1
|
Duranti E, Villa C. From Brain to Muscle: The Role of Muscle Tissue in Neurodegenerative Disorders. BIOLOGY 2024; 13:719. [PMID: 39336146 PMCID: PMC11428675 DOI: 10.3390/biology13090719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/02/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024]
Abstract
Neurodegenerative diseases (NDs), like amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), and Parkinson's disease (PD), primarily affect the central nervous system, leading to progressive neuronal loss and motor and cognitive dysfunction. However, recent studies have revealed that muscle tissue also plays a significant role in these diseases. ALS is characterized by severe muscle wasting as a result of motor neuron degeneration, as well as alterations in gene expression, protein aggregation, and oxidative stress. Muscle atrophy and mitochondrial dysfunction are also observed in AD, which may exacerbate cognitive decline due to systemic metabolic dysregulation. PD patients exhibit muscle fiber atrophy, altered muscle composition, and α-synuclein aggregation within muscle cells, contributing to motor symptoms and disease progression. Systemic inflammation and impaired protein degradation pathways are common among these disorders, highlighting muscle tissue as a key player in disease progression. Understanding these muscle-related changes offers potential therapeutic avenues, such as targeting mitochondrial function, reducing inflammation, and promoting muscle regeneration with exercise and pharmacological interventions. This review emphasizes the importance of considering an integrative approach to neurodegenerative disease research, considering both central and peripheral pathological mechanisms, in order to develop more effective treatments and improve patient outcomes.
Collapse
Affiliation(s)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
2
|
Li C, Li M, Jin Y, An Q, Dang H, Gong W. Investigating the therapeutic effects of a Japanese sake yeast supplement on a zebrafish model of Parkinson's disease: Antioxidant and inflammatory responses. Exp Gerontol 2024; 194:112509. [PMID: 38964429 DOI: 10.1016/j.exger.2024.112509] [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: 05/28/2024] [Revised: 06/23/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Sake may potentially halt the progression of Parkinson's disease due to its properties, yet no studies have explored its effects. This preliminary study aimed to assess the impact of sake supplementation on Parkinson's disease using a zebrafish model. Sixty fish were divided into six groups: control, rotenone (ROT), and groups administered rotenone along with sake at concentrations of 25, 50, 75, and 100 mg/L (25S, 50S, 75S, and 100S). After 28 days of treatment, behavioral responses and the activities of catalase (CAT), superoxide dismutase (SOD), reduced glutathione (GSH), and glutathione-S-transferase (GST), as well as the expressions of TNF-α, IL-1β, and COX-2, were evaluated. The results indicated that rotenone administration significantly reduced crossing number (P = 0.001), entries in the top area (P = 0.001), and time spent in the top area (P = 0.001). It also markedly increased levels of TBARS and SH compared to the control group (P = 0.001). Rotenone significantly decreased CAT, SOD, and GSH activities while increasing GST levels. Furthermore, it upregulated the expressions of TNF-α (P = 0.001), IL-1β (P = 0.001), and COX-2 (P = 0.001). Supplementation with sake, particularly at higher doses, reversed the adverse effects of rotenone on behavioral, oxidative, and inflammatory responses. In conclusion, sake shows promise for preventing Parkinson's disease pending further clinical studies.
Collapse
Affiliation(s)
- Chang Li
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, China; Department of Traditional Chinese Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meihe Li
- Department of Renal Transplantation, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Jin
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Qing An
- First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang 712046, China
| | - Huimin Dang
- Department of Traditional Chinese Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Wei Gong
- Department of Pediatric Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
3
|
Park TY, Jeon J, Cha Y, Kim KS. Past, present, and future of cell replacement therapy for parkinson's disease: a novel emphasis on host immune responses. Cell Res 2024; 34:479-492. [PMID: 38777859 PMCID: PMC11217403 DOI: 10.1038/s41422-024-00971-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/28/2024] [Indexed: 05/25/2024] Open
Abstract
Parkinson's disease (PD) stands as the second most common neurodegenerative disorder after Alzheimer's disease, and its prevalence continues to rise with the aging global population. Central to the pathophysiology of PD is the specific degeneration of midbrain dopamine neurons (mDANs) in the substantia nigra. Consequently, cell replacement therapy (CRT) has emerged as a promising treatment approach, initially supported by various open-label clinical studies employing fetal ventral mesencephalic (fVM) cells. Despite the initial favorable results, fVM cell therapy has intrinsic and logistical limitations that hinder its transition to a standard treatment for PD. Recent efforts in the field of cell therapy have shifted its focus towards the utilization of human pluripotent stem cells, including human embryonic stem cells and induced pluripotent stem cells, to surmount existing challenges. However, regardless of the transplantable cell sources (e.g., xenogeneic, allogeneic, or autologous), the poor and variable survival of implanted dopamine cells remains a major obstacle. Emerging evidence highlights the pivotal role of host immune responses following transplantation in influencing the survival of implanted mDANs, underscoring an important area for further research. In this comprehensive review, building upon insights derived from previous fVM transplantation studies, we delve into the functional ramifications of host immune responses on the survival and efficacy of grafted dopamine cells. Furthermore, we explore potential strategic approaches to modulate the host immune response, ultimately aiming for optimal outcomes in future clinical applications of CRT for PD.
Collapse
Affiliation(s)
- Tae-Yoon Park
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, Belmont, MA, USA
- Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Jeha Jeon
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, Belmont, MA, USA
- Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Young Cha
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, Belmont, MA, USA
- Program in Neuroscience, Harvard Medical School, Belmont, MA, USA
| | - Kwang-Soo Kim
- Molecular Neurobiology Laboratory, Department of Psychiatry and McLean Hospital, Harvard Medical School, Belmont, MA, USA.
- Program in Neuroscience, Harvard Medical School, Belmont, MA, USA.
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard Medical School, Belmont, MA, USA.
| |
Collapse
|
4
|
Mao Z, Nie Q, Xue Z, Li Z. Coexistence of Parkinson's disease and myasthenia gravis: A case report and literature review. Exp Ther Med 2024; 28:282. [PMID: 38800046 PMCID: PMC11117104 DOI: 10.3892/etm.2024.12570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
The coexistence of Parkinson's disease (PD) and myasthenia gravis (MG) is rare. When similar symptoms of both diseases overlap, it is challenging to make a concomitant diagnosis of PD and MG. The present study describes the case of a patient with concomitant PD and MG. In addition, a systematic literature review was conducted by searching PubMed and Embase for reports on all patients with concomitant PD and MG, which were then grouped and compared according to different preexisting diseases. Finally, a total of 47 cases of concomitant PD and MG (35 men; 12 women), including the present case, were analyzed. The median age of the patients at first diagnosis was 66.59±9.91 years. The interval between the two diseases varied from 2 months to 22 years. Based on the sequential occurrence of these two diseases, the patients were categorized into three groups: The prePD-MG (30 cases), preMG-PD (12 cases), and coPD-MG (5 cases) groups. In the prePD-MG group, the onset age of MG was older and head drop was more common. In the preMG-PD group, the patients were more likely to have comorbid immune diseases.
Collapse
Affiliation(s)
- Zhijuan Mao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qing Nie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zheng Xue
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhijun Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
5
|
Shen T, Cui G, Chen H, Huang L, Song W, Zu J, Zhang W, Xu C, Dong L, Zhang Y. TREM-1 mediates interaction between substantia nigra microglia and peripheral neutrophils. Neural Regen Res 2024; 19:1375-1384. [PMID: 37905888 PMCID: PMC11467918 DOI: 10.4103/1673-5374.385843] [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: 02/14/2023] [Revised: 03/22/2023] [Accepted: 05/29/2023] [Indexed: 11/02/2023] Open
Abstract
Microglia-mediated neuroinflammation is considered a pathological feature of Parkinson’s disease. Triggering receptor expressed on myeloid cell-1 (TREM-1) can amplify the inherent immune response, and crucially, regulate inflammation. In this study, we found marked elevation of serum soluble TREM-1 in patients with Parkinson’s disease that positively correlated with Parkinson’s disease severity and dyskinesia. In a mouse model of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson’s disease, we found that microglial TREM-1 expression also increased in the substantia nigra. Further, TREM-1 knockout alleviated dyskinesia in a mouse model of Parkinson’s disease and reduced dopaminergic neuronal injury. Meanwhile, TREM-1 knockout attenuated the neuroinflammatory response, dopaminergic neuronal injury, and neutrophil migration. Next, we established an in vitro 1-methyl-4-phenyl-pyridine-induced BV2 microglia model of Parkinson’s disease and treated the cells with the TREM-1 inhibitory peptide LP17. We found that LP17 treatment reduced apoptosis of dopaminergic neurons and neutrophil migration. Moreover, inhibition of neutrophil TREM-1 activation diminished dopaminergic neuronal apoptosis induced by lipopolysaccharide. TREM-1 can activate the downstream CARD9/NF-κB proinflammatory pathway via interaction with SYK. These findings suggest that TREM-1 may play a key role in mediating the damage to dopaminergic neurons in Parkinson’s disease by regulating the interaction between microglia and peripheral neutrophils.
Collapse
Affiliation(s)
- Tong Shen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, Jiangsu Province, China
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Guiyun Cui
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Hao Chen
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Long Huang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, Jiangsu Province, China
| | - Wei Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, Jiangsu Province, China
| | - Jie Zu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Wei Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Chuanying Xu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Liguo Dong
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yongmei Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou, Jiangsu Province, China
| |
Collapse
|
6
|
Hong J, Li Y, Chen L, Han D, Li Y, Mi X, Liu K, Wang Q, Song Y, Liu T, Yang N, Liu Y, Li Z, Guo X. A53T α-synuclein mutation increases susceptibility to postoperative delayed neurocognitive recovery via hippocampal Ang-(1-7)/MasR axis. Biochem Pharmacol 2024; 224:116261. [PMID: 38705534 DOI: 10.1016/j.bcp.2024.116261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Delayed neurocognitive recovery (dNCR) is a common complication in geriatric surgical patients. The impact of anesthesia and surgery on patients with neurodegenerative diseases, such as Parkinson's disease (PD) or prion disease, has not yet been reported. In this study, we aimed to determine the association between a pre-existing A53T genetic background, which involves a PD-related point mutation, and the development of postoperative dNCR. We observed that partial hepatectomy induced hippocampus-dependent cognitive deficits in 5-month-old A53T transgenic mice, a model of early-stage PD without cognitive deficits, unlike in age-matched wild-type (WT) mice. We respectively examined molecular changes at 6 h, 1 day, and 2 days after partial hepatectomy and observed that cognitive changes were accompanied by weakened angiotensin-(1-7)/Mas receptor [Ang-(1-7)/MasR] axis, increased alpha-synuclein (α-syn) expression and phosphorylation, decreased methylated protein phosphatase-2A (Me-PP2A), and prompted microglia M1 polarization and neuronal apoptosis in the hippocampus at 1 day after surgery. Nevertheless, no changes in blood-brain barrier (BBB) integrity or plasma α-syn levels in either A53T or WT mice. Furthermore, intranasal administration of selective MasR agonist AVE 0991, reversed the mentioned cognitive deficits in A53T mice, enhanced MasR expression, reduced α-syn accumulation and phosphorylation, and attenuated microglia activation and apoptotic response. Our findings suggest that individuals with the A53T genetic background may be more susceptible to developing postoperative dNCR. This susceptibility could be linked to central α-syn accumulation mediated by the weakened Ang-(1-7)/MasR/methyl-PP2A signaling pathway in the hippocampus following surgery, independent of plasma α-syn level and BBB.
