, a key pathway for mDAn neurogenesis, neuroprotection, and immunomodulation, adding to the already complex "signaling puzzle," a novel actor in mDAn-glial regulatory machinery. Here, we propose an autoregulatory feedback system allowing DA to act as an endogenous Nrf2/Wnt innate modulator and trace the importance of DA receptor agonists applied to the clinic as immune modifiers.
Collapse
Affiliation(s)
- Bianca Marchetti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC)Pharmacology SectionMedical SchoolUniversity of CataniaCataniaItaly
- OASI Research Institute‐IRCCS, Troina (EN), ItalyTroinaItaly
| | | | - Cataldo Tirolo
- OASI Research Institute‐IRCCS, Troina (EN), ItalyTroinaItaly
| | - Maria F. Serapide
- Department of Biomedical and Biotechnological Sciences (BIOMETEC)Pharmacology SectionMedical SchoolUniversity of CataniaCataniaItaly
| |
Collapse
|
13
|
Onaolapo OJ, Odeniyi AO, Onaolapo AY. Parkinson's Disease: Is there a Role for Dietary and Herbal Supplements? CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 20:343-365. [PMID: 33602107 DOI: 10.2174/1871527320666210218082954] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/19/2020] [Accepted: 09/29/2020] [Indexed: 12/18/2022]
Abstract
Parkinson's Disease (PD) is characterised by degeneration of the neurons of the nigrostriatal dopaminergic pathway of the brain. The pharmacological cornerstone of PD management is mainly the use of dopamine precursors, dopamine receptor agonists, and agents that inhibit the biochemical degradation of dopamine. While these drugs initially provide relief to the symptoms and improve the quality of life of the patients, progression of the underlying pathological processes, such as oxidative stress and neuroinflammation (which have been strongly associated with PD and other neurodegenerative disorders), eventually reduce their benefits, making further benefits achievable, only at high doses due to which the magnitude and frequency of side-effects are amplified. Also, while it is becoming obvious that mainstream pharmacological agents may not always provide the much-needed answer, the question remains what succour can nature provide through dietary supplements, nutraceuticals and herbal remedies? This narrative review examines current literature for evidence of the possible roles (if any) of nutraceuticals, dietary supplements and herbal remedies in the prevention or management of PD by examining how these compounds could modulate key factors and pathways that are crucial to the pathogenesis and/or progression of PD. The likely limitations of this approach and its possible future roles in PD prevention and management are also considered.
Collapse
Affiliation(s)
- Olakunle J Onaolapo
- Behavioural Neuroscience Unit, Neuropharmacology Subdivision, Department of Pharmacology, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
| | - Ademola O Odeniyi
- Behavioural Neuroscience Unit, Neuropharmacology Subdivision, Department of Pharmacology, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
| | - Adejoke Y Onaolapo
- Behavioural Neuroscience Unit, Neurobiology Subdivision, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| |
Collapse
|
14
|
Mao L, Zhang Y, Tian J, Sang M, Zhang G, Zhou Y, Wang P. Cross-Sectional Study on the Gut Microbiome of Parkinson's Disease Patients in Central China. Front Microbiol 2021; 12:728479. [PMID: 34650532 PMCID: PMC8506127 DOI: 10.3389/fmicb.2021.728479] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/27/2021] [Indexed: 01/14/2023] Open
Abstract
Gastrointestinal dysfunction plays an important role in the occurrence and development of Parkinson's disease (PD). This study investigates the composition of the gut microbiome using shotgun metagenomic sequencing in PD patients in central China. Fecal samples from 39 PD patients (PD group) and the corresponding 39 healthy spouses of the patients (SP) were collected for shotgun metagenomics sequencing. Results showed a significantly altered microbial composition in the PD patients. Bilophila wadsworthia enrichment was found in the gut microbiome of PD patients, which has not been reported in previous studies. The random forest (RF) model, which identifies differences in microbiomes, reliably discriminated patients with PD from controls; the area under the receiver operating characteristic curve was 0.803. Further analysis of the microbiome and clinical symptoms showed that Klebsiella and Parasutterella were positively correlated with the duration and severity of PD, whereas hydrogen-generating Prevotella was negatively correlated with disease severity. The Cluster of Orthologous Groups of protein database, the KEGG Orthology database, and the carbohydrate-active enzymes of gene-category analysis showed that branched-chain amino acid-related proteins were significantly increased, and GH43 was significantly reduced in the PD group. Functional analysis of the metagenome confirmed differences in microbiome metabolism in the PD group related to short-chain fatty acid precursor metabolism.
Collapse
Affiliation(s)
- Liangwei Mao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Biological Resources, School of Life Sciences, Hubei University, Wuhan, China
| | - Yu Zhang
- Hubei Clinical Research Center of Parkinson’s Disease, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Jing Tian
- Hubei Clinical Research Center of Parkinson’s Disease, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Ming Sang
- Hubei Clinical Research Center of Parkinson’s Disease, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Guimin Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Biological Resources, School of Life Sciences, Hubei University, Wuhan, China
| | - Yuling Zhou
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Collaborative Innovation Center for Green Transformation of Biological Resources, School of Life Sciences, Hubei University, Wuhan, China
| | - Puqing Wang
- Hubei Clinical Research Center of Parkinson’s Disease, Xiangyang No. 1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| |
Collapse
|
15
|
Tucker HR, Mahoney E, Akhtar K, Kao TJ, Mamone G, Mikkilineni S, Ravi M, Watkins H, Terrelonge DL, Martin C, Unger K, Kim G, Fiber K, Gupta M, Indajang J, Kochman EM, Sachs N, Feustel P, Molho ES, Pilitsis JG, Shin DS. Motor Thalamic Deep Brain Stimulation Alters Cortical Activity and Shows Therapeutic Utility for Treatment of Parkinson's Disease Symptoms in a Rat Model. Neuroscience 2021; 460:88-106. [PMID: 33631218 DOI: 10.1016/j.neuroscience.2021.02.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 02/12/2021] [Accepted: 02/15/2021] [Indexed: 12/17/2022]
Abstract
Deep brain stimulation (DBS) in Parkinson's disease (PD) alters neuronal function and network communication to improve motor symptoms. The subthalamic nucleus (STN) is the most common DBS target for PD, but some patients experience adverse effects on memory and cognition. Previously, we reported that DBS of the ventral anterior (VA) and ventrolateral (VL) nuclei of the thalamus and at the interface between the two (VA|VL), collectively VA-VL, relieved forelimb akinesia in the hemiparkinsonian 6-hydroxydopamine (6-OHDA) rat model. To determine the mechanism(s) underlying VA-VL DBS efficacy, we examined how motor cortical neurons respond to VA-VL DBS using single-unit recording electrodes in anesthetized 6-OHDA lesioned rats. VA-VL DBS increased spike frequencies of primary (M1) and secondary (M2) motor cortical pyramidal cells and M2, but not M1, interneurons. To explore the translational merits of VA-VL DBS, we compared the therapeutic window, rate of stimulation-induced dyskinesia onset, and effects on memory between VA-VL and STN DBS. VA-VL and STN DBS had comparable therapeutic windows, induced dyskinesia at similar rates in hemiparkinsonian rats, and adversely affected performance in the novel object recognition (NOR) test in cognitively normal and mildly impaired sham animals. Interestingly, a subset of sham rats with VA-VL implants showed severe cognitive deficits with DBS off. VA-VL DBS improved NOR test performance in these animals. We conclude that VA-VL DBS may exert its therapeutic effects by increasing pyramidal cell activity in the motor cortex and interneuron activity in the M2, with plausible potential to improve memory in PD.
Collapse
Affiliation(s)
- Heidi R Tucker
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Emily Mahoney
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Kainat Akhtar
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | | | - Gianna Mamone
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Saisree Mikkilineni
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Maya Ravi
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Hanel Watkins
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Danielle-Lee Terrelonge
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Caryn Martin
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Kristen Unger
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Gabrielle Kim
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Kyra Fiber
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Megan Gupta
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Jonathan Indajang
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Eliyahu M Kochman
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Natasha Sachs
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Paul Feustel
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Eric S Molho
- Department of Neurology, Albany Medical Center, Albany, NY, USA
| | - Julie G Pilitsis
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA; Department of Neurosurgery, Albany Medical Center, Albany, NY, USA
| | - Damian S Shin
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, USA; Department of Neurology, Albany Medical Center, Albany, NY, USA.
| |
Collapse
|
16
|
Abstract
14-3-3 proteins are mostly expressed in the brain and are closely involved in numerous brain functions and various brain disorders. Among the isotypes of the 14-3-3 proteins, 14-3-3γ is mainly expressed in neurons and is highly produced during brain development, which could indicate that it has a significance in neural development. Furthermore, the distinctive levels of temporally and locally regulated 14-3-3γ expression in various brain disorders suggest that it could play a substantial role in brain plasticity of the diseased states. In this review, we introduce the various brain disorders reported to be involved with 14-3-3γ, and summarize the changes of 14-3-3γ expression in each brain disease. We also discuss the potential of 14-3-3γ for treatment and the importance of research on specific 14-3-3 isotypes for an effective therapeutic approach.
