1
|
Mahnoor, Jahan S, Elahi L, Zakria M, Rabia, Ikram M, Ullah N. Alpha-Linolenic Acid for Mitigating Neuroinflammation and Dopaminergic Neuronal Loss in Parkinson's Disease: Insights From In Vivo and In Silico Studies. Clin Exp Pharmacol Physiol 2025; 52:e70043. [PMID: 40269665 DOI: 10.1111/1440-1681.70043] [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: 01/19/2025] [Revised: 03/05/2025] [Accepted: 04/07/2025] [Indexed: 04/25/2025]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterised by dopaminergic neuronal loss and chronic neuroinflammation, leading to significant motor and non-motor deficits. This study explores the therapeutic potential of alpha-linolenic acid (ALA), a known antioxidant and anti-inflammatory agent, in a lipopolysaccharide (LPS)-induced murine model of PD. Male Balb-C mice were divided into control, LPS-treated, LPS + ALA-treated and ALA-only groups. Behavioural assessments, including the pole test, rotarod test and open field test, revealed significant motor impairments in LPS-treated mice. Co-treatment with ALA partially ameliorated motor deficits in LPS-treated mice compared to the healthy control group. However, no direct comparison was made with standard PD treatments such as levodopa. Immunohistochemistry analysis showed a 68% reduction in tyrosine hydroxylase-positive (TH+) neurons in the substantia nigra pars compacta (SNpc) of LPS-treated mice. Notably, ALA co-treatment preserved dopaminergic neurons, demonstrating its neuroprotective effects. Western blotting and ELISA revealed heightened expression of inflammatory mediators, including TNF-α, IL-1β and NF-κB, in LPS-treated mice. ALA treatment significantly reduced these markers, indicating its capacity to mitigate neuroinflammation. Molecular docking analysis revealed moderate binding affinities of ALA to NF-κB (-5.1 kcal/mol), TNF-α (-5.7 kcal/mol) and IL-1β (-3.9 kcal/mol), suggesting possible interactions with key inflammatory pathways. These interactions were comparable to known inhibitors, indicating ALA's potential for neuroprotection. This study highlights the neuroprotective and anti-inflammatory effects of ALA in reducing dopaminergic neuronal loss and mitigating neuroinflammation in an LPS-induced PD model. Although behavioural improvements were moderate, these findings underscore ALA's potential as an adjunct therapeutic candidate for PD and other neurodegenerative diseases. Further research is warranted to explore its translational applications in clinical settings.
Collapse
Affiliation(s)
- Mahnoor
- Institute of Basic Medical Sciences (IBMS), Khyber Medical University, Peshawar, Pakistan
| | - Sarwat Jahan
- Institute of Pharmaceutical Sciences (IPS), Khyber Medical University, Peshawar, Pakistan
- Department of Pharmacology, Northwest School of Medicine, Hayatabad, Pakistan
| | - Laila Elahi
- Institute of Basic Medical Sciences (IBMS), Khyber Medical University, Peshawar, Pakistan
| | - Muhammad Zakria
- Institute of Pharmaceutical Sciences (IPS), Khyber Medical University, Peshawar, Pakistan
| | - Rabia
- Institute of Basic Medical Sciences (IBMS), Khyber Medical University, Peshawar, Pakistan
| | - Muhammad Ikram
- Institute of Pharmaceutical Sciences (IPS), Khyber Medical University, Peshawar, Pakistan
- Division of Life Science and Applied Life Science (BK 21), College of Natural Science, Gyeongsang National University, Jinju, Republic of Korea
| | - Najeeb Ullah
- Institute of Basic Medical Sciences (IBMS), Khyber Medical University, Peshawar, Pakistan
| |
Collapse
|
2
|
Bellini G, D'Antongiovanni V, Palermo G, Antonioli L, Fornai M, Ceravolo R, Bernardini N, Derkinderen P, Pellegrini C. α-Synuclein in Parkinson's Disease: From Bench to Bedside. Med Res Rev 2025; 45:909-946. [PMID: 39704040 PMCID: PMC11976381 DOI: 10.1002/med.22091] [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: 05/12/2024] [Revised: 09/24/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024]
Abstract
α-Synuclein (α-syn), a pathological hallmark of PD, is emerging as a bridging element at the crossroads between neuro/immune-inflammatory responses and neurodegeneration in PD. Several evidence show that pathological α-syn accumulates in neuronal and non-neuronal cells (i.e., neurons, microglia, macrophages, skin cells, and intestinal cells) in central and peripheral tissues since the prodromal phase of the disease, contributing to brain pathology. Indeed, pathological α-syn deposition can promote neurogenic/immune-inflammatory responses that contribute to systemic and central neuroinflammation associated with PD. After providing an overview of the structure and functions of physiological α-syn as well as its pathological forms, we review current studies about the role of neuronal and non-neuronal α-syn at the crossroads between neuroinflammation and neurodegeneration in PD. In addition, we provide an overview of the correlation between the accumulation of α-syn in central and peripheral tissues and PD, related symptoms, and neuroinflammation. Special attention was paid to discussing whether targeting α-syn can represent a suitable therapeutical approach for PD.