Collapse
Affiliation(s)
- Jingshu Hong
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yue Li
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Lei Chen
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Dengyang Han
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yitong Li
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Xinning Mi
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Kaixi Liu
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Qian Wang
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yanan Song
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Beijing Center of Quality Control and Improvement on Clinical Anesthesia, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Taotao Liu
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Ning Yang
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Yajie Liu
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China
| | - Zhengqian Li
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Beijing Center of Quality Control and Improvement on Clinical Anesthesia, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Anesthesia and Perioperative Medicine Branch of China International Exchange and Promotive Association for Medical and Health Care (CPAM), No. 49, North Garden Street, Haidian District, Beijing 100191, China.
| | - Xiangyang Guo
- Department of Anesthesiology, Peking University Third Hospital, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Beijing Center of Quality Control and Improvement on Clinical Anesthesia, No. 49, North Garden Street, Haidian District, Beijing 100191, China; Anesthesia and Perioperative Medicine Branch of China International Exchange and Promotive Association for Medical and Health Care (CPAM), No. 49, North Garden Street, Haidian District, Beijing 100191, China.
| |
Collapse
|
7
|
Guan Y, Cao M, Wu X, Yan J, Hao Y, Zhang C. CD28 null T cells in aging and diseases: From biology to assessment and intervention. Int Immunopharmacol 2024; 131:111807. [PMID: 38471362 DOI: 10.1016/j.intimp.2024.111807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/25/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024]
Abstract
CD28null T cells, an atypical subset characterized by the loss of CD28 costimulatory molecule expression, exhibit functional variants and progressively expand with age. Moreover, T cells with these phenotypes are found in both typical and atypical humoral immune responses. Consequently, they accumulate during infectious diseases, autoimmune disorders, cardiovascular conditions, and neurodegenerative ailments. To provide an in-depth review of the current knowledge regarding CD28null T cells, we specifically focus on their phenotypic and functional characteristics as well as their physiological roles in aging and diseases. While uncertainties regarding the clinical utility remains, we will review the following two crucial research perspectives to explore clinical translational applications of the research on this specific T cell subset: 1) addressing the potential utility of CD28null T cells as immunological markers for prognosis and adverse outcomes in both aging and disease, and 2) speculating on the potential of targeting CD28null T cells as an interventional strategy for preventing or delaying immune aging processes and disease progression.
Collapse
Affiliation(s)
- Yuqi Guan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Ming Cao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Xiaofen Wu
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Jinhua Yan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China
| | - Yi Hao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, China.
| |
Collapse
|
8
|
Saadullah M, Tariq H, Chauhdary Z, Saleem U, Anwer Bukhari S, Sehar A, Asif M, Sethi A. Biochemical properties and biological potential of Syzygium heyneanum with antiparkinson's activity in paraquat induced rodent model. PLoS One 2024; 19:e0298986. [PMID: 38551975 PMCID: PMC10980224 DOI: 10.1371/journal.pone.0298986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/01/2024] [Indexed: 04/01/2024] Open
Abstract
Syzygium heyneanum is a valuable source of flavonoids and phenols, known for their antioxidant and neuroprotective properties. This research aimed to explore the potential of Syzygium heyneanum ethanol extract (SHE) in countering Parkinson's disease. The presence of phenols and flavonoids results in SHE displaying an IC50 value of 42.13 when assessed in the DPPH scavenging assay. Rats' vital organs (lungs, heart, spleen, liver, and kidney) histopathology reveals little or almost no harmful effect. The study hypothesized that SHE possesses antioxidants that could mitigate Parkinson's symptoms by influencing α-synuclein, acetylcholinesterase (AChE), TNF-α, and IL-1β. Both in silico and in vivo investigations were conducted. The Parkinson's rat model was established using paraquat (1 mg/kg, i.p.), with rats divided into control, disease control, standard, and SHE-treated groups (150, 300, and 600 mg/kg) for 21 days. According to the ELISA statistics, the SHE treated group had lowers levels of IL-6 and TNF-α than the disease control group, which is a sign of neuroprotection. Behavioral and biochemical assessments were performed, alongside mRNA expression analyses using RT-PCR to assess SHE's impact on α-synuclein, AChE, TNF-α, and interleukins in brain homogenates. Behavioral observations demonstrated dose-dependent improvements in rats treated with SHE (600 > 300 > 150 mg/kg). Antioxidant enzyme levels (catalase, superoxide dismutase, glutathione) were significantly restored, particularly at a high dose, with notable reduction in malondialdehyde. The high dose of SHE notably lowered acetylcholinesterase levels. qRT-PCR results indicated reduced mRNA expression of IL-1β, α-synuclein, TNF-α, and AChE in SHE-treated groups compared to disease controls, suggesting neuroprotection. In conclusion, this study highlights Syzygium heyneanum potential to alleviate Parkinson's disease symptoms through its antioxidant and modulatory effects on relevant biomarkers.
Collapse
Affiliation(s)
- Malik Saadullah
- Department of Pharmaceutical Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Hafsa Tariq
- Department of Pharmaceutical Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Zunera Chauhdary
- Department of Pharmacology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Uzma Saleem
- Department of Pharmacology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Shazia Anwer Bukhari
- Department of Biochemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Amna Sehar
- Department of Pharmaceutical Chemistry, Government College University Faisalabad, Faisalabad, Pakistan
| | - Muhammad Asif
- Department of Pharmacology, Islamia University Bahawalpur, Bahawalpur, Pakistan
| | - Aisha Sethi
- Department of Pharmaceutics, Government College University Faisalabad, Faisalabad, Pakistan
| |
Collapse
|
9
|
Eser P, Kocabicak E, Bekar A, Temel Y. The interplay between neuroinflammatory pathways and Parkinson's disease. Exp Neurol 2024; 372:114644. [PMID: 38061555 DOI: 10.1016/j.expneurol.2023.114644] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/25/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024]
Abstract
Parkinson's disease, a progressive neurodegenerative disorder predominantly affecting elderly, is marked by the gradual degeneration of the nigrostriatal dopaminergic pathway, culminating in neuronal loss within the substantia nigra pars compacta (SNpc) and dopamine depletion. At the molecular level, neuronal loss in the SNpc has been attributed to factors including neuroinflammation, impaired protein homeostasis, as well as mitochondrial dysfunction and the resulting oxidative stress. This review focuses on the interplay between neuroinflammatory pathways and Parkinson's disease, drawing insights from current literature.
Collapse
Affiliation(s)
- Pinar Eser
- Bursa Uludag University School of Medicine, Department of Neurosurgery, Bursa, Turkey.
| | - Ersoy Kocabicak
- Ondokuz Mayis University, Health Practise and Research Hospital, Neuromodulation Center, Samsun, Turkey
| | - Ahmet Bekar
- Bursa Uludag University School of Medicine, Department of Neurosurgery, Bursa, Turkey
| | - Yasin Temel
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
10
|
Wei BR, Zhao YJ, Cheng YF, Huang C, Zhang F. Helicobacter pylori infection and Parkinson's Disease: etiology, pathogenesis and levodopa bioavailability. Immun Ageing 2024; 21:1. [PMID: 38166953 PMCID: PMC10759355 DOI: 10.1186/s12979-023-00404-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024]
Abstract
Parkinson's disease (PD), a neurodegenerative disorder with an unknown etiology, is primarily characterized by the degeneration of dopamine (DA) neurons. The prevalence of PD has experienced a significant surge in recent years. The unidentified etiology poses limitations to the development of effective therapeutic interventions for this condition. Helicobacter pylori (H. pylori) infection has affected approximately half of the global population. Mounting evidences suggest that H. pylori infection plays an important role in PD through various mechanisms. The autotoxin produced by H. pylori induces pro-inflammatory cytokines release, thereby facilitating the occurrence of central inflammation that leads to neuronal damage. Simultaneously, H. pylori disrupts the equilibrium of gastrointestinal microbiota with an overgrowth of bacteria in the small intestinal known as small intestinal bacterial overgrowth (SIBO). This dysbiosis of the gut flora influences the central nervous system (CNS) through microbiome-gut-brain axis. Moreover, SIBO hampers levodopa absorption and affects its therapeutic efficacy in the treatment of PD. Also, H. pylori promotes the production of defensins to regulate the permeability of the blood-brain barrier, facilitating the entry of harmful factors into the CNS. In addition, H. pylori has been found to induce gastroparesis, resulting in a prolonged transit time for levodopa to reach the small intestine. H. pylori may exploit levodopa to facilitate its own growth and proliferation, or it can inflict damage to the gastrointestinal mucosa, leading to gastrointestinal ulcers and impeding levodopa absorption. Here, this review focused on the role of H. pylori infection in PD from etiology, pathogenesis to levodopa bioavailability.
Collapse
Affiliation(s)
- Bang-Rong Wei
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yu-Jia Zhao
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yu-Feng Cheng
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China
| | - Chun Huang
- The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Feng Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education and Key Laboratory of Basic Pharmacology of Guizhou Province and Laboratory Animal Centre, Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
11
|
Hu Y, Xu S. Association between Parkinson's disease and the risk of adverse cardiovascular events: a systematic review and meta-analysis. Front Cardiovasc Med 2023; 10:1284826. [PMID: 38144366 PMCID: PMC10748497 DOI: 10.3389/fcvm.2023.1284826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/21/2023] [Indexed: 12/26/2023] Open
Abstract
Background This review aims to examine the association of Parkinson's disease (PD) with the increased risk of cardiovascular events. Methods PubMed, Embase, CENTRAL, and Scopus databases were electronically searched for papers published up to 5 May 2023. Studies reporting the association between PD and the subsequent risks of stroke, myocardial infarction (MI), and cardiovascular mortality were included. Results Sixteen studies were included in this review. The clinical data of 101,712 PD patients were compared with that of the control group of 204,901 patients without PD in the included studies. Meta-analysis showed that PD patients had an increased risk of stroke compared with patients without PD (odds ratio (OR): 1.49; 95% confidence interval (CI): 1.30, 1.72; I2 = 76%). The pooled analysis demonstrated no significant increase in the risk of MI (OR: 1.16; 95% CI: 0.85, 1.59; I2 = 82%) and cardiovascular mortality (OR: 1.20; 95% CI: 0.93, 1.54; I2 = 65%) in PD patients. However, data from cohort studies indicated a possibility of higher risk of MI (OR: 1.36; 95% CI: 1.01, 1.84; I2 = 80%) and cardiovascular mortality (OR: 1.22; 95% CI: 1.00, 1.60; I2 = 62%) in patients with PD. Conclusion Patients with PD may have an increased risk of stroke as compared with the age- and gender-matched general population. While our results show that PD does not increase the overall risk of MI and cardiovascular mortality, analysis of cohort studies alone demonstrated that these risks may be higher in patients with PD. The current evidence is of very low quality. Further prospective cohort studies from different countries that would account for important cardiovascular risk factors are needed to improve the current evidence. Systematic Review Registration https://www.crd.york.ac.uk/prospero/, PROSPERO (CRD42023421924).
Collapse
Affiliation(s)
| | - Shanxia Xu
- Department of Neurology, Huzhou Central Hospital, Affiliated Central Hospital of HuZhou University, Huzhou, Zhejiang, China
| |
Collapse
|
12
|
Mohammed HS, Hosny EN, Sawie HG, Khadrawy YA. Transcranial photobiomodulation ameliorates midbrain and striatum neurochemical impairments and behavioral deficits in reserpine-induced parkinsonism in rats. Photochem Photobiol Sci 2023; 22:2891-2904. [PMID: 37917308 DOI: 10.1007/s43630-023-00497-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/15/2023] [Indexed: 11/04/2023]
Abstract
Photobiomodulation (PBM) of deep brain structures through transcranial infrared irradiation might be an effective treatment for Parkinson's disease (PD). However, the mechanisms underlying this intervention should be elucidated to optimize the therapeutic outcome and maximize therapeutic efficacy. The present study aimed at investigating the oxidative stress-related parameters of malondialdehyde (MDA), nitric oxide (NO), and reduced glutathione (GSH) and the enzymatic activities of sodium-potassium-ATPase (Na+, K+-ATPase), Acetylcholinesterase (AChE), and monoamine oxidase (MAO) and monoamine levels (dopamine (DA), norepinephrine (NE) and serotonin (5-HT) in the midbrain and striatum of reserpine-induced PD in an animal model treated with PBM. Furthermore, the locomotor behavior of the animals has been determined by the open field test. Animals were divided into three groups; the control group, the PD-induced model group, and the PD-induced model treated with the PBM group. Non-invasive treatment of animals for 14 days with 100 mW, 830 nm laser has demonstrated successful attainment in the recovery of oxidative stress, and enzymatic activities impairments induced by reserpine (0.2 mg/kg) in both midbrain and striatum of adult male Wistar rats. PBM also improved the decrease in DA, NE, and 5-HT in the investigated brain regions. On a behavioral level, animals showed improvement in their locomotion activity. These findings have shed more light on some mechanisms underlying the treatment potential of PBM and displayed the safety, easiness, and efficacy of PBM treatment as an alternative to pharmacological treatment for PD.