Collapse
Affiliation(s)
- Eunsil Cho
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| |
Collapse
|
17
|
Almeida SS, Zizzi FB, Cattaneo A, Comandini A, Di Dato G, Lubrano E, Pellicano C, Spallone V, Tongiani S, Torta R. Management and Treatment of Patients With Major Depressive Disorder and Chronic Diseases: A Multidisciplinary Approach. Front Psychol 2020; 11:542444. [PMID: 33101117 PMCID: PMC7546762 DOI: 10.3389/fpsyg.2020.542444] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 09/01/2020] [Indexed: 12/26/2022] Open
Abstract
In patients with physical chronic diseases, the prevalence of major depressive disorder (MDD) is approximately 2- to 3-fold higher than in the general population, and it can reach up to 20-40%. The comorbidity of MDD with chronic medical diseases is associated with poorer quality of life, increased medical symptom burden, poor adherence to self-care regimens, increased risk of functional impairment, morbidity, and mortality, and also higher medical costs. Despite this evidence, in routine practice, psychological issues and concerns are frequently inadequately managed. This consensus document proposes that a proper diagnosis, a multidisciplinary approach, and a personalized treatment plan would allow patients with MDD and chronic comorbidities to be more compliant, to improve the outcomes, to reduce possible relapses in the long term, and to prevent or better manage complications and adverse events. This proposal might be useful for any health professionals who deal with patients with chronic diseases, as it can help to pay more attention to the emotional impact of these conditions, in particular in terms of depressive symptoms.
Collapse
Affiliation(s)
- Susana Sousa Almeida
- Portuguese Institute of Oncology Porto (IPO Porto), Hospital Cuf Porto (HCuf Porto), University of Porto (FMUP), Porto, Portugal
| | | | - Agnese Cattaneo
- Angelini RR&D (Research, Regulatory & Development) - Angelini S.p.A, Rome, Italy
| | - Alessandro Comandini
- Angelini RR&D (Research, Regulatory & Development) - Angelini S.p.A, Rome, Italy
| | - Giorgio Di Dato
- Angelini RR&D (Research, Regulatory & Development) - Angelini S.p.A, Rome, Italy
| | - Ennio Lubrano
- Academic Rheumatology Unit, Dipartimento di Medicina e Scienze della salute "Vincenzo Tiberio", Università degli Studi del Molise, Campobasso, Italy
| | - Clelia Pellicano
- Laboratory of Neuropsychiatry, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Santa Lucia Foundation, Rome, Italy
| | - Vincenza Spallone
- Division of Endocrinology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Serena Tongiani
- Angelini RR&D (Research, Regulatory & Development) - Angelini S.p.A, Rome, Italy
| | - Riccardo Torta
- Clinical Psychology and Psycho-Oncology Unit, Department of Neuroscience "Rita Levi Montalcini", University of Turin, A.O.U. "Città della Salute e della Scienza" Hospital, Turin, Italy
| |
Collapse
|
18
|
Goldenberg MDF, Huang X, Chen H, Kong L, Postolache TT, Stiller JW, Ryan KA, Pavlovich M, Pollin TI, Shuldiner AR, Mailman RB, Mitchell BD. Parkinson's Disease-Related Motor and Nonmotor Symptoms in the Lancaster Amish. Neuroepidemiology 2020; 54:392-397. [PMID: 32739915 DOI: 10.1159/000509394] [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: 04/14/2020] [Accepted: 06/09/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Previous research has suggested that the Amish may experience a relatively high prevalence of Parkinson's disease (PD) and/or parkinsonian motor signs. METHODS In a large sample from the Amish community in Lancaster County, Pennsylvania, age ≥18 years, we assessed the prevalence of self-reported PD diagnosis. For those without self-reported PD diagnosis, we assessed the frequency of PD-related motor symptoms using a 9-item questionnaire that was designed by the PD Epidemiology Research Group. Lastly, we queried study participants for the presence of 2 nonmotor symptoms that have been commonly linked to PD: bowel movement frequency and daytime sleepiness. RESULTS Among 2,025 subjects who answered the PD questionnaire, 430 were older than 60 years. Of 430 participants ≥60 years, 5 (1.2%) reported a PD diagnosis. Of those without a PD diagnosis, 10.5% reported ≥1 and 1.2% ≥ 4 motor symptoms for the 9-item PD screening questionnaire. Of the 3,789 subjects who answered the question about bowel movement frequency, 0.7% reported ≤3 bowel movements per week. Among 1,710 subjects who answered the question about daytime sleepiness, 8.1% of the participants reported "always" sleepy during the day. DISCUSSION These data neither support a markedly higher PD prevalence in the older Lancaster Amish nor do they show dramatically higher motor and/or selected nonmotor symptoms than the general population. Future studies that employ more rigorous procedures for case identification and PD-specific preclinical symptoms/tests are needed to determine the potential differences and similarities among different Amish populations and between Amish and non-Amish populations.
Collapse
Affiliation(s)
- Michael D F Goldenberg
- Department of Neurology, Penn State Health-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Xuemei Huang
- Department of Neurology, Penn State Health-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA,
| | - Honglei Chen
- Department of Epidemiology, Michigan State University, East Lansing, Michigan, USA
| | - Lan Kong
- Department of Public Health Sciences, Penn State University-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Teodor T Postolache
- Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, MD, Rocky Mountain Mental Illness, Research Education and Clinical Center, Denver, Colorado, USA.,Rocky Mountain Mental Illness Research, Education and Clinical Center, Denver, Colorado, USA.,VISN 5 Capitol Health Care Network Mental Illness Research Education and Clinical Center, Baltimore, Maryland, USA
| | - John W Stiller
- Mood and Anxiety Program, University of Maryland School of Medicine, Baltimore, MD, Rocky Mountain Mental Illness, Research Education and Clinical Center, Denver, Colorado, USA.,St. Elizabeth's Hospital, Neurology Consultation Service, Washington, District of Columbia, USA
| | - Katherine A Ryan
- VISN 5 Capitol Health Care Network Mental Illness Research Education and Clinical Center, Baltimore, Maryland, USA
| | - Mary Pavlovich
- Program for Personalized and Genomic Medicine, Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Toni I Pollin
- Program for Personalized and Genomic Medicine, Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Alan R Shuldiner
- Program for Personalized and Genomic Medicine, Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Richard B Mailman
- Department of Neurology, Penn State Health-Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA.,Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Braxton D Mitchell
- Program for Personalized and Genomic Medicine, Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Geriatric Research and Education Clinical Center, Veterans Administration Medical Center, Baltimore, Maryland, USA
| |
Collapse
|
19
|
Scialò C, Legname G. How would defining Parkinson’s as a prion disease impact the search of a cure? Expert Rev Neurother 2020; 20:417-420. [DOI: 10.1080/14737175.2020.1753504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Carlo Scialò
- Department of Neuroscience, Laboratory of Prion Biology, Scuola Internazionale Superiore Di Studi Avanzati (SISSA), Trieste, Italy
| | - Giuseppe Legname
- Department of Neuroscience, Laboratory of Prion Biology, Scuola Internazionale Superiore Di Studi Avanzati (SISSA), Trieste, Italy
| |
Collapse
|
20
|
Pavlenko TA, Chesnokova NB, Nodel MR, Kim AR, Ugrumov MV. Molecular Mechanisms and Clinical Manifestations of Catecholamine Dysfunction in the Eye in Parkinson's Disease As a Basis for Developing Early Diagnosis. Acta Naturae 2020; 12:52-62. [PMID: 32742727 PMCID: PMC7385097 DOI: 10.32607/actanaturae.10906] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 04/16/2020] [Indexed: 01/08/2023] Open
Abstract
This review provides information on the non-motor peripheral manifestations of Parkinson's disease (PD) associated with a pathology of the visual analyzer and the auxiliary apparatus of the eye. The relationship between neurodegenerative processes that take place in the brain and in the eye opens new prospects to use preventive ophthalmologic examination to diagnose PD long before the characteristic motor symptoms appear. This will encourage the use of neuroprotective therapy, which stops, or at least slows down, neuronal death, instead of the current replacement therapy with dopamine agonists. An important result of an eye examination of patients with PD may be a non-invasive identification of new peripheral biomarkers manifesting themselves as changes in the composition of the lacrimal fluid.