Collapse
Affiliation(s)
- Gabriele Bellini
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
- Department of NeurologyThe Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, NYU Langone HealthNew York CityNew YorkUSA
| | - Vanessa D'Antongiovanni
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Giovanni Palermo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Roberto Ceravolo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Nunzia Bernardini
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Pascal Derkinderen
- Department of NeurologyNantes Université, CHU Nantes, INSERMNantesFrance
| | - Carolina Pellegrini
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| |
Collapse
|
3
|
Hu R, Shi G, Wu C, Jiao Y, Li Y, Qi A, Li Y, Zhang Q, Liu Q, Wang K, Huang M. Paraquat exposure triggers amyloid-β and α-synuclein aggregation in the prefrontal cortex of mice: Suppression of microglial phagocytosis via IL-17A. Int Immunopharmacol 2025; 157:114746. [PMID: 40300355 DOI: 10.1016/j.intimp.2025.114746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/30/2025] [Accepted: 04/24/2025] [Indexed: 05/01/2025]
Abstract
Paraquat (PQ), an environmental neurotoxin, has been demonstrated to induce pathological protein aggregation and thus neurotoxicity. Nevertheless, the exact mechanisms remain elusive. In this investigation, we explored the involvement of interleukin-17A (IL-17 A) in the aggregation of amyloid-β (Aβ) and α-synuclein (α-syn) induced by PQ. Combining in vitro and in vivo, we explored whether PQ leads to Aβ and α-syn aggregation through IL-17 A-mediated reduction in microglia phagocytosis, thereby aggravating neurotoxicity. The results demonstrated that low-dose PQ continuous exposure significantly elevated IL-17 A levels in the peripheral blood serum and prefrontal cortical regions of mice. It also suppressed microglial phagocytosis of pathological proteins and promoted the aggregation of Aβ and α-syn in the prefrontal cortex. These changes ultimately resulted in depression, anxiety, and cognitive impairments. Mechanistically, IL-17 A inhibited the expression of the microglial phagocytic receptor CD36, impairing the microglial ability to clear Aβ and α-syn. Furthermore, administering an anti-IL-17 A effectively restored microglial phagocytosis in PQ-exposed mice, reduced Aβ and α-syn aggregation in prefrontal cortical areas, and alleviated behavioral deficits. In conclusion, this paper highlights IL-17 A as a pivotal mediator in PQ-induced neurotoxicity. It provides a potential target for developing novel therapeutic strategies against neurodegenerative pathologies induced by such environmental toxicants.