Collapse
Affiliation(s)
- Haitham S Mohammed
- Faculty of Science, Biophysics Department, Cairo University, Giza, Egypt.
| | - Eman N Hosny
- Medical Division, Medical Physiology Department, National Research Centre, Giza, Egypt
| | - Hussein G Sawie
- Medical Division, Medical Physiology Department, National Research Centre, Giza, Egypt
| | - Yasser A Khadrawy
- Medical Division, Medical Physiology Department, National Research Centre, Giza, Egypt
| |
Collapse
|
13
|
Zhou Z, Zhang M, Fang Q, Huang J. Relationship between Parkinson's disease and cardio-cerebrovascular diseases: a Mendelian randomized study. Sci Rep 2023; 13:20428. [PMID: 37993489 PMCID: PMC10665329 DOI: 10.1038/s41598-023-47708-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 11/17/2023] [Indexed: 11/24/2023] Open
Abstract
Parkinson's disease (PD) and cardio-cerebrovascular diseases are related, according to earlier studies, but these studies have some controversy. Our aim was to assess the impact of PD on cardiocerebrovascular diseases using a Mendelian randomization (MR) method. The data for PD were single nucleotide polymorphisms (SNPs) from a publicly available genome-wide association study (GWAS) dataset containing data on 482,730 individuals. And the outcome SNPs data is were derived from five different GWAS datasets. The basic method for MR analysis was the inverse variance weighted (IVW) approach. We use the weighted median method and the MR-Egger method to supplement the MR analysis conclusion. Finally, We used Cochran's Q test to test heterogeneity, MR-PRESSO method and leave-one-out analysis method to perform sensitivity analysis. We used ratio ratios (OR) to assess the strength of the association between exposure and outcome, and 95% confidence intervals (CI) to show the reliability of the results. Our findings imply that PD is linked to a higher occurrence of coronary artery disease (CAD) (OR = 1.055, 95% CI 1.020-1.091, P = 0.001), stroke (OR = 1.039, 95% CI 1.007-1.072, P = 0.014). IVW analyses for stroke's subgroups of ischemic stroke (IS) and 95% CI 1.007-1.072, P = 0.014). IVW analyses for stroke's subgroups of ischemic stroke (IS) and cardioembolic stroke (CES) also yielded positive results, respectively (OR = 1.043, 95% CI 1.008-1.079, P = 0.013), (OR = 1.076, 95% CI 1.008-1.149, P = 0.026). There is no evidence of a relationship between PD and other cardio-cerebrovascular diseases. Additionally, sensitivity analysis revealed reliable outcomes. Our MR study analysis that PD is related with an elevated risk of CAD, stroke, IS, and CES.
Collapse
Affiliation(s)
- Zhongzheng Zhou
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Muzi Zhang
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qinghua Fang
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Huang
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
14
|
Numakawa T, Kajihara R. Involvement of brain-derived neurotrophic factor signaling in the pathogenesis of stress-related brain diseases. Front Mol Neurosci 2023; 16:1247422. [PMID: 37781095 PMCID: PMC10537938 DOI: 10.3389/fnmol.2023.1247422] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/31/2023] [Indexed: 10/03/2023] Open
Abstract
Neurotrophins including brain-derived neurotrophic factor, BDNF, have critical roles in neuronal differentiation, cell survival, and synaptic function in the peripheral and central nervous system. It is well known that a variety of intracellular signaling stimulated by TrkB, a high-affinity receptor for BDNF, is involved in the physiological and pathological neuronal aspects via affecting cell viability, synaptic function, neurogenesis, and cognitive function. As expected, an alteration of the BDNF/TrkB system is suspected to be one of the molecular mechanisms underlying cognitive decline in cognitive diseases and mental disorders. Recent evidence has also highlighted a possible link between the alteration of TrkB signaling and chronic stress. Furthermore, it has been demonstrated that downregulation of the BDNF/TrkB system and chronic stress have a role in the pathogenesis of Alzheimer's disease (AD) and mental disorders. In this review, we introduce current evidence showing a close relationship between the BDNF/TrkB system and the development of cognition impairment in stress-related disorders, and the possible contribution of the upregulation of the BDNF/TrkB system in a therapeutic approach against these brain diseases.
Collapse
Affiliation(s)
- Tadahiro Numakawa
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Ryutaro Kajihara
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
- Department of Biomedical Laboratory Sciences, Faculty of Life Science, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
15
|
Zhang Y, Li S, Hou L, Wu M, Liu J, Wang R, Wang Q, Zhao J. NLRP3 mediates the neuroprotective effects of SVHRSP derived from scorpion venom in rotenone-induced experimental Parkinson's disease model. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116497. [PMID: 37072089 DOI: 10.1016/j.jep.2023.116497] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/31/2023] [Accepted: 04/11/2023] [Indexed: 05/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In traditional Chinese medicine, scorpion is used to treat diseases with symptoms such as trembling, convulsion and dementia. Our laboratory employs patented technology to extract and purify the active single component from scorpion venom. We then utilize mass spectrometry to determine the amino acid sequence of the polypeptide and synthesize it artificially to acquire the polypeptide with a purity of 99.3%, named SVHRSP (Scorpion Venom Heat-Resistant Peptide). SVHRSP has been demonstrated to display potent neuroprotective efficacy in Parkinson's disease. AIM OF THE STUDY To explore the molecular mechanisms and potential molecular targets of SVHRSP-afforded neuroprotection in PD mouse models, as well as to investigate the role of NLRP3 in SVHRSP-mediated neuroprotection. MATERIALS AND METHODS The PD mouse model was induced by rotenone and the neuroprotective role of SVHRSP on the PD mouse model was measured using the gait test, rotarod test, the number of dopaminergic neurons, and the activation of microglia. RNA sequencing and GSEA analysis were performed to find the differentially biological pathways regulated by SVHRSP. Primary mid-brain neuron-glial cultures and NLRP3-/- mice were applied to verify the role of NLRP3 by using qRT-PCR, western blotting, enzyme-linked immunosorbent assay (ELISA) and immunostaining. RESULTS SVHRSP-afforded dopaminergic neuroprotection was accompanied with inhibition of microglia-mediated neuroinflammatory pathways. Importantly, depletion of microglia markedly reduced the neuroprotective efficacy of SVHRSP against rotenone-induced dopaminergic neurotoxicity in vitro. SVHRSP inhibited microglial NOD-like receptor pathway, mRNA expression and protein level of NLRP3 in rotenone PD mice. SVHRSP also reduced rotenone-induced caspse-1 activation and IL-1β maturation, indicating that SVHRSP mitigated activation of NLRP3 inflammasome. Moreover, inactivation of NLRP3 inflammasome by MCC950 or genetic deletion of NLRP3 almost abolished SVHRSP-afforded anti-inflammatory, neuroprotective effects and improvement of motor performance in response to rotenone. CONCLUSIONS NLRP3 mediated the neuroprotective effects of SVHRSP in rotenone-induced experimental PD model, providing additional evidence for the mechanisms of SVHRSP-afforded anti-inflammatory and neuroprotective effects in PD.
Collapse
Affiliation(s)
- Yu Zhang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China; Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Sheng Li
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Liyan Hou
- Dalian Medical University Library, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Mingyang Wu
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Jianing Liu
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Ruonan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China
| | - Qingshan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China; School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China.
| | - Jie Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, 116044, China.
| |
Collapse
|
16
|
Nabizadeh F, Valizadeh P, Sharifi P, Zafari R, Mirmosayyeb O. Risk of myocardial infarction in Parkinson's disease: A systematic review and meta-analysis. Eur J Neurol 2023; 30:2557-2569. [PMID: 37151196 DOI: 10.1111/ene.15838] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/06/2023] [Accepted: 04/16/2023] [Indexed: 05/09/2023]
Abstract
BACKGROUND AND PURPOSE Previous studies investigating cardiovascular disorders in patients with Parkinson's disease (PD) showed heterogeneous results regarding whether there is a higher or lower risk of myocardial infarction (MI) in these patients compared to the general population. Because of the inconsistency in findings, herein the aim was to perform a systematic review and meta-analysis to investigate the risk of MI in patients with PD. METHODS A comprehensive literature search was performed using four databases, PubMed, Web of Science, Scopus and Embase, in June 2022. Peer-reviewed observational studies comprising case-controls, cohort, cross-sectional and longitudinal studies that reported MI in the PD population were included. RESULTS After the screening, 20 studies with a total of 80,441 patients with PD and 802,857 controls were included in our qualitative and quantitative synthesis. The pooled estimated odds ratio for MI in PD patients compared to controls was 0.80 (95% confidence interval [CI] 0.56-1.05) which indicates that there is no association. The pooled prevalence of MI was 5% (95% CI 3%-7%) with a range of 1%-20% amongst patients with PD. The men (6%, 95% CI 1%-13%) and women (6%, 95% CI 1%-14%, Q = 29.27, I2 = 98.50%, p < 0.001) had similar MI prevalence. CONCLUSION This comprehensive systematic review and meta-analysis provide compelling evidence that PD is associated with a reduced risk of MI. Whilst the exact mechanism underlying this association remains to be fully elucidated, it is clear that certain risk factors for cardiac events appear to be less present in PD patients, which may serve as a protective factor. However, given the reports of increased risk for cerebrovascular events in PD patients, it is possible that the major risk factors for MI and cardiovascular accidents in this population differ. These findings have important implications for clinical management and further research in this area is warranted.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- Neuroscience Research Group (NRG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parya Valizadeh
- School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Parisa Sharifi
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Rasa Zafari
- School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Omid Mirmosayyeb
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
17
|
Mohapatra L, Mishra D, Shiomurti Tripathi A, Kumar Parida S. Immunosenescence as a convergence pathway in neurodegeneration. Int Immunopharmacol 2023; 121:110521. [PMID: 37385122 DOI: 10.1016/j.intimp.2023.110521] [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: 05/08/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/01/2023]
Abstract
Immunity refers to the body's defense mechanism to protect itself against illness or to produce antibodies against pathogens. Senescence is a cellular phenomenon that integrates a sustainable growth restriction, other phenotypic abnormalities and including a pro-inflammatory secretome. It is highly involved in regulating developmental stages, tissue homeostasis, and tumor proliferation monitoring. Contemporary experimental reports imply that abolition of senescent cells employing evolved genetic and therapeutic approaches augment the chances of survival and boosts the health span of an individual. Immunosenescence is considered as a process in which dysfunction of the immune system occurs with aging and greatly includes remodeling of lymphoid organs. This in turn causes fluctuations in the immune function of the elderly that has strict relation with the expansion of autoimmune diseases, infections, malignant tumors and neurodegenerative disorders. The interaction of the nervous and immune systems during aging is marked by bi-directional influence and mutual correlation of variations. The enhanced systemic inflammatory condition in the elderly, and the neuronal immune cell activity can be modulated by inflamm-aging and peripheral immunosenescence resulting in chronic low-grade inflammatory processes in the central Nervous system known as neuro-inflammaging. For example, glia excitation by cytokines and glia pro-inflammatory productions contribute significantly to memory injury as well as in acute systemic inflammation, which is associated with high levels of Tumor necrosis factor -α and a rise in cognitive decline. In recent years its role in the pathology of Alzheimer's disease has caught research interest to a large extent. This article reviews the connection concerning the immune and nervous systems and highlights how immunosenescence and inflamm-aging can affect neurodegenerative disorders.
Collapse
Affiliation(s)
- Lucy Mohapatra
- Amity Institute of Pharmacy, Lucknow, Amity University Uttar Pradesh Sector-125, Noida, 201313, India.
| | - Deepak Mishra
- Amity Institute of Pharmacy, Lucknow, Amity University Uttar Pradesh Sector-125, Noida, 201313, India
| | | | | |
Collapse
|
18
|
Miyazaki I, Asanuma M. Multifunctional Metallothioneins as a Target for Neuroprotection in Parkinson's Disease. Antioxidants (Basel) 2023; 12:antiox12040894. [PMID: 37107269 PMCID: PMC10135286 DOI: 10.3390/antiox12040894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/27/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
Parkinson's disease (PD) is characterized by motor symptoms based on a loss of nigrostriatal dopaminergic neurons and by non-motor symptoms which precede motor symptoms. Neurodegeneration accompanied by an accumulation of α-synuclein is thought to propagate from the enteric nervous system to the central nervous system. The pathogenesis in sporadic PD remains unknown. However, many reports indicate various etiological factors, such as oxidative stress, inflammation, α-synuclein toxicity and mitochondrial impairment, drive neurodegeneration. Exposure to heavy metals contributes to these etiopathogenesis and increases the risk of developing PD. Metallothioneins (MTs) are cysteine-rich metal-binding proteins; MTs chelate metals and inhibit metal-induced oxidative stress, inflammation and mitochondrial dysfunction. In addition, MTs possess antioxidative properties by scavenging free radicals and exert anti-inflammatory effects by suppression of microglial activation. Furthermore, MTs recently received attention as a potential target for attenuating metal-induced α-synuclein aggregation. In this article, we summarize MTs expression in the central and enteric nervous system, and review protective functions of MTs against etiopathogenesis in PD. We also discuss neuroprotective strategies for the prevention of central dopaminergic and enteric neurodegeneration by targeting MTs. This review highlights multifunctional MTs as a target for the development of disease-modifying drugs for PD.