Collapse
Affiliation(s)
- T. A. Pavlenko
- Helmholtz Moscow Research Institute of Eye Diseases of Ministry of Health of the Russian Federation, Moscow, 105062 Russia
| | - N. B. Chesnokova
- Helmholtz Moscow Research Institute of Eye Diseases of Ministry of Health of the Russian Federation, Moscow, 105062 Russia
| | - M. R. Nodel
- Sechenov First Moscow State Medical University, Moscow, 119991 Russia
- Pirogov Russian National Research Medical University, Russian Clinical and Research Center of Gerontology, Moscow, 129226 Russia
| | - A. R. Kim
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, 119334 Russia
| | - M. V. Ugrumov
- Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, 119334 Russia
| |
Collapse
|
21
|
Gruden’ MA, Solov’eva OA, Kudrin VS, Narkevich VB, Sherstnev VV. Neurochemical and Behavioral Features of Action of Pre-Fibrillar Oligomeric Structures of α-Sinuclein in Adult Mice. NEUROCHEM J+ 2020. [DOI: 10.1134/s1819712420010092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
22
|
Non-motor Symptoms in Parkinson's Disease Patients with Parkin Mutations: More Depression and Less Executive Dysfunction. J Mol Neurosci 2020; 70:246-253. [PMID: 31927768 DOI: 10.1007/s12031-019-01444-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 11/13/2019] [Indexed: 01/19/2023]
Abstract
The purpose of this study was to identify differences between genetically undefined (GU) early-onset Parkinson's disease (EOPD) patients and carriers of Parkin mutations on non-motor symptoms (NMSs). EOPD patients (N = 261) underwent targeted sequencing of Parkinson's disease (PD) related genes. Among them, 53 cases carried homozygous or compound heterozygous Parkin mutations (Parkin group) while 208 did not carry known causative PD mutations or risk factors of GBA or Parkin heterozygous mutations (GU group). NMSs were evaluated by face-to-face interviews, self-completed questionnaires and results on a neuropsychological battery. Linear regression and logistic regression models were applied to assess the predictors of NMSs. Parkin patients had younger ages of onset (AOO) (p < 0.001), longer disease durations (p < 0.001) and lower grades of Hoehn and Yarh (H&Y) (p = 0.007). Results on the neuropsychological battery showed a shorter time in Trail Making Test (TMT) (part B) in Parkin patients (p = 0.034) compared to GU patients. After adjusting for AOO, disease duration, H&Y, and levodopa equivalent daily dose (LEDD), there was a higher depression index on the Beck Depression Inventory (BDI) (p = 0.013) and better performance (p = 0.038) on executive function in the Parkin group compared to the GU group. No significant differences were found for autonomic functions, sleep-wake problems or other domains of cognitive function. Our study showed that the Parkin mutation status might be a good predictor of symptoms of depression without an impact on executive function. While these findings need to be confirmed in larger cohorts, they identify a need to screen for depression. Graphical Abstract Flow chart of genetic tests.
Collapse
|
23
|
Marino S, Cartella E, Donato N, Muscarà N, Sorbera C, Cimino V, De Salvo S, Micchìa K, Silvestri G, Bramanti A, Di Lorenzo G. Quantitative assessment of Parkinsonian tremor by using biosensor device. Medicine (Baltimore) 2019; 98:e17897. [PMID: 31860947 PMCID: PMC6940115 DOI: 10.1097/md.0000000000017897] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 06/12/2019] [Accepted: 10/11/2019] [Indexed: 01/12/2023] Open
Abstract
Parkinson disease (PD) is the second most common neurodegenerative disease which affects population older than 65 years. Tremor represents one of the main symptomatic triads in PD, particularly in rest state.We enrolled 41 idiopathic PD patients, to validate the assessment of tremor symptoms.To be enrolled in the study, patients had to fulfill the movement disorder society clinical diagnostic criteria for PD.We used an innovative home-made, low-cost device, able to quantify the frequency and amplitude of rest tremor and stress conditionOur results confirmed the presence of tremor during muscular effort in a significant number of patients and the influence of emotional stress.We suppose that this new device should be validated in clinical practice as a support of differential diagnosis and therapeutic management of PD patients.
Collapse
Affiliation(s)
| | | | - Nicola Donato
- Laboratory of Electronics for Sensors and for Systems of Transduction, Department of Engineering, University of Messina
| | | | | | | | | | | | | | - Alessia Bramanti
- Institute of Applied Sciences and Intelligent Systems “Edoardo Caianello” (ISASI), National Research Council of Italy, Messina, Italy
| | | |
Collapse
|
24
|
Abstract
Despite recent successes in understanding the genetics of Parkinson’s disease (PD), the causes of late-onset sporadic PD remain elusive. Many of the epidemiologic findings on PD etiology have been challenged by alternative explanations such as reverse causation. This is mainly because PD often takes decades to develop before it can be diagnosed late in life. Convincing evidence shows that this prodromal stage of PD is characterized by various prodromal symptoms such as olfactory impairment and rapid-eye-movement sleep behavior disorder (RBD). As they likely reflect PD pathogenesis years, if not decades, before nigrostriatal involvement, research on these symptoms may represent an unprecedented opportunity to dissect the etiology of PD. Using PD prodromal symptoms as intermediate phenotypes, we may be able to identify factors that contribute to the development of these symptoms and factors that modify their progression to clinical PD. Further, this line of research will also enable examinations of novel etiological hypotheses of PD development such as the microbiome and prion hypotheses. In this article, the author used olfactory impairment and RBD as examples to illustrate the promises and challenges of epidemiologic research on prodromal symptoms to understand PD etiology.
Collapse
Affiliation(s)
- Honglei Chen
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
25
|
Ghaisas S, Langley MR, Palanisamy BN, Dutta S, Narayanaswamy K, Plummer PJ, Sarkar S, Ay M, Jin H, Anantharam V, Kanthasamy A, Kanthasamy AG. MitoPark transgenic mouse model recapitulates the gastrointestinal dysfunction and gut-microbiome changes of Parkinson's disease. Neurotoxicology 2019; 75:186-199. [PMID: 31505196 DOI: 10.1016/j.neuro.2019.09.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/28/2019] [Accepted: 09/03/2019] [Indexed: 02/06/2023]
Abstract
Gastrointestinal (GI) disturbances are one of the earliest symptoms affecting most patients with Parkinson's disease (PD). In many cases, these symptoms are observed years before motor impairments become apparent. Hence, the molecular and cellular underpinnings that contribute to this early GI dysfunction in PD have actively been explored using a relevant animal model. The MitoPark model is a chronic, progressive mouse model recapitulating several key pathophysiological aspects of PD. However, GI dysfunction and gut microbiome changes have not been categorized in this model. Herein, we show that decreased GI motility was one of the first non-motor symptoms to develop, evident as early as 8 weeks with significantly different transit times from 12 weeks onwards. These symptoms were observed well before motor symptoms developed, thereby paralleling PD progression in humans. At age 24 weeks, we observed increased colon transit time and reduced fecal water content, indicative of constipation. Intestinal inflammation was evidenced with increased expression of iNOS and TNFα in the small and large intestine. Specifically, iNOS was observed mainly in the enteric plexi, indicating enteric glial cell activation. A pronounced loss of tyrosine hydroxylase-positive neurons occurred at 24 weeks both in the mid-brain region as well as the gut, leading to a corresponding decrease in dopamine (DA) production. We also observed decreased DARPP-32 expression in the colon, validating the loss of DAergic neurons in the gut. However, the total number of enteric neurons did not significantly differ between the two groups. Metabolomic gas chromatography-mass spectrometry analysis of fecal samples showed increased sterol, glycerol, and tocopherol production in MitoPark mice compared to age-matched littermate controls at 20 weeks of age while 16 s microbiome sequencing showed a transient temporal increase in the genus Prevotella. Altogether, the data shed more light on the role of the gut dopaminergic system in maintaining intestinal health. Importantly, this model recapitulates the chronology and development of GI dysfunction along with other non-motor symptoms and can become an attractive translational animal model for pre-clinical assessment of the efficacy of new anti-Parkinsonian drugs that can alleviate GI dysfunction in PD.
Collapse
Affiliation(s)
- Shivani Ghaisas
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University (ISU), Ames, IA, 50011, USA
| | - Monica R Langley
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University (ISU), Ames, IA, 50011, USA
| | - Bharathi N Palanisamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University (ISU), Ames, IA, 50011, USA
| | - Somak Dutta
- Department of Statistics, Iowa State University, Ames, IA, USA
| | - Kirthi Narayanaswamy
- W M Keck Metabolomics Research Laboratory, Office of Biotechnology, ISU, Ames, IA, USA
| | - Paul J Plummer
- Veterinary Diagnostic and Production Animal Medicine, College of Veterinary Medicine, ISU, Ames, IA, USA; Veterinary Microbiology and Preventative Medicine, College of Veterinary Medicine, ISU, Ames, IA, USA
| | - Souvarish Sarkar
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University (ISU), Ames, IA, 50011, USA
| | - Muhammet Ay
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University (ISU), Ames, IA, 50011, USA
| | - Huajun Jin
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University (ISU), Ames, IA, 50011, USA
| | - Vellareddy Anantharam
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University (ISU), Ames, IA, 50011, USA
| | - Arthi Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University (ISU), Ames, IA, 50011, USA
| | - Anumantha G Kanthasamy
- Parkinson Disorders Research Program, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University (ISU), Ames, IA, 50011, USA.