Collapse
Affiliation(s)
- Rong Hu
- School of Public Health, Ningxia Medical University, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, the Street of Shengli, Xingqing District, Yinchuan, Ningxia 750004, China
| | - Ge Shi
- School of Public Health, Ningxia Medical University, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, the Street of Shengli, Xingqing District, Yinchuan, Ningxia 750004, China
| | - Chenyang Wu
- School of Public Health, Ningxia Medical University, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, the Street of Shengli, Xingqing District, Yinchuan, Ningxia 750004, China
| | - Yuxuan Jiao
- School of Public Health, Ningxia Medical University, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, the Street of Shengli, Xingqing District, Yinchuan, Ningxia 750004, China
| | - Yonghang Li
- School of Public Health, Ningxia Medical University, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, the Street of Shengli, Xingqing District, Yinchuan, Ningxia 750004, China
| | - Ai Qi
- School of Public Health, Ningxia Medical University, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, the Street of Shengli, Xingqing District, Yinchuan, Ningxia 750004, China
| | - Yujing Li
- School of Public Health, Ningxia Medical University, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, the Street of Shengli, Xingqing District, Yinchuan, Ningxia 750004, China
| | - Qianrong Zhang
- School of Public Health, Ningxia Medical University, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, the Street of Shengli, Xingqing District, Yinchuan, Ningxia 750004, China
| | - Qi Liu
- School of Public Health, Ningxia Medical University, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, the Street of Shengli, Xingqing District, Yinchuan, Ningxia 750004, China
| | - Kaidong Wang
- School of Public Health, Ningxia Medical University, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, the Street of Shengli, Xingqing District, Yinchuan, Ningxia 750004, China
| | - Min Huang
- School of Public Health, Ningxia Medical University, Ningxia 750004, China; Key Laboratory of Environmental Factors and Chronic Disease Control, No.1160, the Street of Shengli, Xingqing District, Yinchuan, Ningxia 750004, China.
| |
Collapse
|
4
|
Zhou KG, Huang YB, Zhu ZW, Jiang M, Jin LJ, Guan Q, Tian LL, Zhang JX. Mesencephalic astrocyte-derived neurotrophic factor inhibits neuroinflammation through autophagy-mediated α-synuclein degradation. Arch Gerontol Geriatr 2025; 131:105738. [PMID: 39761611 DOI: 10.1016/j.archger.2024.105738] [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: 10/07/2024] [Revised: 12/14/2024] [Accepted: 12/30/2024] [Indexed: 02/25/2025]
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder marked by the progressive loss of dopamine neurons in the substantia nigra. α-synuclein (SNCA) aggregation-induced microglia activation and neuroinflammation play vital role in the pathology of PD. Our previous studies showed that mesencephalic astrocyte-derived neurotrophic factor (MANF) could inhibit SNCA accumulation and Lipopolysaccharides (LPS)-induced neuroinflammation, but the specific molecular mechanism remains unclear. In this study, we showed that knock-down the expression of MANF leads to the up-regulation of inflammatory factor tumor necrosis factor-α (TNF-α). Exogenous MANF protein inhibits LPS-induced neuroinflammation in BV2 cells. Additionally, our results indicated that knock-down of the expression of MANF triggered autophagic pathway dysfunction, while exogenous addition of MANF protein or using adeno-associated virus 8 (AAV8) mediated MANF over-expression could activate the autophagic system and subsequently suppress SNCA accumulation. Furthermore, using autophagy inhibitor to block autophagic flux, we found that MANF prevented neuroinflammation by autophagy-mediated SNCA degradation. Collectively, this study indicated that MANF has potential therapeutic value for PD. Autophagy and its role in MANF-mediated anti-inflammatory properties may provide new sights that target SNCA pathology in PD.
Collapse
Affiliation(s)
- Kai-Ge Zhou
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Yi-Bin Huang
- School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Zi-Wen Zhu
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Ming Jiang
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China; Biomedical Research Center, Tongji University Suzhou Institute, Jiangsu, 215101, China
| | - Ling-Jing Jin
- Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qiang Guan
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Lu-Lu Tian
- Department of Pharmacy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Jing-Xing Zhang
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| |
Collapse
|
5
|
Armanville S, Tocco C, Haj Mohamad Z, Clarke D, Robitaille R, Drouin-Ouellet J. Chemically Induced Senescence Prompts Functional Changes in Human Microglia-Like Cells. J Immunol Res 2025; 2025:3214633. [PMID: 40041406 PMCID: PMC11876530 DOI: 10.1155/jimr/3214633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
In response to various stressors, cells can enter a state called cellular senescence which is characterized by irreversible cell cycle arrest and a senescence-associated secretory phenotype (SASP). The progressive accumulation of senescent glial cells in the central nervous system (CNS) with aging suggests a potential role for senescence as driver of aging and inflammation in the brain. As the main immune cell population residing in the CNS, microglia are thought to play a pivotal role in the progression of age-associated neuroinflammation. Furthermore, due to their slow turnover, microglia are highly susceptible to undergoing cellular senescence. However, current understanding of age-related changes in microglia and their impact on brain aging is limited. Due to the challenge in accessing human primary microglia and the lack of models to adequately recapitulate aging, this knowledge is predominantly limited to rodent studies. Here, we chemically induced senescence in a human immortalized microglia cell line with a cocktail of senescence-inducing molecules. We demonstrate that chemically induced senescent microglia adopt a proinflammatory phenotype, have reduced phagocytic activity, and impaired calcium activity. Our results show that chemically induced senescence can mimic features of cellular aging and can provide insight into the impact of aging and cellular senescence on human microglia.