Collapse
Affiliation(s)
- Ikuko Miyazaki
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Masato Asanuma
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
19
|
Roussakis AA, Gennaro M, Gordon MF, Reilmann R, Borowsky B, Rynkowski G, Lao-Kaim NP, Papoutsou Z, Savola JM, Hayden MR, Owen DR, Kalk N, Lingford-Hughes A, Gunn RN, Searle G, Tabrizi SJ, Piccini P. A PET-CT study on neuroinflammation in Huntington's disease patients participating in a randomized trial with laquinimod. Brain Commun 2023; 5:fcad084. [PMID: 37020532 PMCID: PMC10069663 DOI: 10.1093/braincomms/fcad084] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 12/19/2022] [Accepted: 03/17/2023] [Indexed: 04/05/2023] Open
Abstract
Microglia activation, an indicator of central nervous system inflammation, is believed to contribute to the pathology of Huntington's disease. Laquinimod is capable of regulating microglia. By targeting the translocator protein, 11C-PBR28 PET-CT imaging can be used to assess the state of regional gliosis in vivo and explore the effects of laquinimod treatment. This study relates to the LEGATO-HD, multi-centre, double-blinded, Phase 2 clinical trial with laquinimod (US National Registration: NCT02215616). Fifteen patients of the UK LEGATO-HD cohort (mean age: 45.2 ± 7.4 years; disease duration: 5.6 ± 3.0 years) were treated with laquinimod (0.5 mg, N = 4; 1.0 mg, N = 6) or placebo (N = 5) daily. All participants had one 11C-PBR28 PET-CT and one brain MRI scan before laquinimod (or placebo) and at the end of treatment (12 months apart). PET imaging data were quantified to produce 11C-PBR28 distribution volume ratios. These ratios were calculated for the caudate and putamen using the reference Logan plot with the corpus callosum as the reference region. Partial volume effect corrections (Müller-Gartner algorithm) were applied. Differences were sought in Unified Huntington's Disease Rating Scale scores and regional distribution volume ratios between baseline and follow-up and between the two treatment groups (laquinimod versus placebo). No significant change in 11C-PBR28 distribution volume ratios was found post treatment in the caudate and putamen for both those treated with laquinimod (N = 10) and those treated with placebo (N = 5). Over time, the patients treated with laquinimod did not show a significant clinical improvement. Data from the 11C-PBR28 PET-CT study indicate that laquinimod may not have affected regional translocator protein expression and clinical performance over the studied period.
Collapse
Affiliation(s)
| | - Marta Gennaro
- Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | | | | | | | | | - Nicholas P Lao-Kaim
- Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Zoe Papoutsou
- Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | | | - Michael R Hayden
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital and Research Institute, University of British Columbia, Vancouver V5Z 4H4, Canada
| | - David R Owen
- Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Nicola Kalk
- Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Anne Lingford-Hughes
- Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Roger N Gunn
- Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
- Invicro, Hammersmith Hospital,, London W12 0NN, UK
| | | | - Sarah J Tabrizi
- Huntington’s Disease Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
| | - Paola Piccini
- Brain Sciences, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| |
Collapse
|
20
|
Nabizadeh F, Mohamadzadeh O, Hosseini H, Rasouli K, Afyouni NE. Serum neurofilament light chain in LRRK2 related Parkinson's disease: A five years follow-up. J Clin Neurosci 2023; 110:12-18. [PMID: 36780781 DOI: 10.1016/j.jocn.2023.01.015] [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: 11/03/2022] [Revised: 12/30/2022] [Accepted: 01/31/2023] [Indexed: 02/12/2023]
Abstract
BACKGROUND Studies revealed that serum neurofilament light chain (NFL) levels not only increase considerably over time in Parkinson's disease (PD) but also have a significant association with disease progression. However, there is no evidence of the level of serum NFL in PD patients with leucine-rich repeat kinase 2 (LRRK2) mutation (LRRK2-PD) which is the most common mutation that causes familial and sporadic PD. AIM Here we aimed to investigate the difference and longitudinal alteration of the serum level of NFL in LRRK2-PD and idiopathic PD (iPD) patients. METHODS We entered 228 iPD and 103 LRRK2-PD patients and 176 healthy controls (HCs) from PPMI. We compared the level of serum NFL at baseline, six months, one year, two years, three years, and five years visits. Also, we used linear mixed models to assess longitudinal changes of serum NFL over six months, one year, two years, three years, and five years within groups. RESULTS We found a significant difference in the level of serum NFL between three groups at baseline, two years, three years, and five years time points. Also, our analysis showed that LRRK2-PD patients had significantly lower serum NFL compared to iPD subjects at baseline. In the longitudinal analysis, there was no significant change in the HCs group over five years. The level of serum NFL was significantly increased after two, three, and five years from baseline in LRRK2-PD patients. Also, we found similar results for iPD subjects after three and five years from baseline. CONCLUSION We can conclude that the overall neurodegeneration might be similar in LRRK2-PD and healthy subjects and lower than the idiopathic form of PD at the early stages, which may disappear in the later stages. Moreover, our findings suggest that the serum NFL might be a more accurate biomarker to distinguish iPD from healthy subjects rather than all PD patients or LRRK2-PD from healthy subjects at the early stages.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- Neuroscience Research Group (NRG), Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Omid Mohamadzadeh
- Department of Neurosurgery, Tehran University of Medical Science, Tehran, Iran
| | - Helia Hosseini
- Faculty of Medicine, Tehran University of Medical Sciences, Iran
| | - Kimia Rasouli
- Student Research Committee, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nazgol Esmalian Afyouni
- Isfahan Neurosciences Research Center, Alzahra Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
21
|
Chang L, Dong WW, Luo B, Qiu C, Lu Y, Lin XJ, Zhang WB. Deep brain stimulation improves central nervous system inflammation in Parkinson's disease: Evidence and perspectives. CNS Neurosci Ther 2023. [PMID: 36942520 DOI: 10.1111/cns.14167] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/25/2023] [Accepted: 02/27/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND In Parkinson's disease (PD), inflammation may lead to the degeneration of dopaminergic (DAergic) neurons. Previous studies showed that inflammatory mediators mainly contributed to this phenomenon. On the other hand, invasive neuromodulation methods such as deep brain stimulation (DBS) have better therapeutic effects for PD. One possibility is that DBS improves PD by influencing inflammation. Therefore, we further explored the mechanisms underlying inflammatory mediators and DBS in the pathogenesis of PD. METHODS We measured serum levels of two inflammatory markers, namely RANTES (regulated on activation, normal T cell expressed and secreted) and tumor necrosis factor-alpha (TNF-α), using Luminex assays in 109 preoperative DBS PD patients, 49 postoperative DBS PD patients, and 113 age- and sex-matched controls. The plasma protein data of the different groups were then statistically analyzed. RESULTS RANTES (p < 0.001) and TNF-α (p = 0.005) levels differed significantly between the three groups. A strong and significant correlation between RANTES levels and Hoehn-Yahr (H-Y) stage was observed in preoperative PD patients (rs = 0.567, p < 0.001). Significant correlations between RANTES levels and Unified Parkinson's Disease Rating Scale III (UPDRS III) score (rs1 = 0.644, p = 0.033 and rs2 = 0.620, p = 0.042) were observed in matched patients. No correlation was observed for TNF-α levels. CONCLUSION The results of this study indicate that PD patients have a persistent inflammatory profile, possibly via recruitment of activated monocytes, macrophages, and T lymphocytes to the central nervous system (CNS). DBS was shown to have a significant therapeutic effect on PD, which may arise by improving the inflammatory environment of the central nervous system.
Collapse
Affiliation(s)
- Lei Chang
- Department of Functional Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Wen-Wen Dong
- Department of Functional Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Bei Luo
- Department of Functional Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Chang Qiu
- Department of Functional Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Lu
- Department of Functional Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Xing-Jian Lin
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Wen-Bin Zhang
- Department of Functional Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
22
|
Essam RM, Kandil EA. p-CREB and p-DARPP-32 orchestrating the modulatory role of cAMP/PKA signaling pathway enhanced by Roflumilast in rotenone-induced Parkinson's disease in rats. Chem Biol Interact 2023; 372:110366. [PMID: 36706892 DOI: 10.1016/j.cbi.2023.110366] [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: 08/30/2022] [Revised: 01/13/2023] [Accepted: 01/23/2023] [Indexed: 01/26/2023]
Abstract
Recently, phosphodiesterases (PDEs) have gained great attention due to their implication in Parkinson's disease (PD) pathogenesis. Noteworthy, the PDE4 enzyme is highly expressed in the striatum and selectively degrades cyclic adenosine monophosphate (cAMP). The cAMP was shown to play a vital role in dopamine (DA) signaling besides maintaining the plasticity of dopaminergic neurons as well as protecting them from inflammation and oxidative stress-mediated death. Thus, PDE4 inhibition could be a promising strategy for treating PD. Accordingly, the present study investigated the neuroprotective efficacy of roflumilast, a PDE4 inhibitor, in abolishing neurodegeneration in the rotenone-induced PD model. Rotenone (1.5 mg/kg, s.c) was delivered via 11 injections on matching days. Roflumilast treatment (0.5 mg/kg, p.o) was given daily after the fifth rotenone injection. Roflumilast significantly reversed rotenone's adverse effects, as it enhanced trophic factors expression and abrogated inflammation as well as oxidative stress. Thus, promoting dopaminergic neuronal plasticity and survival, as well as restoring striatal DA level and function, which resulted in enhanced motor performance. The beneficial effect of roflumilast was mediated through inhibition of striatal PDE4 with consequent activation of cAMP-dependent protein kinase A (PKA) signaling pathways, including the cAMP response element-binding protein (CREB) pathway and dopamine and cAMP-regulated phosphoprotein 32,000 (DARPP-32) pathway that is essential for maintaining dopaminergic function. Therefore, the present work sheds light on the substantial neuroprotective potential of roflumilast in treating PD through the activation of the cAMP/PKA cascade.
Collapse
Affiliation(s)
- Reham M Essam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Department of Biology, School of Pharmacy, Newgiza University, First 6th of October, Giza, 3296121, Egypt.
| | - Esraa A Kandil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
23
|
An Update on Peripheral Blood Extracellular Vesicles as Biomarkers for Parkinson's Disease Diagnosis. Neuroscience 2023; 511:131-146. [PMID: 36435476 DOI: 10.1016/j.neuroscience.2022.11.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/27/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022]
Abstract
Parkinson's disease (PD) is the world's second primary neurodegenerative disease, and the diagnosis and treatment of PD have become mainstream research. Over the past decades, several studies have identified potential biomarkers for diagnosing PD. Among them, extracellular vesicles (EVs) can carry specific biomarkers reflecting the physiological and pathological state of the body. Due to the blood-brain barrier (BBB) limitation, peripheral blood is limited in diagnosing neurodegenerative diseases. With the increasing research on EVs, their ability to pass through BBB indicated that peripheral blood could depict disease status like cerebrospinal fluid (CSF). Peripheral blood is a clinically available sample and has recently been widely used by researchers in various studies. In this review, we summarized previous studies on PD diagnosis biomarkers in peripheral blood EVs and evaluated their diagnostic value. Some EV surface markers were also described, which can extract EVs from specific cell origins. In addition, the combination of several biomarkers demonstrated good diagnostic performance in PD diagnosis compared with a single biomarker, suggesting the focus of future research.
Collapse
|
24
|
The complex role of inflammation and gliotransmitters in Parkinson's disease. Neurobiol Dis 2023; 176:105940. [PMID: 36470499 PMCID: PMC10372760 DOI: 10.1016/j.nbd.2022.105940] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/09/2022] Open
Abstract
Our understanding of the role of innate and adaptive immune cell function in brain health and how it goes awry during aging and neurodegenerative diseases is still in its infancy. Inflammation and immunological dysfunction are common components of Parkinson's disease (PD), both in terms of motor and non-motor components of PD. In recent decades, the antiquated notion that the central nervous system (CNS) in disease states is an immune-privileged organ, has been debunked. The immune landscape in the CNS influences peripheral systems, and peripheral immunological changes can alter the CNS in health and disease. Identifying immune and inflammatory pathways that compromise neuronal health and survival is critical in designing innovative and effective strategies to limit their untoward effects on neuronal health.