| |
Collapse
|
26
|
Li C, Cui L, Yang Y, Miao J, Zhao X, Zhang J, Cui G, Zhang Y. Gut Microbiota Differs Between Parkinson's Disease Patients and Healthy Controls in Northeast China. Front Mol Neurosci 2019; 12:171. [PMID: 31354427 PMCID: PMC6637281 DOI: 10.3389/fnmol.2019.00171] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/20/2019] [Indexed: 01/20/2023] Open
Abstract
Background: There is accumulating evidence suggesting a connection between the gut and Parkinson's disease (PD). Gut microbiota may play an important role in the intestinal lesions in PD patients. Objective: This study aims to determine whether gut microbiota differs between PD patients and healthy controls in Northeast of China, and to identify the factors that influence the changes in the gut microbiota. Methods: We enrolled 51 PD patients and 48 healthy controls in this study. Microbial species in stool samples were determined through 16S-rRNA gene sequencing. Dietary intakes were collected from a subset of 42 patients and 23 controls using a food frequency questionnaire (FFQ). Gut microbiota species richness, diversity, differential abundance of individual taxa between PD patients and controls, and the relationship between the gut microbiota abundance and the dietary and clinical factors were analyzed. Results: PD patients showed decreased species richness, phylogenetic diversity, β- diversity, and altered relative abundance in several taxa compared to the controls. PD- associated clinical scores appeared to be the most influential factors that correlated with the abundance of a variety of taxa. The most consistent findings suggested by multiple analyses used in this study were the increase of Akkermansia and the decrease of Lactobacillus in PD patients in Northeast China. Conclusion: Gut microbiota significantly differed between a group of PD patients and healthy controls in Northeast China, with decreased species richness, phylogenetic diversity, β-diversity, and altered relative abundance in several taxa compared to the controls.
Collapse
Affiliation(s)
- Chunxiao Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Li Cui
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yimin Yang
- Department of Intensive Care Unit, First Hospital of Jilin University, Changchun, China
| | - Jing Miao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Xiuzhen Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jingdian Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Guohong Cui
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, NC, United States
| | - Ying Zhang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
27
|
Tucker HR, Mahoney E, Chhetri A, Unger K, Mamone G, Kim G, Audil A, Moolick B, Molho ES, Pilitsis JG, Shin DS. Deep brain stimulation of the ventroanterior and ventrolateral thalamus improves motor function in a rat model of Parkinson's disease. Exp Neurol 2019; 317:155-167. [PMID: 30890329 DOI: 10.1016/j.expneurol.2019.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/26/2019] [Accepted: 03/14/2019] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease with affected individuals exhibiting motor symptoms of bradykinesia, muscle rigidity, tremor, postural instability and gait dysfunction. The current gold standard treatment is pharmacotherapy with levodopa, but long-term use is associated with motor response fluctuations and can cause abnormal movements called dyskinesias. An alternative treatment option is deep brain stimulation (DBS) with the two FDA-approved brain targets for PD situated in the basal ganglia; specifically, in the subthalamic nucleus (STN) and globus pallidus pars interna (GPi). Both improve quality of life and motor scores by ~50-70% in well-selected patients but can also elicit adverse effects on cognition and other non-motor symptoms. Therefore, identifying a novel DBS target that is efficacious for patients not optimally responsive to current DBS targets with fewer side-effects has clear clinical merit. Here, we investigate whether the ventroanterior (VA) and ventrolateral (VL) motor nuclei of the thalamus can serve as novel and effective DBS targets for PD. In the limb-use asymmetry test (LAT), hemiparkinsonian rats showcased left forelimb akinesia and touched only 6.5 ± 1.3% with that paw. However, these animals touched equally with both forepaws with DBS at 10 Hz, 100 μsec pulse width and 100 uA cathodic stimulation in the VA (n = 7), VL (n = 8) or at the interface between the two thalamic nuclei which we refer to as the VA|VL (n = 12). With whole-cell patch-clamp recordings, we noted that VA|VL stimulation in vitro increased the number of induced action potentials in proximal neurons in both areas albeit VL neurons transitioned from bursting to non-bursting action potentials (APs) with large excitatory postsynaptic potentials time-locked to stimulation. In contrast, VA neurons were excited with VA|VL electrical stimulation but with little change in spiking phenotype. Overall, our findings show that DBS in the VA, VL or VA|VL improved motor function in a rat model of PD; plausibly via increased excitation of residing neurons.
Collapse
Affiliation(s)
- Heidi R Tucker
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, United States of America
| | - Emily Mahoney
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, United States of America
| | - Ashok Chhetri
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, United States of America
| | - Kristen Unger
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, United States of America
| | - Gianna Mamone
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, United States of America
| | - Gabrielle Kim
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, United States of America
| | - Aliyah Audil
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, United States of America
| | - Benjamin Moolick
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, United States of America
| | - Eric S Molho
- Department of Neurology, Albany Medical Center, Albany, NY, United States of America
| | - Julie G Pilitsis
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, United States of America; Department of Neurosurgery, Albany Medical Center, Albany, NY, United States of America
| | - Damian S Shin
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, Albany, NY, United States of America; Department of Neurology, Albany Medical Center, Albany, NY, United States of America.
| |
Collapse
|
28
|
Micó-Amigo ME, Kingma I, Heinzel S, Rispens SM, Heger T, Nussbaum S, van Lummel RC, Berg D, Maetzler W, van Dieën JH. Potential Markers of Progression in Idiopathic Parkinson's Disease Derived From Assessment of Circular Gait With a Single Body-Fixed-Sensor: A 5 Year Longitudinal Study. Front Hum Neurosci 2019; 13:59. [PMID: 30837857 PMCID: PMC6389786 DOI: 10.3389/fnhum.2019.00059] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 02/04/2019] [Indexed: 12/03/2022] Open
Abstract
Background and Aim: Development of objective, reliable and easy-to-use methods to obtain progression markers of Parkinson's disease (PD) is required to evaluate interventions and to advance research in PD. This study aimed to provide quantitative markers of progression in idiopathic PD from the assessment of circular gait (walking in circles) with a single body-fixed inertial sensor placed on the lower back. Methods: The assessments were performed every 6 months over a (up to) 5 years period for 22 patients in early-stage PD, 27 patients in middle-stage PD and 25 healthy controls (HC). Longitudinal changes of 24 gait features extracted from accelerometry were compared between PD groups and HCs with generalized estimating equations (GEE) analysis, accounting for gait speed, age and levodopa medication state confounders when required. Results: Five gait features indicated progressive worsening in early stages of PD: number of steps, total duration and harmonic ratios calculated from vertical (VT), medio-lateral (ML), and anterior-posterior (AP) accelerations. For middle stages of PD, three gait features were identified as potential progression markers: stride time variability, and stride regularity from VT and AP acceleration. Conclusion: Faster progressive worsening of gait features in early and middle stages of PD relative to healthy controls over 5 years confirmed the potential of accelerometry-based assessments as quantitative progression markers in early and middle stages of the disease. The difference in significant parameters between both PD groups suggests that distinct domains of gait deteriorate in these PD stages. We conclude that instrumented circular walking assessment is a practical and useful tool in the assessment of PD progression that may have relevant potential to be implemented in clinical trials and even clinical routine, particularly in a developing digital era.
Collapse
Affiliation(s)
- M. Encarna Micó-Amigo
- Department of Human Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, Netherlands
| | - Idsart Kingma
- Department of Human Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, Netherlands
| | - Sebastian Heinzel
- Department of Neurology, Christian-Albrechts-University, Kiel, Germany
| | - Sietse M. Rispens
- Department of Human Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, Netherlands
- Personal Health Department, Philips Research Europe, Eindhoven, Netherlands
| | - Tanja Heger
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- DZNE, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Susanne Nussbaum
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- DZNE, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | | | - Daniela Berg
- Department of Neurology, Christian-Albrechts-University, Kiel, Germany
- Personal Health Department, Philips Research Europe, Eindhoven, Netherlands
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Walter Maetzler
- Department of Neurology, Christian-Albrechts-University, Kiel, Germany
- Personal Health Department, Philips Research Europe, Eindhoven, Netherlands
- Department of Neurodegeneration, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Jaap H. van Dieën
- Department of Human Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, Netherlands
| |
Collapse
|
29
|
Berrío Sánchez J, Cucarian Hurtado J, Barcos Nunes R, de Oliveira AA. Mesenchymal stem cell transplantation and aerobic exercise for Parkinson's disease: therapeutic assets beyond the motor domain. Rev Neurosci 2019; 30:165-178. [PMID: 29959887 DOI: 10.1515/revneuro-2018-0011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/19/2018] [Indexed: 11/15/2022]
Abstract
Parkinson's disease (PD) is a very common neurodegenerative condition in which both motor and nonmotor deficits evolve throughout the course of the disease. Normally characterized as a movement disorder, PD has been broadly studied from a motor perspective. However, mild to moderate cognitive deficits began to appear in the early phases of the disease, even before motor disturbances actually manifest, and continue to progress relentlessly. These nonmotor manifestations are also a source of detriment to the patients' already strained functionality and quality of life, and pose a therapeutic challenge seeing that replacing therapies have had conflicting results. Considering that the currently approved therapies can hardly be considered curative, efforts to find therapeutic approaches with an actual disease-modifying quality and capable of addressing not only motor but also cognitive dysfunctions are clearly needed. Among possible alternatives with such attribute, mesenchymal stem cell transplantation and exercise are worth highlighting given their common neuroprotective, neuroplastic, and immunomodulatory properties. In this paper, we will summarize the existent literature on the topic, focusing on the mechanisms of action through which these two approaches might beget therapeutic benefits for PD beyond the commonly assessed motor dysfunctions, alluding, at the same time, toward a potential synergic association of both therapies as an optimized approach for PD.