Collapse
Affiliation(s)
- S. Armanville
- Faculty of Pharmacy, University of Montreal, Montreal, Quebec, Canada
| | - C. Tocco
- Faculty of Pharmacy, University of Montreal, Montreal, Quebec, Canada
| | - Z. Haj Mohamad
- Faculty of Pharmacy, University of Montreal, Montreal, Quebec, Canada
| | - D. Clarke
- Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada
| | - R. Robitaille
- Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada
- Research Group on Neural Signalling and Circuits (SNC), University of Montreal, Montreal, Quebec, Canada
- Center for Interdisciplinary Research on Brain and Learning (CIRCA), Montreal, Quebec, Canada
| | - J. Drouin-Ouellet
- Faculty of Pharmacy, University of Montreal, Montreal, Quebec, Canada
- Research Group on Neural Signalling and Circuits (SNC), University of Montreal, Montreal, Quebec, Canada
- Center for Interdisciplinary Research on Brain and Learning (CIRCA), Montreal, Quebec, Canada
| |
Collapse
|
6
|
Sarallah R, Jahani S, Soltani Khaboushan A, Moaveni AK, Amiri M, Majidi Zolbin M. The role of CXCL12/CXCR4/CXCR7 axis in cognitive impairment associated with neurodegenerative diseases. Brain Behav Immun Health 2025; 43:100932. [PMID: 39834554 PMCID: PMC11743895 DOI: 10.1016/j.bbih.2024.100932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 12/03/2024] [Accepted: 12/21/2024] [Indexed: 01/22/2025] Open
Abstract
Neurodegenerative diseases, including Alzheimer's Disease (AD), Parkinson's Disease (PD), Multiple Sclerosis (MS), and Amyotrophic Lateral Sclerosis (ALS), are characterized by progressive neuronal loss and cognitive impairment (CI). The: Cysteine-X-cysteine chemokine ligand 12(CXCL12)/CXC chemokine receptor type 4 (CXCR4)/CXC chemokine receptor type 7 (CXCR7) axis has emerged as a critical molecular pathway in the development of CI in these disorders. This review explores the role of this axis in the pathogenesis of CI across these neurodegenerative diseases, synthesizing current evidence and its implications for targeted therapies. In AD, dysregulation of this axis contributes to amyloid-β accumulation and tau hyperphosphorylation, leading to synaptic dysfunction and cognitive decline. PD studies reveal that CXCL12/CXCR4 signaling influences dopaminergic neuron survival and microglial activation, affecting cognitive function. In MS, the axis modulates neuroinflammation and demyelination processes, impacting cognitive performance. ALS research indicates that the CXCL12/CXCR4/CXCR7 pathway is involved in motor neuron degeneration and associated cognitive deficits. Across these diseases, the axis influences neuroinflammation, synaptic plasticity, and neuronal survival through various signaling cascades, including PI3K/AKT, MAPK, and JAK/STAT pathways. Emerging evidence suggests that modulating this axis could provide neuroprotective effects and potentially alleviate cognitive symptoms. This review highlights the potential of the CXCL12/CXCR4/CXCR7 axis as a therapeutic target for addressing CI in neurodegenerative diseases. It also underscores the need for further research to fully elucidate its role and develop effective interventions, potentially leading to improved clinical management strategies for these devastating disorders.