Collapse
|
25
|
Coleman C, Martin I. Unraveling Parkinson's Disease Neurodegeneration: Does Aging Hold the Clues? JOURNAL OF PARKINSON'S DISEASE 2022; 12:2321-2338. [PMID: 36278358 PMCID: PMC9837701 DOI: 10.3233/jpd-223363] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Aging is the greatest risk factor for Parkinson's disease (PD), suggesting that mechanisms driving the aging process promote PD neurodegeneration. Several lines of evidence support a role for aging in PD. First, hallmarks of brain aging such as mitochondrial dysfunction and oxidative stress, loss of protein homeostasis, and neuroinflammation are centrally implicated in PD development. Second, mutations that cause monogenic PD are present from conception, yet typically only cause disease following a period of aging. Third, lifespan-extending genetic, dietary, or pharmacological interventions frequently attenuate PD-related neurodegeneration. These observations support a central role for aging in disease development and suggest that new discoveries in the biology of aging could be leveraged to elucidate novel mechanisms of PD pathophysiology. A recent rapid growth in our understanding of conserved molecular pathways that govern model organism lifespan and healthspan has highlighted a key role for metabolism and nutrient sensing pathways. Uncovering how metabolic pathways involving NAD+ consumption, insulin, and mTOR signaling link to the development of PD is underway and implicates metabolism in disease etiology. Here, we assess areas of convergence between nervous system aging and PD, evaluate the link between metabolism, aging, and PD and address the potential of metabolic interventions to slow or halt the onset of PD-related neurodegeneration drawing on evidence from cellular and animal models.
Collapse
Affiliation(s)
- Colin Coleman
- Department of Neurology, Jungers Center for Neurosciences, Oregon Health and Science University, Portland, OR, USA
| | - Ian Martin
- Department of Neurology, Jungers Center for Neurosciences, Oregon Health and Science University, Portland, OR, USA,Correspondence to: Ian Martin, Jungers Center for Neurosciences Research, Department of Neurology - Mail Code L623, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA. Tel.: +1 503 494 9140; E-mail:
| |
Collapse
|
26
|
Xanthotoxin modulates oxidative stress, inflammation, and MAPK signaling in a rotenone-induced Parkinson's disease model. Life Sci 2022; 310:121129. [DOI: 10.1016/j.lfs.2022.121129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/22/2022] [Accepted: 10/23/2022] [Indexed: 11/05/2022]
|
27
|
Zhang L, Li Q. Neuroprotective effects of tanshinone IIA in experimental model of Parkinson disease in rats. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
28
|
Chua SKK, Saffari SE, Lee SJY, Tan EK. Association Between Parkinson's Disease and Coronary Artery Disease: A Systematic Review and Meta-Analysis. JOURNAL OF PARKINSON'S DISEASE 2022; 12:1737-1748. [PMID: 35694936 PMCID: PMC9789484 DOI: 10.3233/jpd-223291] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND The relationship between Parkinson's disease (PD) and coronary artery disease (CAD) is unclear. OBJECTIVE This study aims to investigate whether PD and CAD are associated through systematic review and meta-analysis of observational studies. METHODS Electronic database search of PubMed, EMBASE, and Web of Science for observational studies published from 1 January 2010 to 1 August 2021 was conducted using terms related to PD and CAD. Unadjusted risk ratios (RR) and odds ratios (OR) of included cohort and case-control studies respectively were used to ascertain the association between PD and CAD. Study heterogeneity was evaluated using the I2 test. RESULTS Forty-one full-text studies were initially retrieved for eligibility assessment. Five studies that satisfied the inclusion criteria, consisting of three cohort and two case-control studies, were eventually included in this meta-analysis. The five studies enrolled 35,237 PD patients and 650,866 non-PD patients. PD and CAD were found to be significantly associated in cohort studies (RR = 2.23, 95% CI = 1.08-4.59, p = 0.03; Fig. 2), which held after sensitivity analysis (RR = 1.45, 95% CI = 1.31-1.60, p < 0.001; Fig. 3). Case-control studies found a trend towards association of PD and CAD approaching significance (OR = 1.47, 95% CI = 0.84-2.56, p = 0.18; Fig. 2). CONCLUSION Overall, this meta-analysis suggests that PD is associated with CAD. The underlying mechanisms, as well as the role of ethnicity and other comorbidities on the relationship between PD and CAD should be further explored.
Collapse
Affiliation(s)
- Shaun Kai Kiat Chua
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Seyed Ehsan Saffari
- National Neuroscience Institute, Singapore, Singapore,
Health Services & Systems Research, Duke-NUS Medical School, National University of Singapore, Singapore
| | | | - Eng-King Tan
- National Neuroscience Institute, Singapore, Singapore,Correspondence to: Eng-King Tan, MD, Department of Neurology, National Neuroscience Institute, Duke NUS Medical School, Outram Road, Singapore 169608, Singapore. Tel.: +65 63265003; Fax: +65 62203322; E-mail:
| |
Collapse
|
29
|
Kim S, Kim DK, Jeong S, Lee J. The Common Cellular Events in the Neurodegenerative Diseases and the Associated Role of Endoplasmic Reticulum Stress. Int J Mol Sci 2022; 23:5894. [PMID: 35682574 PMCID: PMC9180188 DOI: 10.3390/ijms23115894] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/17/2022] [Accepted: 05/20/2022] [Indexed: 12/28/2022] Open
Abstract
Neurodegenerative diseases are inseparably linked with aging and increase as life expectancy extends. There are common dysfunctions in various cellular events shared among neurogenerative diseases, such as calcium dyshomeostasis, neuroinflammation, and age-associated decline in the autophagy-lysosome system. However, most of all, the prominent pathological feature of neurodegenerative diseases is the toxic buildup of misfolded protein aggregates and inclusion bodies accompanied by an impairment in proteostasis. Recent studies have suggested a close association between endoplasmic reticulum (ER) stress and neurodegenerative pathology in cellular and animal models as well as in human patients. The contribution of mutant or misfolded protein-triggered ER stress and its associated signaling events, such as unfolded protein response (UPR), to the pathophysiology of various neurodegenerative disorders, including Alzheimer's, Parkinson's, and Huntington's disease, amyotrophic lateral sclerosis, and prion disease, is described here. Impaired UPR action is commonly attributed to exacerbated ER stress, pathogenic protein aggregate accumulation, and deteriorating neurodegenerative pathologies. Thus, activating certain UPR components has been shown to alleviate ER stress and its associated neurodegeneration. However, uncontrolled activation of some UPR factors has also been demonstrated to worsen neurodegenerative phenotypes, suggesting that detailed molecular mechanisms around ER stress and its related neurodegenerations should be understood to develop effective therapeutics against aging-associated neurological syndromes. We also discuss current therapeutic endeavors, such as the development of small molecules that selectively target individual UPR components and address ER stress in general.
Collapse
Affiliation(s)
- Soojeong Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; (S.K.); (D.K.K.); (S.J.)
| | - Doo Kyung Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; (S.K.); (D.K.K.); (S.J.)
| | - Seho Jeong
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; (S.K.); (D.K.K.); (S.J.)
| | - Jaemin Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea; (S.K.); (D.K.K.); (S.J.)
- New Biology Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
- Well Aging Research Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, Korea
| |
Collapse
|
30
|
Rauf A, Badoni H, Abu-Izneid T, Olatunde A, Rahman MM, Painuli S, Semwal P, Wilairatana P, Mubarak MS. Neuroinflammatory Markers: Key Indicators in the Pathology of Neurodegenerative Diseases. Molecules 2022; 27:molecules27103194. [PMID: 35630670 PMCID: PMC9146652 DOI: 10.3390/molecules27103194] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/05/2022] [Accepted: 05/12/2022] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation, a protective response of the central nervous system (CNS), is associated with the pathogenesis of neurodegenerative diseases. The CNS is composed of neurons and glial cells consisting of microglia, oligodendrocytes, and astrocytes. Entry of any foreign pathogen activates the glial cells (astrocytes and microglia) and overactivation of these cells triggers the release of various neuroinflammatory markers (NMs), such as the tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-1β (IL-10), nitric oxide (NO), and cyclooxygenase-2 (COX-2), among others. Various studies have shown the role of neuroinflammatory markers in the occurrence, diagnosis, and treatment of neurodegenerative diseases. These markers also trigger the formation of various other factors responsible for causing several neuronal diseases including Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), multiple sclerosis (MS), ischemia, and several others. This comprehensive review aims to reveal the mechanism of neuroinflammatory markers (NMs), which could cause different neurodegenerative disorders. Important NMs may represent pathophysiologic processes leading to the generation of neurodegenerative diseases. In addition, various molecular alterations related to neurodegenerative diseases are discussed. Identifying these NMs may assist in the early diagnosis and detection of therapeutic targets for treating various neurodegenerative diseases.
Collapse
Affiliation(s)
- Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar 23561, Khyber Pakhtunkhwa, Pakistan
- Correspondence: (A.R.); (P.W.); (M.S.M.)
| | - Himani Badoni
- Department of Biotechnology, School of Applied and Life Sciences, Uttaranchal University, Premnagar, Dehradun 248006, India;
| | - Tareq Abu-Izneid
- Pharmaceutical Sciences Department, College of Pharmacy, Al Ain University for Science and Technology, Al Ain 64141, United Arab Emirates;
| | - Ahmed Olatunde
- Department of Medical Biochemistry, Abubakar Tafawa Balewa University, Bauchi 740272, Nigeria;
| | - Md. Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh;
| | - Sakshi Painuli
- Uttarakhand Council for Biotechnology (UCB), Premnagar, Dehradun 248007, India;
| | - Prabhakar Semwal
- Department of Life Sciences, Graphic Era (Deemed To Be University), Dehradun 248002, India;
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
- Correspondence: (A.R.); (P.W.); (M.S.M.)
| | - Mohammad S. Mubarak
- Department of Chemistry, The University of Jordan, Amman 11942, Jordan
- Correspondence: (A.R.); (P.W.); (M.S.M.)
| |
Collapse
|
31
|
Li D, Hong X, Chen T. Association Between Rheumatoid Arthritis and Risk of Parkinson's Disease: A Meta-Analysis and Systematic Review. Front Neurol 2022; 13:885179. [PMID: 35645965 PMCID: PMC9130734 DOI: 10.3389/fneur.2022.885179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/11/2022] [Indexed: 12/24/2022] Open
Abstract
Background Rheumatoid arthritis (RA) and Parkinson's disease (PD) are two common chronic diseases worldwide, and any potential link between the two would significantly impact public health practice. Considering the current inconsistent evidence, we conducted a meta-analysis and systematic review to examine the risk of PD in patients with RA. Methods Two investigators (DL and XH) conducted a comprehensive search of PubMed, Embase, and Web of Science using medical subject headings terms combined with free words to identify relevant papers published from inception through December 31, 2021. All studies that explored the relationship between RA and PD were included for quantitative analysis and qualitative review. Random- and fixed-effects models were used to pool the risk ratios (RRs) of PD in patients with RA. The Newcastle-Ottawa scale was used to assess the quality of included studies. This study followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA 2020) guideline. Results Four population-based studies involving 353,246 patients and one Mendelian randomized study were included in our study. The pooled result showed a significantly reduced risk of PD in patients with RA than in the general population (RR = 0.74, 95% CI: 0.56-0.98, P = 0.034). No apparent effects of gender, age, region, follow-up time, or study design on PD risk were observed. Sensitivity analysis showed that pooled results were relatively stable, and no publication bias was detected. The Mendelian randomization study indicated a significant inverse association between RA and PD (genetic correlation: −0.10, P = 0.0033) and that each one standard deviation increase in the risk of RA was significantly associated with a lower risk of PD. Of note, the current study is limited by the relatively small number of included studies and unmeasured confounding factors, especially for RA-related anti-inflammatory agents. Conclusions This study supports that people with RA had a lower PD risk than those without RA. Further studies are needed to explore the underlying molecular mechanisms of the interaction between the two diseases.