Collapse
Affiliation(s)
- Jenny Berrío Sánchez
- Graduate Program in Rehabilitation Science, Department of Psychology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Rio Grande do Sul, CEP 90050-170, Brazil
| | - Jaison Cucarian Hurtado
- Graduate Program in Rehabilitation Science, Department of Psychology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Rio Grande do Sul, CEP 90050-170, Brazil
| | - Ramiro Barcos Nunes
- Research Department, Instituto Federal de Educação, Ciência e Tecnologia. SUL-RIO-GRANDENSE, Rua Men de Sá, 800, Bom Sucesso, Gravataí, CEP 94.135-300, Brazil
| | - Alcyr Alves de Oliveira
- Graduate Program in Psychology and Health, Department of Psychology, Federal University of Health Sciences of Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Rio Grande do Sul, CEP 90050-170, Brazil
| |
Collapse
|
30
|
Bermúdez ML, Skelton MR, Genter MB. Intranasal carnosine attenuates transcriptomic alterations and improves mitochondrial function in the Thy1-aSyn mouse model of Parkinson's disease. Mol Genet Metab 2018; 125:305-313. [PMID: 30146452 DOI: 10.1016/j.ymgme.2018.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 08/07/2018] [Indexed: 12/17/2022]
Abstract
Mitochondrial dysfunction plays a central role in the pathogenesis of neurodegenerative diseases such as Parkinson's disease (PD). This study was designed to determine whether the dipeptide carnosine, which has been shown to protect against oxidative stress and mitochondrial dysfunction, would provide a beneficial effect on mitochondrial function in the Thy1-aSyn mouse model of PD. Thy1-aSyn mice, which overexpress wild-type human alpha-synuclein (aSyn), exhibit progressive non-motor and motor deficits as early as 2 months of age. Two-month old Thy1-aSyn mice and wild-type littermates were randomly assigned to treatment groups with intranasal (IN) and drinking water carnosine, with controls receiving 10 μl of sterile waster intranasally or carnosine-free drinking water, respectively. After two months of treatment, mice were euthanized, and the midbrain was dissected for the evaluation of the gene expression and mitochondrial function. Transcriptional deficiencies associated with the aSyn overexpression in Thy1-aSyn mice were related to ribosomal and mitochondrial function. These deficiencies were attenuated by IN carnosine administration, which increased the expression of mitochondrial genes and enhanced mitochondrial function. These results suggest a potential neuroprotective role for IN-carnosine in PD patients.
Collapse
Affiliation(s)
- Mei-Ling Bermúdez
- Department of Environmental Health, University of Cincinnati, ML 670056, Cincinnati, OH 45267-0056, United States of America.
| | - Matthew R Skelton
- Department of Pediatrics, UC COM, Division of Neurology, Cincinnati Children's Research Foundation, 3333 Burnet Avenue, MLC 7044, Cincinnati, OH 45229-3039, United States of America
| | - Mary Beth Genter
- Department of Environmental Health, University of Cincinnati, ML 670056, Cincinnati, OH 45267-0056, United States of America.
| |
Collapse
|
31
|
Maraki MI, Yannakoulia M, Stamelou M, Stefanis L, Xiromerisiou G, Kosmidis MH, Dardiotis E, Hadjigeorgiou GM, Sakka P, Anastasiou CA, Simopoulou E, Scarmeas N. Mediterranean diet adherence is related to reduced probability of prodromal Parkinson's disease. Mov Disord 2018; 34:48-57. [PMID: 30306634 DOI: 10.1002/mds.27489] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/31/2018] [Accepted: 08/09/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The International Parkinson and Movement Disorder Society recently introduced a methodology for probability score calculation for prodromal PD. OBJECTIVES To assess the probability of prodromal PD in an older population and investigate its possible association with Mediterranean diet adherence. METHODS Data from a population-based cohort study of older adults (HEllenic Longitudinal Investigation of Aging and Diet) in Greece were used. Probability of prodromal PD was calculated according to International Parkinson and Movement Disorder Society research criteria. A detailed food frequency questionnaire was used to evaluate dietary intake and calculate Mediterranean diet adherence score, ranging from 0 to 55, with higher scores indicating higher adherence. RESULTS Median probability of prodromal PD was 1.9%, ranging from 0.2 to 96.7% in 1,731 PD-free individuals aged ≥ 65 (41% male). Lower probability for prodromal PD (P < 0.001) in the higher Mediterranean diet adherence groups was noted, driven mostly by nonmotor markers of prodromal PD, depression, constipation, urinary dysfunction, and daytime somnolence. Each unit increase in Mediterranean diet score was associated with a 2% decreased probability for prodromal PD (P < 0.001). Compared to participants in the lowest quartile of Mediterranean diet adherence, those in the highest quartile were associated with a ∼21% lower probability for prodromal PD. CONCLUSIONS Adherence to the Mediterranean diet is associated with lower probability of prodromal PD in older people. Further studies are needed to elucidate the potential causality of this association, potential relation of the Mediterranean diet to delayed onset or lower incidence of PD, as well as the underlying neurobiological mechanisms. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Maria I Maraki
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Mary Yannakoulia
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Maria Stamelou
- Department of Neurology, Philipps University, Marburg, Germany.,1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Greece
| | - Leonidas Stefanis
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Greece.,Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Mary H Kosmidis
- Lab of Cognitive Neuroscience, School of Psychology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | | | - Paraskevi Sakka
- Athens Association of Alzheimer's Disease and Related Disorders, Marousi, Greece
| | - Costas A Anastasiou
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece.,1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Greece
| | | | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Greece.,Taub Institute for Research in Alzheimer's Disease and the Aging Brain, The Gertrude H. Sergievsky Center, Department of Neurology, Columbia University, New York, USA
| |
Collapse
|
32
|
Evaluation of sleep quality in individuals with Parkinson’s disease using objective and subjective measures. Sleep Biol Rhythms 2018. [DOI: 10.1007/s41105-018-0185-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
33
|
Abstract
Parkinson's disease (PD) is a debilitating neurodegenerative condition associated with tremor, rigidity, dementia, and gastrointestinal symptoms such as constipation, nausea and vomiting. The pathological hallmarks of PD are Lewy bodies and neurites in the brain and peripheral nerves. The major constituent of Lewy bodies is the neuronal protein α-synuclein. Misfolding of α-synuclein confers prion-like properties enabling its spread from cell to cell. Misfolded α-synuclein also serves as a template and induces misfolding of endogenous α-synuclein in recipient cells leading to the formation of oligomers that progress to fibrils and eventually Lewy bodies. Accumulating evidence suggests that PD may arise in the gut. Clinically, gastrointestinal symptoms often appear in patients before other neurological signs and aggregates of α-synuclein have been found in enteric nerves of PD patients. Importantly, patients undergoing vagotomy have a reduced risk of developing PD. Experimentally, abnormal forms of α-synuclein appear in enteric nerves before they appear in the brain and injection of abnormal α-synuclein into the wall of the intestine spreads to the vagus nerve. Ingested toxins and alterations in gut microbiota can induce α-synuclein aggregation and PD, however, it is not known how PD starts. Recently, it has been shown that sensory cells of the gut known as enteroendocrine cells (EECs) contain α-synuclein and synapse with enteric nerves, thus providing a connection from the gut to the brain. It is possible that abnormal α-synuclein first develops in EECs and spreads to the nervous system.
Collapse
Affiliation(s)
- Rodger A Liddle
- Department of Medicine, Duke University Medical Center and Department of Veterans Affairs Health Care System, Durham, NC 27710, United States.
| |
Collapse
|
34
|
Getz SJ, Levin B. Cognitive and Neuropsychiatric Features of Early Parkinson's Disease. Arch Clin Neuropsychol 2018; 32:769-785. [PMID: 29077803 DOI: 10.1093/arclin/acx091] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/05/2017] [Indexed: 12/31/2022] Open
Abstract
The clinical definition of Parkinson's disease (PD) is based on cardinal motor features including bradykinesia as well as an additional symptom of tremor, postural instability, or rigidity. Evidence from neuropathological, imaging, and clinical research suggests a premotor, early phase of PD pathology. Further understanding of the earliest biomarkers of PD is crucial for the development of neuroprotective, disease modifying, cognitive, and psychiatric interventions. Recent research has explored early non-motor markers of PD pathology. This issue is especially timely as the International Parkinson and Movement Disorder Society has recently provided a research definition for prodromal PD which includes combinations of prodromal markers and risk factors aimed at identifying target populations for disease-prevention trials. In this review of early PD, we will outline early non-motor symptoms, early cognitive and neuropsychiatric features, neuropsychological assessment strategies, emerging evidence for early biomarkers, and treatment recommendations.