Collapse
|
32
|
Zhang Y, Chu JMT, Wong GTC. Cerebral Glutamate Regulation and Receptor Changes in Perioperative Neuroinflammation and Cognitive Dysfunction. Biomolecules 2022; 12:biom12040597. [PMID: 35454185 PMCID: PMC9029551 DOI: 10.3390/biom12040597] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 12/23/2022] Open
Abstract
Glutamate is the major excitatory neurotransmitter in the central nervous system and is intricately linked to learning and memory. Its activity depends on the expression of AMPA and NMDA receptors and excitatory amino transporters on neurons and glial cells. Glutamate transporters prevent the excess accumulation of glutamate in synapses, which can lead to aberrant synaptic signaling, excitotoxicity, or cell death. Neuroinflammation can occur acutely after surgical trauma and contributes to the development of perioperative neurocognitive disorders, which are characterized by impairment in multiple cognitive domains. In this review, we aim to examine how glutamate handling and glutamatergic function are affected by neuroinflammation and their contribution to cognitive impairment. We will first summarize the current data regarding glutamate in neurotransmission, its receptors, and their regulation and trafficking. We will then examine the impact of inflammation on glutamate handling and neurotransmission, focusing on changes in glial cells and the effect of cytokines. Finally, we will discuss these changes in the context of perioperative neuroinflammation and the implications they have for perioperative neurocognitive disorders.
Collapse
|
33
|
Neurofunctional characteristics of executive control in older people with HIV infection: a comparison with Parkinson's disease. Brain Imaging Behav 2022; 16:1776-1793. [PMID: 35294979 PMCID: PMC10124990 DOI: 10.1007/s11682-022-00645-6] [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: 01/28/2022] [Indexed: 11/02/2022]
Abstract
Expression of executive dysfunctions is marked by substantial heterogeneity in people living with HIV infection (PLWH) and attributed to neuropathological degradation of frontostriatal circuitry with age and disease. We compared the neurophysiology of executive function in older PLWH and Parkinson's disease (PD), both affecting frontostriatal systems. Thirty-one older PLWH, 35 individuals with PD, and 28 older healthy controls underwent executive task-activated fMRI, neuropsychological testing, and a clinical motor exam. fMRI task conditions distinguished cognitive control operations, invoking a lateral frontoparietal network, and motor control operations, activating a cerebellar-precentral-medial prefrontal network. HIV-specific findings denoted a prominent sensorimotor hypoactivation during cognitive control and striatal hypoactivation during motor control related to CD4+ T cell count and HIV disease duration. Activation deficits overlapped for PLWH and PD, relative to controls, in dorsolateral frontal, medial frontal, and middle cingulate cortices for cognitive control, and in limbic, frontal, parietal, and cerebellar regions for motor control. Thus, despite well-controlled HIV infection, frontostriatal and sensorimotor activation deficits occurred during executive control in older PLWH. Overlapping activation deficits in posterior cingulate and hippocampal regions point toward similarities in mesocorticolimbic system aberrations among older PLWH and PD. The extent of pathophysiology in PLWH was associated with variations in immune system health, neural signature consistent with subclinical parkinsonism, and mild neurocognitive impairment. The failure to adequately engage these pathways could be an early sign for cognitive and motor functional decline in the aging population of PLWH.
Collapse
|
34
|
Weiss F, Labrador-Garrido A, Dzamko N, Halliday G. Immune responses in the Parkrtdinson's disease brain. Neurobiol Dis 2022; 168:105700. [DOI: 10.1016/j.nbd.2022.105700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 12/15/2022] Open
|
35
|
Zhao Y, Zhang X, Guo N, Tian D, Zhang C, Mu C, Han C, Zhu R, Zhang J, Liu X. Genetically Predicted Levels of Circulating Inflammatory Cytokines and the Risk and Age at Onset of Parkinson’s Disease: A Two-Sample Mendelian Randomization Study. Front Aging Neurosci 2022; 14:811059. [PMID: 35299944 PMCID: PMC8923644 DOI: 10.3389/fnagi.2022.811059] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/31/2022] [Indexed: 12/14/2022] Open
Abstract
Parkinson’s disease (PD) is widely considered to be a disabling neurodegenerative disorder, which has been ranked second worldwide just after Alzheimer’s disease. Until present, a wide range of studies has focused on the role of circulating inflammatory cytokines in the development of PD. However, the causal relationship between circulating inflammatory cytokines and the risk and age at the onset of PD has not been elucidated. Hence, to evaluate the effects of circulating inflammatory cytokines on the risk or age at the onset of PD more accurately, we conducted this two-sample Mendelian randomization (MR) study involving summary statistics from genome-wide association studies (GWASs). Totally, we included a GWAS for inflammatory cytokines (8,293 participants), a meta-analysis of GWASs for PD risk (482,730 participants), and a GWAS dataset for age at the onset of PD (17,996 patients with PD). A total of 149 and 131 polymorphisms for exploring relationships between 19 inflammatory cytokines and the risk and age at the onset of PD were obtained as instrumental variants. Then, we used a total of five MR methods, including inverse-variance weighted (IVW), Wald ratio, MR Egger regression, weighted median, and MR-pleiotropy residual sum and outlier (MR-PRESSO) methods. Finally, we found a causal association between circulating levels of macrophage inflammatory protein-1 beta (MIP1b) and PD risk in the IVW method (OR: 1.06; 95% CI: 1.02–1.10; P = 0.001). Meanwhile, other MR estimates by weighted median and MR-PRESSO methods yielded similar effect estimates. Besides, we identified a suggestive association of interleukin-16 (IL-16) levels with PD risk (OR: 1.08; 95% CI: 1.00–1.17; P = 0.037). For age at PD onset, there was no evidence supporting its correlation with inflammatory cytokines. Our findings implied that MIP1b and IL-16 may be novel biomarkers and promising therapeutic targets for PD development.
Collapse
Affiliation(s)
- Yating Zhao
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiaoqian Zhang
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Na Guo
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Dandan Tian
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Chenguang Zhang
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Changqing Mu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Chen Han
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ruixia Zhu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jian Zhang
- Key Laboratory of Cell Biology, Ministry of Public Health, Department of Cell Biology, China Medical University, Shenyang, China
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Xu Liu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Xu Liu,
| |
Collapse
|
36
|
Liu Z, Chan RB, Cai Z, Liu X, Wu Y, Yu Z, Feng T, Yang Y, Zhang J. α-Synuclein-containing erythrocytic extracellular vesicles: essential contributors to hyperactivation of monocytes in Parkinson's disease. J Neuroinflammation 2022; 19:53. [PMID: 35193594 PMCID: PMC8862590 DOI: 10.1186/s12974-022-02413-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 02/09/2022] [Indexed: 01/06/2023] Open
Abstract
Background Immune system dysfunction, including higher levels of peripheral monocytes and inflammatory cytokines, is an important feature of Parkinson’s disease (PD) pathogenesis, although the mechanism underlying the process remains to be investigated. In the central nervous system, it is well-known that α-synuclein (α-syn), a key protein involved in PD, activates microglia potently, and it is also reported that α-syn exists in the peripheral system, especially in erythrocytes or red blood cells (RBC) at exceedingly high concentration. The current study focused on the possibility that RBC-derived α-syn mediates the sensitization of peripheral monocytes in PD patients. Methods The hyperactivation of monocytes was assessed quantitatively by measuring mRNA levels of typical inflammatory cytokines (including IL-1β, IL-6 and TNF-α) and protein levels of secreted inflammatory cytokines (including pro-inflammatory cytokines: IL-1β, IL-6, TNF-α, IL-8, IFN-γ, IL-2, and IL-12p70 and anti-inflammatory cytokines: IL-4, IL-10, and IL-13). Western blot, nanoparticle tracking analysis and electron microscopy were used to characterize RBC-derived extracellular vesicles (RBC-EVs). Inhibitors of endocytosis and leucine-rich repeat kinase 2 (LRRK2), another key protein involved in PD, were used to investigate how these two factors mediated the process of monocyte sensitization by RBC-EVs. Results Increased inflammatory sensitization of monocytes was observed in PD patients and PD model mice. We found that α-syn-containing RBC-EVs isolated from PD model mice or free form oligomeric α-syn induced the inflammatory sensitization of THP-1 cells, and demonstrated that endocytosis was a requirement for this pathophysiological pathway. Furthermore, the hyperactivation of THP-1 cells induced by RBC-EVs was associated with increased LRRK2 production and kinase activity. The phenomenon of inflammatory sensitization of human monocytes and increased LRRK2 were also observed by the treatment of RBC-EVs isolated from PD patients. Conclusions Our data provided new insight into how hyperactivation of monocytes occurs in PD patients, and identified the central role played by α-syn-containing RBC-EVs in this process. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02413-1.
Collapse
Affiliation(s)
- Zongran Liu
- Department of Pathology, Peking University Health Science Center, Beijing, 100191, China
| | | | - Zhijian Cai
- School of Basic Medicine, Zhejiang University, Hangzhou, 310002, Zhejiang, China
| | - Xiaodan Liu
- Department of Pathology, Peking University Health Science Center, Beijing, 100191, China
| | - Yufeng Wu
- Department of Laboratory Medicine, Peking University Third Hospital, Peking University Health Science Center, Beijing, China
| | - Zhenwei Yu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, 100050, China
| | - Tao Feng
- Department of Neurology, TianTan Hospital, Capital Medical University, Beijing, 100050, China
| | - Ying Yang
- Department of Pathology, Peking University Health Science Center, Beijing, 100191, China.
| | - Jing Zhang
- Department of Pathology, Zhejiang University School of Medicine and First Affiliated Hospital, Hangzhou, 310002, Zhejiang, China. .,National Health and Disease Human Brain Tissue Resource Center, Zhejiang University, Hangzhou, 310002, Zhejiang, China.
| |
Collapse
|
37
|
Li KL, Huang HY, Ren H, Yang XL. Role of exosomes in the pathogenesis of inflammation in Parkinson's disease. Neural Regen Res 2022; 17:1898-1906. [PMID: 35142665 PMCID: PMC8848593 DOI: 10.4103/1673-5374.335143] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Inflammatory responses, including glial cell activation and peripheral immune cell infiltration, are involved in the pathogenesis of Parkinson’s disease (PD). These inflammatory responses appear to be closely related to the release of extracellular vesicles, such as exosomes. However, the relationships among different forms of glial cell activation, synuclein dysregulation, mitochondrial dysfunction, and exosomes are complicated. This review discusses the multiple roles played by exosomes in PD-associated inflammation and concludes that exosomes can transport toxic α-synuclein oligomers to immature neurons and into the extracellular environment, inducing the oligomerization of α-synuclein in normal neurons. Misfolded α-synuclein causes microglia and astrocytes to activate and secrete exosomes. Glial cell-derived exosomes participate in communications between glial cells and neurons, triggering anti-stress and anti-inflammatory responses, in addition to axon growth. The production and release of mitochondrial vesicles and exosomes establish a new mechanism for linking mitochondrial dysfunction to systemic inflammation associated with PD. Given the relevance of exosomes as mediators of neuron-glia communication in neuroinflammation and neuropathogenesis, new targeted treatment strategies are currently being developed that use these types of extracellular vesicles as drug carriers. Exosome-mediated inflammation may be a promising target for intervention in PD patients.
Collapse
Affiliation(s)
- Ke-Lu Li
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Hong-Yan Huang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hui Ren
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| | - Xing-Long Yang
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China
| |
Collapse
|
38
|
Neuroinflammation and Neuropathology. NEUROSCIENCE AND BEHAVIORAL PHYSIOLOGY 2022; 52:196-201. [PMID: 35317271 PMCID: PMC8930459 DOI: 10.1007/s11055-022-01223-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/18/2021] [Indexed: 11/25/2022]
Abstract
This review addresses the current understanding of the role of autoimmune neuroinflammation in the pathogenesis of vascular, neurodegenerative, and other diseases of the nervous system. The mechanisms of responses of resident CNS cells (glial cells, astrocytes) and peripheral immune system cells are presented. The therapeutic potentials of phosphodiesterase inhibitors, which have antiaggregant properties and can suppress autoimmune inflammation, are discussed. The phosphodiesterase inhibitor dipyridamole is regarded as a potential drug for this purpose.