Collapse
Affiliation(s)
- Sarah J Getz
- Department of Neurology, Division of Neuropsychology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Bonnie Levin
- Department of Neurology, Division of Neuropsychology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
35
|
Abstract
Parkinson's disease (PD), diffuse Lewy body disease (DLBD), and multiple system atrophy (MSA) constitute the three major neurodegenerative disorders referred to as synucleinopathies because both genetic and pathological results implicate the α-synuclein protein in their pathogenesis. PD and DLBD are recognized as closely related diseases with substantial clinical and pathological overlap. MSA, on the other hand, has a distinctive clinical presentation and neuropathological profile. In this review, we will summarize the evidence linking α-synuclein to these three disorders. Hundreds of patients with point or copy number mutations in the gene encoding α-synuclein, SNCA, have been reported in the literature in association with hereditary, autosomal dominant forms of PD, DLBD, or neurodegenerative disease with parkinsonism. The copy number mutations show a dosage effect with age at onset and severity correlating with the number of extra copies of SNCA a patient carries. Common variation in and around the SNCA gene has also been found by genome-wide association studies to be associated with increased risk for apparently sporadic PD, with some evidence that these variants exert their effect through modest increases in α-synuclein expression. Complementing the genetic evidence linking α-synuclein to PD and DLBD is the pathological finding that α-synuclein is a major constituent of Lewy bodies and Lewy neurites in the brains of patients with the common sporadic form of PD. On the other hand, there is little genetic evidence linking SNCA to MSA despite strong neuropathological evidence of α-synuclein aggregation in oligodendroglial cells in MSA patients. Evidence is now emerging that α-synuclein aggregates can have different protein conformations, referred to as strains, similar to what has been shown in prion disease. The different phenotypes in hereditary PD/DLBD versus MSA are likely, therefore, to be the result not only of how specific mutations affect protein expression and turnover, but also a more complex interaction between intrinsic and extrinsic factors governing aggregation and strain formation.
Collapse
Affiliation(s)
- Robert L Nussbaum
- Volunteer Clinical Faculty, UCSF School of Medicine, University of California, San Francisco, San Francisco, California 94143
| |
Collapse
|
36
|
Chen H. Author response: Olfaction and incident Parkinson disease in US white and black older adults. Neurology 2018; 90:941. [DOI: 10.1212/wnl.0000000000005515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
37
|
Nussbaum RL. The Identification of Alpha-Synuclein as the First Parkinson Disease Gene. JOURNAL OF PARKINSONS DISEASE 2017; 7:S43-S49. [PMID: 28282812 PMCID: PMC5345646 DOI: 10.3233/jpd-179003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In this Commentary, I describe the events that led from an NINDS-sponsored Workshop on Parkinson Disease Research in 1995, where I was asked to speak about the genetics of Parkinson disease, to the identification a mere two years later of a mutation in alpha-synuclein as the cause of autosomal dominant Parkinson disease in the Contursi kindred. I review the steps we took to first map and then find the mutation in the alpha-synuclein locus and describe the obstacles and the role of serendipity in facilitating the work. Although alpha-synuclein mutations are a rare cause of hereditary PD, the importance of this finding goes far beyond the rare families with hereditary disease because it pinpointed alpha-synuclein as a key contributor to the far more common sporadic form of Parkinson disease. This work confirms William Harvey's observation from 350 years ago that studying rarer forms of a disease is an excellent way to understand the more common forms of that disease. The identification of synuclein's role in hereditary Parkinson disease has opened new avenues of research into the pathogenesis and potential treatments of the common form of Parkinson disease that affects many millions of Americans and tens of millions of human beings worldwide.
Collapse
Affiliation(s)
- Robert L. Nussbaum
- Correspondence to: Robert L. Nussbaum, MD, Chief Medical Officer, Invitae Corporation and former Holly Smith Professor, University of California San Francisco, CA, USA. Tel.: +1 415 264 2589; Fax: +1 415 230 4991; E-mail:
| |
Collapse
|
38
|
Rodríguez-Violante M, de Saráchaga AJ, Cervantes-Arriaga A, Davila-Avila NM, Carreón-Bautista E, Estrada-Bellmann I, Parra-López G, Cruz-Fino D, Pascasio-Astudillo F. Premotor symptoms and the risk of Parkinson's disease: A case-control study in Mexican population. Clin Neurol Neurosurg 2017. [PMID: 28644969 DOI: 10.1016/j.clineuro.2017.06.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To assess the prevalence of pre-motor symptoms and estimate the risk for developing Parkinson's disease in Mexican population. PATIENTS AND METHODS A case-control study was carried out with consecutive subjects with Parkinson's disease from two different referral centers in Mexico. Gender- and age-matched controls were randomly selected from the participating hospitals. All subjects were assessed using a structured questionnaire for the assessment of pre-motor symptoms (hyposmia, depression, anxiety, constipation, and sleep disorders). Odds ratios (OR) were calculated using logistic regression analysis. RESULTS A total of 430 subjects with PD and 430 healthy subjects were included. Premotor symptoms prevalence was 77.7% (n=334) for the PD group, compared to 41.3% (n=178) in the control group (p<0.001). After logistic multivariate analysis, previous history of hyposmia (OR 2.02 [95% CI 1.33-3.06]), depression (OR 2.52 [95% CI 1.67-3.84]), anxiety (OR 4.37 [95% CI 2.73-6.98]) and sleep disorders (OR 2.03 [95% CI 1.41-2.93]) were independently associated with Parkinson's disease. Overall prediction success of the model was 81.2% for controls and 61.2% for subjects with PD. CONCLUSION All five premotor symptoms assessed were more commonly reported in PD subjects than healthy controls. The presence of non-motor symptoms yield a prediction success of 71.2% to discriminate between PD subjects and healthy controls.
Collapse
Affiliation(s)
- Mayela Rodríguez-Violante
- Clinical Neurodegenerative Research Unit, National Institute of Neurology and Neurosurgery, Mexico City, Mexico; Movement Disorder Clinic, National Institute of Neurology and Neurosurgery, Mexico City, Mexico.
| | - Adib Jorge de Saráchaga
- Clinical Neurodegenerative Research Unit, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - Amin Cervantes-Arriaga
- Clinical Neurodegenerative Research Unit, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - Ned Merari Davila-Avila
- Clinical Neurodegenerative Research Unit, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - Edith Carreón-Bautista
- Clinical Neurodegenerative Research Unit, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | | | - Guillermo Parra-López
- Clinical Neurodegenerative Research Unit, National Institute of Neurology and Neurosurgery, Mexico City, Mexico; Movement Disorder Clinic, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - Diego Cruz-Fino
- Clinical Neurodegenerative Research Unit, National Institute of Neurology and Neurosurgery, Mexico City, Mexico; Movement Disorder Clinic, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - Francisco Pascasio-Astudillo
- Clinical Neurodegenerative Research Unit, National Institute of Neurology and Neurosurgery, Mexico City, Mexico; Movement Disorder Clinic, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| |
Collapse
|
39
|
Chandra R, Hiniker A, Kuo YM, Nussbaum RL, Liddle RA. α-Synuclein in gut endocrine cells and its implications for Parkinson's disease. JCI Insight 2017; 2:92295. [PMID: 28614796 DOI: 10.1172/jci.insight.92295] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 05/05/2017] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with devastating clinical manifestations. In PD, neuronal death is associated with intracellular aggregates of the neuronal protein α-synuclein known as Lewy bodies. Although the cause of sporadic PD is not well understood, abundant clinical and pathological evidence show that misfolded α-synuclein is found in enteric nerves before it appears in the brain. This suggests a model in which PD pathology originates in the gut and spreads to the central nervous system via cell-to-cell prion-like propagation, such that transfer of misfolded α-synuclein initiates misfolding of native α-synuclein in recipient cells. We recently discovered that enteroendocrine cells (EECs), which are part of the gut epithelium and directly face the gut lumen, also possess many neuron-like properties and connect to enteric nerves. In this report, we demonstrate that α-synuclein is expressed in the EEC line, STC-1, and native EECs of mouse and human intestine. Furthermore, α-synuclein-containing EECs directly connect to α-synuclein-containing nerves, forming a neural circuit between the gut and the nervous system in which toxins or other environmental influences in the gut lumen could affect α-synuclein folding in the EECs, thereby beginning a process by which misfolded α-synuclein could propagate from the gut epithelium to the brain.
Collapse
Affiliation(s)
- Rashmi Chandra
- Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, USA
| | | | - Yien-Ming Kuo
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Robert L Nussbaum
- Department of Medicine, UCSF, San Francisco, California, USA.,Invitae Corporation, San Francisco, California, USA
| | - Rodger A Liddle
- Department of Medicine, Duke University and Durham VA Medical Centers, Durham, North Carolina, USA.,Duke Institute for Brain Sciences, Durham, North Carolina, USA
| |
Collapse
|
40
|
Belghali M, Chastan N, Cignetti F, Davenne D, Decker LM. Loss of gait control assessed by cognitive-motor dual-tasks: pros and cons in detecting people at risk of developing Alzheimer's and Parkinson's diseases. GeroScience 2017; 39:305-329. [PMID: 28551877 DOI: 10.1007/s11357-017-9977-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 05/02/2017] [Indexed: 12/26/2022] Open
Abstract
Alzheimer's and Parkinson's diseases are age-related progressive neurodegenerative diseases of increasing prevalence worldwide. In the absence of curative therapy, current research is interested in prevention, by identifying subtle signs of early-stage neurodegeneration. Today, the field of behavioral neuroscience has emerged as one of the most promising areas of research on this topic. Recently, it has been shown that the exacerbation of gait disorders under dual-task conditions (i.e., simultaneous performance of cognitive and motor tasks) could be a characteristic feature of Alzheimer's and Parkinson's diseases. The cognitive-motor dual-task paradigm during walking allows to assess whether (i) executive attention is abnormally impaired in prodromal Alzheimer's disease or (ii) compensation strategies are used in order to preserve gait function when the basal ganglia system is altered in prodromal Parkinson's disease. This review aims at (i) identifying patterns of dual-task-related gait changes that are specific to Alzheimer's and Parkinson's diseases, respectively, (ii) demonstrating that these changes could potentially be used as prediagnostic markers for disease onset, (iii) reviewing pros and cons of existing dual-task studies, and (iv) proposing future directions for clinical research.
Collapse
Affiliation(s)
- Maroua Belghali
- Normandie Univ, UNICAEN, INSERM, COMETE, 14000, Caen, France
| | - Nathalie Chastan
- Normandie Univ, UNICAEN, INSERM, COMETE, 14000, Caen, France.,Department of Neurophysiology, UNIROUEN, Rouen University Hospital-Charles Nicolle, Normandie Univ, 76000, Rouen, France
| | - Fabien Cignetti
- CNRS, LNC UMR 7291, Aix-Marseille Univ, 13331, Marseille, France
| | - Damien Davenne
- Normandie Univ, UNICAEN, INSERM, COMETE, 14000, Caen, France
| | - Leslie M Decker
- Normandie Univ, UNICAEN, INSERM, COMETE, 14000, Caen, France.
| |
Collapse
|
41
|
MPTP Mouse Model of Preclinical and Clinical Parkinson’s Disease as an Instrument for Translational Medicine. Mol Neurobiol 2017; 55:2991-3006. [DOI: 10.1007/s12035-017-0559-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 04/12/2017] [Indexed: 02/04/2023]
|
42
|
Moschos MM, Chatziralli IP. Evaluation of Choroidal and Retinal Thickness Changes in Parkinson’s Disease Using Spectral Domain Optical Coherence Tomography. Semin Ophthalmol 2017; 33:494-497. [DOI: 10.1080/08820538.2017.1307423] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Marilita M. Moschos
- Electrophysiology Laboratory, 1st Department of Ophthalmology, University of Athens, Athens, Greece
| | - Irini P. Chatziralli
- Electrophysiology Laboratory, 1st Department of Ophthalmology, University of Athens, Athens, Greece
| |
Collapse
|
43
|
Agricultural activities and the incidence of Parkinson's disease in the general French population. Eur J Epidemiol 2017; 32:203-216. [PMID: 28185034 DOI: 10.1007/s10654-017-0229-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/27/2017] [Indexed: 12/29/2022]
Abstract
Most studies on pesticides and Parkinson's disease (PD) focused on occupational exposure in farmers. Whether non-occupational exposure is associated with PD has been little explored. We investigated the association between agricultural characteristics and PD incidence in a French nationwide ecologic study. We hypothesized that persons living in regions with agricultural activities involving more intensive pesticide use would be at higher risk. We identified incident PD cases from French National Health Insurance databases (2010-2012). The proportion of land dedicated to 18 types of agricultural activities was defined at the canton of residence level. We examined the association between agricultural activities and PD age/sex-standardized incidence ratios using multivariable multilevel Poisson regression adjusted for smoking, deprivation index, density of neurologists, and rurality (proportion of agricultural land); we used a false discovery rate approach to correct for multiple comparisons and compute q-values. We also compared incidence in clusters of cantons with similar agricultural characteristics (k-means algorithm). We identified 69,010 incident PD cases. Rurality was associated with higher PD incidence (p < 0.001). Cantons with higher density of vineyards displayed the strongest association (RRtop/bottom quartile = 1.102, 95% CI = 1.049-1.158; q-trend = 0.040). This association was similar in men, women, and non-farmers, stronger in older than younger persons, and present in all French regions. Persons living in the cluster with greatest vineyards density had 8.5% (4.4-12.6%) higher PD incidence (p < 0.001). In France, vineyards rank among the crops that require most intense pesticide use. Regions with greater presence of vineyards are characterized by higher PD risk; non-professional pesticides exposure is a possible explanation.
Collapse
|
44
|
Passali GC, Bove F, Vargiu L, Bentivoglio AR, Anzivino R, De Corso E, Galli J, Rigante M, Pandolfini M, Sergi B, Giuliani M, Cianfrone F, Paludetti G, Fasano A. New olfactometric findings in Parkinson's disease. Clin Otolaryngol 2017; 42:837-843. [PMID: 28004533 DOI: 10.1111/coa.12816] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2016] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To investigate in Parkinson's disease-affected patients a correlation between hyposmia and gastrointestinal dysfunction and their possible identical etiopathogenesis. DESIGN Retrospective cohort study. SETTING ENT and neurology departments (Gemelli Hospital, Rome, Italy). PARTICIPANTS A total of 78 patients with diagnosis of PD according to the UK Brain Bank criteria. INCLUSION CRITERIA informed consent and olfactory testing executed; exclusion criteria: signs of dementia according to the DSM-IV criteria; Mini Mental State Examination score ≤26; head trauma; central neurological disorders, nasal or systemic diseases potentially affecting olfactory function. Motor condition was assessed by means of Hoehn and Yahr staging and by section III of the Unified PD Rating Scale, performed off and on medications. MAIN OUTCOME MEASURES The patients underwent olfactory evaluation (TDI score), after rhinomanometry with nasal decongestion. A total of 25 non-motor symptoms were evaluated through an interview. RESULTS Olfactory dysfunction was objectively found in 91.0% of patients, a percentage higher than the subjective hyposmia reported (55.1%) P = 0.0001. Seven patients (9.0%) were normosmic, 49 (62.8%) hyposmic and 22 (28.2%) anosmic. Subjective hyposmia, constipation, bloating and dyspepsia differed across groups, being higher in anosmic and hyposmic ones than in the normosmic group. P value was ≤0.05 for each symptom. Despite the original results, this study has the limitation of being based on subjective ratings by a relatively limited group of patients. CONCLUSIONS Hyposmia and gastrointestinal symptoms are correlated, and this would support a possible common origin; the CNS could be reached through two different pathways, both starting in the peripheral nervous system.
Collapse
Affiliation(s)
- G C Passali
- Institute of Otorhinolaryngology, Head and Neck Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - F Bove
- Department of Neurology, Catholic University of Sacred Heart, Rome, Italy
| | - L Vargiu
- Institute of Otorhinolaryngology, Head and Neck Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - A R Bentivoglio
- Department of Neurology, Catholic University of Sacred Heart, Rome, Italy
| | - R Anzivino
- Institute of Otorhinolaryngology, Head and Neck Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - E De Corso
- Institute of Otorhinolaryngology, Head and Neck Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - J Galli
- Institute of Otorhinolaryngology, Head and Neck Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - M Rigante
- Institute of Otorhinolaryngology, Head and Neck Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - M Pandolfini
- Institute of Otorhinolaryngology, Head and Neck Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - B Sergi
- Institute of Otorhinolaryngology, Head and Neck Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - M Giuliani
- Institute of Otorhinolaryngology, Head and Neck Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - F Cianfrone
- Institute of Otorhinolaryngology, Head and Neck Surgery, S. Camillo Hospital, Rome, Italy
| | - G Paludetti
- Institute of Otorhinolaryngology, Head and Neck Surgery, Catholic University of Sacred Heart, Rome, Italy
| | - A Fasano
- Movement Disorders Centre - Toronto Western Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
45
|
Del Tredici K, Braak H. Review: Sporadic Parkinson's disease: development and distribution of α-synuclein pathology. Neuropathol Appl Neurobiol 2016; 42:33-50. [PMID: 26662475 DOI: 10.1111/nan.12298] [Citation(s) in RCA: 286] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 12/04/2015] [Accepted: 12/13/2015] [Indexed: 12/17/2022]
Abstract
The development of α-synuclein immunoreactive aggregates in selectively vulnerable neuronal types of the human central, peripheral, and enteric nervous systems is crucial for the pathogenesis of sporadic Parkinson's disease. The presence of these lesions persists into the end phase of the disease, a process that is not subject to remission. The initial induction of α-synuclein misfolding and subsequent aggregation probably occurs in the olfactory bulb and/or the enteric nervous system. Each of these sites is exposed to potentially hostile environmental factors. Once formed, the aggregates appear to be capable of propagating trans-synaptically from nerve cell to nerve cell in a virtually self-promoting pathological process. A regional distribution pattern of aggregated α-synuclein emerges that entails the involvement of only a few types of susceptible and axonally interconnected projection neurons within the human nervous system. One major route of disease progression may originate in the enteric nervous system and retrogradely reach the dorsal motor nucleus of the vagal nerve in the lower brainstem. From there, the disease process proceeds chiefly in a caudo-rostral direction through visceromotor and somatomotor brainstem centres to the midbrain, forebrain, and cerebral cortex. Spinal cord centres may become involved by means of descending projections from involved lower brainstem nuclei as well as by sympathetic projections connecting the enteric nervous system with postganglionic peripheral ganglia and preganglionic nuclei of the spinal cord. The development of experimental cellular and animal models is helping to explain the mechanisms of how abnormal α-synuclein can undergo aggregation and how transmission along axonal connectivities can occur, thereby encouraging the initiation of potential disease-modifying therapeutic strategies for sporadic Parkinson's disease.