Collapse
|
39
|
Palmas MF, Ena A, Burgaletto C, Casu MA, Cantarella G, Carboni E, Etzi M, De Simone A, Fusco G, Cardia MC, Lai F, Picci L, Tweedie D, Scerba MT, Coroneo V, Bernardini R, Greig NH, Pisanu A, Carta AR. Repurposing Pomalidomide as a Neuroprotective Drug: Efficacy in an Alpha-Synuclein-Based Model of Parkinson's Disease. Neurotherapeutics 2022; 19:305-324. [PMID: 35072912 PMCID: PMC9130415 DOI: 10.1007/s13311-022-01182-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2021] [Indexed: 12/17/2022] Open
Abstract
Marketed drugs for Parkinson's disease (PD) treat disease motor symptoms but are ineffective in stopping or slowing disease progression. In the quest of novel pharmacological approaches that may target disease progression, drug-repurposing provides a strategy to accelerate the preclinical and clinical testing of drugs already approved for other medical indications. Here, we targeted the inflammatory component of PD pathology, by testing for the first time the disease-modifying properties of the immunomodulatory imide drug (IMiD) pomalidomide in a translational rat model of PD neuropathology based on the intranigral bilateral infusion of toxic preformed oligomers of human α-synuclein (H-αSynOs). The neuroprotective effect of pomalidomide (20 mg/kg; i.p. three times/week 48 h apart) was tested in the first stage of disease progression by means of a chronic two-month administration, starting 1 month after H-αSynOs infusion, when an already ongoing neuroinflammation is observed. The intracerebral infusion of H-αSynOs induced an impairment in motor and coordination performance that was fully rescued by pomalidomide, as assessed via a battery of motor tests three months after infusion. Moreover, H-αSynOs-infused rats displayed a 40-45% cell loss within the bilateral substantia nigra, as measured by stereological counting of TH + and Nissl-stained neurons, that was largely abolished by pomalidomide. The inflammatory response to H-αSynOs infusion and the pomalidomide treatment was evaluated both in CNS affected areas and peripherally in the serum. A reactive microgliosis, measured as the volume occupied by the microglial marker Iba-1, was present in the substantia nigra three months after H-αSynOs infusion as well as after H-αSynOs plus pomalidomide treatment. However, microglia differed for their phenotype among experimental groups. After H-αSynOs infusion, microglia displayed a proinflammatory profile, producing a large amount of the proinflammatory cytokine TNF-α. In contrast, pomalidomide inhibited the TNF-α overproduction and elevated the anti-inflammatory cytokine IL-10. Moreover, the H-αSynOs infusion induced a systemic inflammation with overproduction of serum proinflammatory cytokines and chemokines, that was largely mitigated by pomalidomide. Results provide evidence of the disease modifying potential of pomalidomide in a neuropathological rodent model of PD and support the repurposing of this drug for clinical testing in PD patients.
Collapse
Affiliation(s)
| | - Anna Ena
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Chiara Burgaletto
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Giuseppina Cantarella
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Ezio Carboni
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Michela Etzi
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Alfonso De Simone
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Giuliana Fusco
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Maria Cristina Cardia
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Francesco Lai
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Luca Picci
- Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Michael T Scerba
- Drug Design & Development Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Valentina Coroneo
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, Baltimore, MD, USA
| | - Augusta Pisanu
- National Research Council, Institute of Neuroscience, Cagliari, Italy.
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy.
| |
Collapse
|
40
|
Kara SP, Altunan B, Unal A. Investigation of the peripheral inflammation (neutrophil-lymphocyte ratio) in two neurodegenerative diseases of the central nervous system. Neurol Sci 2022; 43:1799-1807. [PMID: 34331157 PMCID: PMC8324446 DOI: 10.1007/s10072-021-05507-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/18/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD), and idiopathic Parkinson's disease (IPD) are the neurodegenerative diseases of the central nervous system (CNS). Cognitive impairment is on the forefront in AD. However, IPD is a movement disorder. Inflammation was suggested to have an effect in the pathophysiology of these two diseases. Neutrophil-lymphocyte ratio (NLR) was shown to be a possible marker showing the peripheral inflammation. We aimed to investigate the NLR of patiens with the diagnosis of AD, and IPD, and individuals with no neurodegenerative disease. MATERIALS AND METHODS A total of 100 patients with the diagnosis of IPD, and 94 with diagnosis of AD, and 61 healthy controls were included into the study. All the demographic, clinical, and laboratory data were retrospectively obtained from the hospital automated database system. RESULTS The NLR in the IPD group was found statistically significantly higher compared with the control group and the AD group (p < 0.001, p = 0.04, respectively). The age-adjusted values were statistically analyzed because of age difference. No statistically significant difference was detected between AD and control groups in terms of NLR (p = 0.6). The age-adjusted NLR value in the Parkinson's group was found significantly higher compared to the control group (p = 0.02) and Alzheimer's group (p = 0.03). DISCUSSION Chronic inflammation has an important role in the emergence and progression of the chronic neurodegenerative diseases of the CNS. Our results show that the inflammation in the peripheral blood in IPD was more significant compared with the inflammation in AD.
Collapse
Affiliation(s)
- Sonat Pınar Kara
- Faculty of Medicine, Department of Internal Medicine, Tekirdag Namık Kemal University, Tekirdag, Turkey
| | - Bengü Altunan
- Faculty of Medicine, Department of Neurology, Tekirdag Namik Kemal University, Tekirdag, Turkey
| | - Aysun Unal
- Faculty of Medicine, Department of Neurology, Tekirdag Namik Kemal University, Tekirdag, Turkey
| |
Collapse
|
41
|
Forero-Rodríguez LJ, Josephs-Spaulding J, Flor S, Pinzón A, Kaleta C. Parkinson's Disease and the Metal-Microbiome-Gut-Brain Axis: A Systems Toxicology Approach. Antioxidants (Basel) 2021; 11:71. [PMID: 35052575 PMCID: PMC8773335 DOI: 10.3390/antiox11010071] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/02/2021] [Accepted: 12/17/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson's Disease (PD) is a neurodegenerative disease, leading to motor and non-motor complications. Autonomic alterations, including gastrointestinal symptoms, precede motor defects and act as early warning signs. Chronic exposure to dietary, environmental heavy metals impacts the gastrointestinal system and host-associated microbiome, eventually affecting the central nervous system. The correlation between dysbiosis and PD suggests a functional and bidirectional communication between the gut and the brain. The bioaccumulation of metals promotes stress mechanisms by increasing reactive oxygen species, likely altering the bidirectional gut-brain link. To better understand the differing molecular mechanisms underlying PD, integrative modeling approaches are necessary to connect multifactorial perturbations in this heterogeneous disorder. By exploring the effects of gut microbiota modulation on dietary heavy metal exposure in relation to PD onset, the modification of the host-associated microbiome to mitigate neurological stress may be a future treatment option against neurodegeneration through bioremediation. The progressive movement towards a systems toxicology framework for precision medicine can uncover molecular mechanisms underlying PD onset such as metal regulation and microbial community interactions by developing predictive models to better understand PD etiology to identify options for novel treatments and beyond. Several methodologies recently addressed the complexity of this interaction from different perspectives; however, to date, a comprehensive review of these approaches is still lacking. Therefore, our main aim through this manuscript is to fill this gap in the scientific literature by reviewing recently published papers to address the surrounding questions regarding the underlying molecular mechanisms between metals, microbiota, and the gut-brain-axis, as well as the regulation of this system to prevent neurodegeneration.
Collapse
Affiliation(s)
- Lady Johanna Forero-Rodríguez
- Research Group Bioinformatics and Systems Biology, Instituto de Genetica, Universidad Nacional de Colombia, Bogotá 111321, Colombia; (L.J.F.-R.); (A.P.)
- Research Group Medical Systems Biology, Christian-Albrechts-Universität Kiel, Brunswiker Straße 10, 24105 Kiel, Germany; (S.F.); (C.K.)
| | - Jonathan Josephs-Spaulding
- Research Group Medical Systems Biology, Christian-Albrechts-Universität Kiel, Brunswiker Straße 10, 24105 Kiel, Germany; (S.F.); (C.K.)
| | - Stefano Flor
- Research Group Medical Systems Biology, Christian-Albrechts-Universität Kiel, Brunswiker Straße 10, 24105 Kiel, Germany; (S.F.); (C.K.)
| | - Andrés Pinzón
- Research Group Bioinformatics and Systems Biology, Instituto de Genetica, Universidad Nacional de Colombia, Bogotá 111321, Colombia; (L.J.F.-R.); (A.P.)
| | - Christoph Kaleta
- Research Group Medical Systems Biology, Christian-Albrechts-Universität Kiel, Brunswiker Straße 10, 24105 Kiel, Germany; (S.F.); (C.K.)
| |
Collapse
|
42
|
Yuan M, Wang Y, Wang S, Huang Z, Jin F, Zou Q, Li J, Pu Y, Cai Z. Bioenergetic Impairment in the Neuro-Glia-Vascular Unit: An Emerging Physiopathology during Aging. Aging Dis 2021; 12:2080-2095. [PMID: 34881087 PMCID: PMC8612602 DOI: 10.14336/ad.2021.04017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/17/2021] [Indexed: 12/28/2022] Open
Abstract
An emerging concept termed the "neuro-glia-vascular unit" (NGVU) has been established in recent years to understand the complicated mechanism of multicellular interactions among vascular cells, glial cells, and neurons. It has been proverbially reported that the NGVU is significantly associated with neurodegenerative disorders, such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Physiological aging is an inevitable progression associated with oxidative damage, bioenergetic alterations, mitochondrial dysfunction, and neuroinflammation, which is partially similar to the pathology of AD. Thus, senescence is regarded as the background for the development of neurodegenerative diseases. With the exacerbation of global aging, senescence is an increasingly serious problem in the medical field. In this review, the coupling of each component, including neurons, glial cells, and vascular cells, in the NGVU is described in detail. Then, various mechanisms of age-dependent impairment in each part of the NGVU are discussed. Moreover, the potential bioenergetic alterations between different cell types in the NGVU are highlighted, which seems to be an emerging physiopathology associated with the aged brain. Bioenergetic intervention in the NGVU may be a new direction for studies on delaying or diminishing aging in the future.
Collapse
Affiliation(s)
- Minghao Yuan
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China.,4Chongqing Medical University, Chongqing, China
| | - Yangyang Wang
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Shengyuan Wang
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China.,4Chongqing Medical University, Chongqing, China
| | - Zhenting Huang
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Feng Jin
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Qian Zou
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Jing Li
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Yinshuang Pu
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China
| | - Zhiyou Cai
- 1Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 400013, Chongqing, China.,2Chongqing School, University of Chinese Academy of Sciences, Chongqing, China.,3Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, 400013, Chongqing, China.,4Chongqing Medical University, Chongqing, China
| |
Collapse
|
43
|
Reduced Immunosenescence of Peripheral Blood T Cells in Parkinson's Disease with CMV Infection Background. Int J Mol Sci 2021; 22:ijms222313119. [PMID: 34884936 PMCID: PMC8658620 DOI: 10.3390/ijms222313119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/25/2021] [Accepted: 12/01/2021] [Indexed: 01/11/2023] Open
Abstract
Immunosenescence is a process of remodeling the immune system under the influence of chronic inflammation during aging. Parkinson’s disease (PD) is a common age-associated neurodegenerative disorder and is frequently accompanied by neuroinflammation. On the other hand, cytomegalovirus (CMV), one of the most spread infections in humans, may induce chronic inflammation which contributes to immunosenescence, differentiation and the inflation of T cells and NK cells. Currently, there is no clear understanding of immunosenescence severity in PD patients infected with CMV. In this study, we analyzed differentiation stages and immunosenescence characteristics of T cells and NK cells in 31 patients with mild and moderate PD severity, 33 age-matched and 30 young healthy donors. The PD patients were 100% CMV-seropositive compared to 76% age-matched and 73% young CMV-infected healthy donors. The proportion of effector memory T cells re-expressing CD45RA, CD57+CD56− T cells and CD57+CD56+ T cells was significantly reduced in PD patients compared with CMV-seropositive age-matched healthy individuals. The CD57+CD56− T cell proportion in PD patients was similar to that of CMV-seropositive young healthy donors. Thus, PD is characterized by reduced peripheral blood T cell immunosenescence, even against the background of CMV infection.
Collapse
|
44
|
The science of matcha: Bioactive compounds, analytical techniques and biological properties. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.10.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
45
|
Kuter KZ, Śmiałowska M, Ossowska K. The influence of preconditioning with low dose of LPS on paraquat-induced neurotoxicity, microglia activation and expression of α-synuclein and synphilin-1 in the dopaminergic system. Pharmacol Rep 2021; 74:67-83. [PMID: 34762280 PMCID: PMC8786770 DOI: 10.1007/s43440-021-00340-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 12/21/2022]
Abstract
Background Prolonged inflammation, oxidative stress, and protein aggregation are important factors contributing to Parkinson’s disease (PD) pathology. A known ROS generator, pesticide paraquat (PQ), was indicated as an environmental substance potentially increasing the incidence of PD and is used to model this disease. We investigated if a combination of inflammation and oxidative stress in subthreshold doses would exacerbate the modelled neuropathology. Methods We examined the late effects of acute or repeated peripheral inflammation induced by low dose of LPS (10 μg/kg, ip) on PQ toxicity in the rat nigrostriatal dopaminergic pathway, microglial activation markers and expression of major Lewy bodies proteins, α-synuclein and synphilin-1. Results We observed that LPS increased, while PQ decreased body temperature and microglia CD11b expression in the SN. Single LPS pretreatment, 3 h before repeated weekly PQ injections (4×) slightly aggravated neuronal degeneration in the SN. Moreover, degeneration of dopaminergic neurons after weekly repeated inflammation itself (4×) was observed. Interestingly, repeated LPS administration combined with each PQ dose counteracted such effect. The expression of α-synuclein decreased after repeated LPS injections, while only combined, repeated LPS and PQ treatment lowered the levels of synphilin-1. Therefore, α-synuclein and synphilin-1 expression change was influenced by different mechanisms. Concomitantly, decreased levels of the two proteins correlated with decreased degeneration of dopaminergic neurons and with a normalized microglia activation marker. Conclusions Our results indicate that both oxidative insult triggered by PQ and inflammation caused by peripheral LPS injection can individually induce neurotoxicity. Those factors act through different mechanisms that are not additive and not selective towards dopaminergic neurons, probably implying microglia. Repeated, but small insults from oxidative stress and inflammation when administered in significant time intervals can counteract each other and even act protective as a preconditioning effect. The timing of such repetitive insults is also of essence. Supplementary Information The online version contains supplementary material available at 10.1007/s43440-021-00340-1.
Collapse
Affiliation(s)
- Katarzyna Z Kuter
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St., 31-343, Kraków, Poland.
| | - Maria Śmiałowska
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Krystyna Ossowska
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St., 31-343, Kraków, Poland
| |
Collapse
|
46
|
Badanjak K, Mulica P, Smajic S, Delcambre S, Tranchevent LC, Diederich N, Rauen T, Schwamborn JC, Glaab E, Cowley SA, Antony PMA, Pereira SL, Venegas C, Grünewald A. iPSC-Derived Microglia as a Model to Study Inflammation in Idiopathic Parkinson's Disease. Front Cell Dev Biol 2021; 9:740758. [PMID: 34805149 PMCID: PMC8602578 DOI: 10.3389/fcell.2021.740758] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease with unknown cause in the majority of patients, who are therefore considered "idiopathic" (IPD). PD predominantly affects dopaminergic neurons in the substantia nigra pars compacta (SNpc), yet the pathology is not limited to this cell type. Advancing age is considered the main risk factor for the development of IPD and greatly influences the function of microglia, the immune cells of the brain. With increasing age, microglia become dysfunctional and release pro-inflammatory factors into the extracellular space, which promote neuronal cell death. Accordingly, neuroinflammation has also been described as a feature of PD. So far, studies exploring inflammatory pathways in IPD patient samples have primarily focused on blood-derived immune cells or brain sections, but rarely investigated patient microglia in vitro. Accordingly, we decided to explore the contribution of microglia to IPD in a comparative manner using, both, iPSC-derived cultures and postmortem tissue. Our meta-analysis of published RNAseq datasets indicated an upregulation of IL10 and IL1B in nigral tissue from IPD patients. We observed increased expression levels of these cytokines in microglia compared to neurons using our single-cell midbrain atlas. Moreover, IL10 and IL1B were upregulated in IPD compared to control microglia. Next, to validate these findings in vitro, we generated IPD patient microglia from iPSCs using an established differentiation protocol. IPD microglia were more readily primed as indicated by elevated IL1B and IL10 gene expression and higher mRNA and protein levels of NLRP3 after LPS treatment. In addition, IPD microglia had higher phagocytic capacity under basal conditions-a phenotype that was further exacerbated upon stimulation with LPS, suggesting an aberrant microglial function. Our results demonstrate the significance of microglia as the key player in the neuroinflammation process in IPD. While our study highlights the importance of microglia-mediated inflammatory signaling in IPD, further investigations will be needed to explore particular disease mechanisms in these cells.
Collapse
Affiliation(s)
- Katja Badanjak
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Patrycja Mulica
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Semra Smajic
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Sylvie Delcambre
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | | | - Nico Diederich
- Centre Hospitalier de Luxembourg (CHL), Luxembourg, Luxembourg
| | - Thomas Rauen
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Jens C. Schwamborn
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Sally A. Cowley
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Paul M. A. Antony
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
- Disease Modeling and Screening Platform (DMSP), Luxembourg Institute of Systems Biomedicine, University of Luxembourg and Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Sandro L. Pereira
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Carmen Venegas
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
| | - Anne Grünewald
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Luxembourg, Luxembourg
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| |
Collapse
|
47
|
Chipurupalli S, Samavedam U, Robinson N. Crosstalk Between ER Stress, Autophagy and Inflammation. Front Med (Lausanne) 2021; 8:758311. [PMID: 34805224 PMCID: PMC8602556 DOI: 10.3389/fmed.2021.758311] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/14/2021] [Indexed: 12/23/2022] Open
Abstract
The endoplasmic reticulum (ER) is not only responsible for protein synthesis and folding but also plays a critical role in sensing cellular stress and maintaining cellular homeostasis. Upon sensing the accumulation of unfolded proteins due to perturbation in protein synthesis or folding, specific intracellular signaling pathways are activated, which are collectively termed as unfolded protein response (UPR). UPR expands the capacity of the protein folding machinery, decreases protein synthesis and enhances ER-associated protein degradation (ERAD) which degrades misfolded proteins through the proteasomes. More recent evidences suggest that UPR also amplifies cytokines-mediated inflammatory responses leading to pathogenesis of inflammatory diseases. UPR signaling also activates autophagy; a lysosome-dependent degradative pathwaythat has an extended capacity to degrade misfolded proteins and damaged ER. Thus, activation of autophagy limits inflammatory response and provides cyto-protection by attenuating ER-stress. Here we review the mechanisms that couple UPR, autophagy and cytokine-induced inflammation that can facilitate the development of novel therapeutic strategies to mitigate cellular stress and inflammation associated with various pathologies.
Collapse
Affiliation(s)
- Sandhya Chipurupalli
- Cellular-Stress and Immune Response Laboratory, Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| | - Unni Samavedam
- College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Nirmal Robinson
- Cellular-Stress and Immune Response Laboratory, Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| |
Collapse
|
48
|
Li B, Yang S, Ye J, Chu S, Chen N, An Z. Flavin-containing monooxygenase 1 deficiency promotes neuroinflammation in dopaminergic neurons in mice. Neurosci Lett 2021; 764:136222. [PMID: 34500002 DOI: 10.1016/j.neulet.2021.136222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 08/09/2021] [Accepted: 09/02/2021] [Indexed: 10/20/2022]
Abstract
A growing body of evidence indicates an association between flavin-containing monooxygenase (FMO) and neurodegeneration, including Parkinson's disease (PD); however, the details of this association are unclear. We previously showed that the level of Fmo1 mRNA is decreased in an in vitro rotenone model of parkinsonism. To further explore the potential involvement of FMO1 deficiency in parkinsonism, we generated Fmo1 knockout (KO) mice and examined the survival of dopaminergic neurons and relative changes. Fmo1 KO mice exhibited loss of tyrosine hydroxylase-positive neurons, decreased levels of tyrosine hydroxylase and Parkin proteins, and increased levels of pro-inflammatory cytokines (IL1β and IL6) in the nigrostriatal region. Moreover, the protein levels of PTEN induced kinase 1 (PINK1) and p62, and the Microtubule associated protein 1 light chain 3 (LC3)-II/I ratio were not significantly altered in Fmo1 KO mice (P > 0.05). FMO1 deficiency promotes neuroinflammation in dopaminergic neurons in mice, thus may plays a potential pathological role in dopaminergic neuronal loss. These findings may provide new insight into the pathogenesis of PD.
Collapse
Affiliation(s)
- Boyu Li
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongtinan Road, Beijing 100020, China
| | - Song Yang
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongtinan Road, Beijing 100020, China
| | - Junrui Ye
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Jia2 nanwei Road, Beijing 100050, China
| | - Shifeng Chu
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Jia2 nanwei Road, Beijing 100050, China
| | - Naihong Chen
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Jia2 nanwei Road, Beijing 100050, China.
| | - Zhuoling An
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongtinan Road, Beijing 100020, China.
| |
Collapse
|
49
|
Butt MS, Tariq U, Iahtisham-Ul-Haq, Naz A, Rizwan M. Neuroprotective effects of oleuropein: Recent developments and contemporary research. J Food Biochem 2021; 45:e13967. [PMID: 34716610 DOI: 10.1111/jfbc.13967] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 12/16/2022]
Abstract
Neurological disorders are increasing at a faster pace due to oxidative stress, protein aggregation, excitotoxicity, and neuroinflammation. It is reported that the Mediterranean diet including olives as a major dietary component prevents and ameliorates neurological anomalies. Oleuropein is the major bioactive component in different parts of the Olive (Olea europaea L.) tree. Several mechanisms have been reported for the neuroprotective role of oleuropein including induction of apoptosis and autophagy, enhancing the antioxidant pool of the cerebral region, decreasing the unnecessary release of proinflammatory cytokines and chemokines by deactivating the microglia cells and astrocytes thus preventing the occurrence of neuroinflammation. Regular intake of oleuropein seems to be correlated with decreased risks of neural disorders including Alzheimer's, Parkinson's, strokes, depression, anxiety, epilepsy, and others. This review majorly discusses the chemistry, biosynthesis, and metabolism of oleuropein along with an updated vision of its neuroprotective role in counteracting the acute and chronic neurodegenerative and neuropsychiatric disorders. Moreover, mechanisms by which oleuropein may prevent neurodegeneration are reviewed. PRACTICAL APPLICATION: Neurological disorders are negatively affecting the health and life quality of individuals around the globe. Although various medicinal solutions are available to tackle such ailments, none has proven to fully cure and being deprived of side effects. In this respect, the prevention of such disorders using natural remedies may be an effective strategy to overcome the incidence of the increasing cases. Furthermore, the natural compounds provide a safer alternative to pharmaceutical drugs. Hence, oleuropein from olive tree products is found to be efficacious against neurological disorders. This review provides an updated insight on the positive effects of oleuropein against neurodegenerative and neuropsychiatric disorders. The diet practitioners and nutraceutical companies may benefit from the provided information to design and develop strategies to improve the mental health of suffering individuals.
Collapse
Affiliation(s)
- Masood Sadiq Butt
- Faculty of Food, Nutrition and Home Sciences, National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| | - Urwa Tariq
- Faculty of Food, Nutrition and Home Sciences, National Institute of Food Science and Technology, University of Agriculture, Faisalabad, Pakistan
| | - Iahtisham-Ul-Haq
- Faculty of Life Sciences, Department of Food Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Ambreen Naz
- Department of Food Science and Technology, Muhammad Nawaz Shareef University of Agriculture, Multan, Pakistan
| | - Muhammad Rizwan
- Faculty of Life Sciences, Department of Food Science and Technology, University of Central Punjab, Lahore, Pakistan
| |
Collapse
|
50
|
Wang XL, Li L. Circadian Clock Regulates Inflammation and the Development of Neurodegeneration. Front Cell Infect Microbiol 2021; 11:696554. [PMID: 34595127 PMCID: PMC8476957 DOI: 10.3389/fcimb.2021.696554] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022] Open
Abstract
The circadian clock regulates numerous key physiological processes and maintains cellular, tissue, and systemic homeostasis. Disruption of circadian clock machinery influences key activities involved in immune response and brain function. Moreover, Immune activation has been closely linked to neurodegeneration. Here, we review the molecular clock machinery and the diurnal variation of immune activity. We summarize the circadian control of immunity in both central and peripheral immune cells, as well as the circadian regulation of brain cells that are implicated in neurodegeneration. We explore the important role of systemic inflammation on neurodegeneration. The circadian clock modulates cellular metabolism, which could be a mechanism underlying circadian control. We also discuss the circadian interventions implicated in inflammation and neurodegeneration. Targeting circadian clocks could be a potential strategy for the prevention and treatment of inflammation and neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiao-Lan Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lianjian Li
- Department of Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|