Collapse
Affiliation(s)
- K Del Tredici
- Clinical Neuroanatomy Section, Department of Neurology, Center for Biomedical Research, University of Ulm, Ulm, Germany
| | - H Braak
- Clinical Neuroanatomy Section, Department of Neurology, Center for Biomedical Research, University of Ulm, Ulm, Germany
| |
Collapse
|
46
|
The efficacy of apomorphine – A non-motor perspective. Parkinsonism Relat Disord 2016; 33 Suppl 1:S28-S35. [DOI: 10.1016/j.parkreldis.2016.11.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/25/2016] [Accepted: 11/30/2016] [Indexed: 01/09/2023]
|
47
|
Stirpe P, Hoffman M, Badiali D, Colosimo C. Constipation: an emerging risk factor for Parkinson's disease? Eur J Neurol 2016; 23:1606-1613. [PMID: 27444575 DOI: 10.1111/ene.13082] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 06/09/2016] [Indexed: 12/11/2022]
Abstract
Constipation is the most prominent and disabling manifestation of lower gastrointestinal (GI) dysfunction in Parkinson's disease (PD). The prevalence of constipation in PD patients ranges from 24.6% to 63%; this variability is due to the different criteria used to define constipation and to the type of population enrolled in the studies. In addition, constipation may play an active role in the pathophysiological changes that underlie motor fluctuations in advanced PD through its negative effects on absorption of levodopa. Several clinical studies now consistently suggest that constipation may precede the first occurrence of classical motor features in PD. Studies in vivo, using biopsies of the GI tract and more recently functional imaging investigations, showed the presence of α-synuclein (α-SYN) aggregates and neurotransmitter alterations in enteric tissues. All these findings support the Braak proposed model for the pathophysiology of α-SYN aggregates in PD, with early pathological involvement of the enteric nervous system and dorsal motor nucleus of the vagus. Therefore, constipation could have the potential sensitivity to be used as a clinical biomarker of the prodromal phase of the disease. The use of colonic biopsies to look at α-SYN pathology, once confirmed by larger prospective studies, might eventually represent a feasible, albeit partially invasive, new diagnostic biomarker for PD.
Collapse
Affiliation(s)
- P Stirpe
- Department of Neurology, Institute for Research and Medical Care (IRCCS) San Raffaele, Rome, Italy
| | - M Hoffman
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - D Badiali
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - C Colosimo
- Department of Neurology, Santa Maria University Hospital, Terni, Italy. ,
| |
Collapse
|
48
|
Sveinbjornsdottir S. The clinical symptoms of Parkinson's disease. J Neurochem 2016; 139 Suppl 1:318-324. [PMID: 27401947 DOI: 10.1111/jnc.13691] [Citation(s) in RCA: 737] [Impact Index Per Article: 81.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 05/21/2016] [Accepted: 05/31/2016] [Indexed: 12/11/2022]
Abstract
In this review, the clinical features of Parkinson's disease, both motor and non-motor, are described in the context of the progression of the disease. Also briefly discussed are the major treatment strategies and their complications. Parkinson's disease is a slowly progressing neurodegenerative disorder, causing impaired motor function with slow movements, tremor and gait and balance disturbances. A variety of non-motor symptoms are common in Parkinson's disease. They include disturbed autonomic function with orthostatic hypotension, constipation and urinary disturbances, a variety of sleep disorders and a spectrum of neuropsychiatric symptoms. This article describes the different clinical symptoms that may occur and the clinical course of the disease. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Sigurlaug Sveinbjornsdottir
- Department of Neurology, Broomfield Hospital, Chelmsford, Essex, CM1 7ET, UK. .,Queen Mary School of Medicine and Dentistry, University of London, London, UK.
| |
Collapse
|
49
|
Augustin AD, Charlett A, Weller C, Dobbs SM, Taylor D, Bjarnason I, Dobbs RJ. Quantifying rigidity of Parkinson's disease in relation to laxative treatment: a service evaluation. Br J Clin Pharmacol 2016; 82:441-50. [PMID: 27062674 PMCID: PMC4972160 DOI: 10.1111/bcp.12967] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 03/28/2016] [Accepted: 04/04/2016] [Indexed: 12/13/2022] Open
Abstract
Aim To estimate whether laxatives prescribed for constipation in Parkinson's disease (PD) could moderate rigidity. Constipation predates diagnosis of PD by decades. Deposition of misfolded protein may begin in the gut, driven by dysbiosis. Successive antimicrobial exposures are associated with cumulative increase in rigidity, and rigidity has biological gradients on circulating leukocyte‐subset counts. Methods Retrospective service evaluation, in a gut/brain axis clinic, yielded an interrupted time series, relating maintenance laxative and other medication to rigidity, in consecutive outpatients identified by inclusion and exclusion criteria. Objective assessment of rigidity was used to bring greater sensitivity to change, validated against subjective gold standard (UPDRS). Results There were 1493 measurements of torque required to extend (flexor rigidity) and flex (extensor rigidity) the forearm in 79 PD patients over 374 person‐years. Both were strongly associated with UPDRS (P < 0.001 and P = 0.008, respectively). Before exhibition of laxative, flexor rigidity increased by 6% (95% CI 1, 10) per year, plateauing at −2% (−4, 1) per year after, with no shift at initiation. Change in slope was significant (P = 0.002), and manifest in those naïve to antiparkinsonian medication. The change was replicated for individual laxative classes (bulk, osmotic, enterokinetic). There was no temporal change in extensor rigidity. Limited experience with a quanylate cyclase‐C receptor agonist (17 patients, 6 person‐years) indicated a large and significant step down in flexor and extensor rigidity, of 19% (1, 34) and 16% (6, 24) respectively (P = 0.04 and <0.001). Conclusions Maintenance laxative usage was associated with apparent stemming of the temporal increase in rigidity in PD, adding to indicative evidence of a continuing role of gastrointestinal dysbiosis in pathogenesis.
Collapse
Affiliation(s)
- Aisha D Augustin
- Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK.,The Maudsley Hospital, London, SE5 8AZ, UK
| | - André Charlett
- Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK.,Statistics Unit, Centre for Infectious Disease Surveillance and Control, Public Health England, London, NW9 5EQ, UK
| | - Clive Weller
- Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK
| | - Sylvia M Dobbs
- Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK.,The Maudsley Hospital, London, SE5 8AZ, UK.,Department of Gastroenterology, King's College Hospital, London, SE5 9RS, UK
| | - David Taylor
- Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK.,The Maudsley Hospital, London, SE5 8AZ, UK
| | - Ingvar Bjarnason
- Department of Gastroenterology, King's College Hospital, London, SE5 9RS, UK
| | - R John Dobbs
- Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK.,The Maudsley Hospital, London, SE5 8AZ, UK.,Department of Gastroenterology, King's College Hospital, London, SE5 9RS, UK
| |
Collapse
|
50
|
Ugrumov MV. [Development of preclinical diagnosis and preventive treatment of neurodegenerative diseases]. Zh Nevrol Psikhiatr Im S S Korsakova 2016; 115:4-14. [PMID: 26978045 DOI: 10.17116/jnevro20151151114-14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Neurodegenerative diseases (NDD) are serious fatal neurological and mental diseases that resulted in disability and fethal outcome. Based on the advances of basic sciences over the last two decades, new knowledge on the risk factors for NDD and molecular mechanisms of the pathogenesis are obtained. It has been shown that the accelerated process of neuronal death which is the main cause of NDD development begins long before the appearance of clinical symptoms. The first symptoms appeared only after the death of most specific regulatory neurons and exhaustion of brain compensatory reserve. Only at that time, one can make the diagnosis and start traditional treatment of patients that accounts for the extremely low efficacy of the latter. Currently, complex preclinical diagnosis based on the identification of relatively specific clinical precursors and peripheral biomarkers has been developing. Development of preclinical diagnosis and preventive treatment is a strategic issue of modern neurology and psychiatry. The resolution of this issue allows to consider NDD as cured, but not fatal, diseases.
Collapse
Affiliation(s)
- M V Ugrumov
- Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|