Collapse
Affiliation(s)
| | - Shima Jahani
- MS Research Center Neuroscience Institute, Tehran University of Medical Science, Iran
| | - Alireza Soltani Khaboushan
- Pediatric and Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children Medical Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Kian Moaveni
- Pediatric and Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children Medical Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Amiri
- Pediatric and Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children Medical Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Majidi Zolbin
- Pediatric and Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children Medical Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Abhilash PL, Bharti U, Rashmi SK, Philip M, Raju TR, Kutty BM, Sagar BKC, Alladi PA. Aging and MPTP Sensitivity Depend on Molecular and Ultrastructural Signatures of Astroglia and Microglia in Mice Substantia Nigra. Cell Mol Neurobiol 2025; 45:13. [PMID: 39833644 PMCID: PMC11753320 DOI: 10.1007/s10571-024-01528-8] [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: 10/27/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025]
Abstract
Both astroglia and microglia show region-specific distribution in CNS and often maladapt to age-associated alterations within their niche. Studies on autopsied substantia nigra (SN) of Parkinson's disease (PD) patients and experimental models propose gliosis as a trigger for neuronal loss. Epidemiological studies propose an ethnic bias in PD prevalence, since Caucasians are more susceptible than non-whites. Similarly, different mice strains are variably sensitive to MPTP. We had earlier likened divergent MPTP sensitivity of C57BL/6 J and CD-1 mice with differential susceptibility to PD, based on the numbers of SN neurons. We examined whether the variability was incumbent to inter-strain differences in glial features of male C57BL/6 J and CD-1 mice. Stereological counts showed relatively more microglia and fewer astrocytes in the SN of normal C57BL/6 J mice, suggesting persistence of an immune-vigilant state. MPTP-induced microgliosis and astrogliosis in both strains suggest their involvement in pathogenesis. ELISA of pro-inflammatory cytokines in the ventral-midbrain revealed augmentation of TNF-α and IL-6 at middle age in both strains that reduced at old age, suggesting middle age as a critical, inflamm-aging-associated time point. TNF-α levels were high in C57BL/6 J, through aging and post-MPTP, while IL-6 and IL-1β were upregulated at old age. CD-1 had higher levels of anti-inflammatory cytokine TGF-β. MPTP challenge caused upregulation of enzymes MAO-A, MAO-B, and iNOS in both strains. Post-MPTP enhancement in fractalkine and hemeoxygenase-1 may be neuron-associated compensatory signals. Ultrastructural observations of elongated astroglial/microglial mitochondria vis-à-vis the shrunken ones in neurons suggest a scale-up of their functions with neurotoxic consequences. Thus, astroglia and microglia may modulate aging and PD susceptibility.
Collapse
Affiliation(s)
- P L Abhilash
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India
| | - Upasna Bharti
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - Santhosh Kumar Rashmi
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - Mariamma Philip
- Department of Biostatistics, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - T R Raju
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India
| | - Bindu M Kutty
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India
| | - B K Chandrasekhar Sagar
- Department of Biostatistics, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - Phalguni Anand Alladi
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India.
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India.
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neuro Sciences, Hosur Road, Bangalore, India.
| |
Collapse
|
8
|
Zhou M, Xu K, Ge J, Luo X, Wu M, Wang N, Zeng J. Targeting Ferroptosis in Parkinson's Disease: Mechanisms and Emerging Therapeutic Strategies. Int J Mol Sci 2024; 25:13042. [PMID: 39684753 DOI: 10.3390/ijms252313042] [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: 10/24/2024] [Revised: 11/30/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the loss of dopaminergic neurons in the substantia nigra and the accumulation of α-synuclein in the brain. Ferroptosis, a recently identified form of regulated cell death, is critical in PD pathogenesis due to its association with iron deposition, overproduction of reactive oxygen species, iron-dependent lipid peroxidation and impaired lipid peroxidation clearance. This cell death mechanism is closely linked to several pathogenic processes in PD, including α-synuclein aggregation, oxidative stress, mitochondrial dysfunction, microglia-induced neuroinflammation, and neuromelanin accumulation. Given the significant role of ferroptosis in these mechanisms, there is increasing interest in targeting ferroptosis for PD treatment. Several drugs have shown potential in alleviating PD symptoms by inhibiting ferroptosis. This review aims to consolidate current knowledge on ferroptosis in PD and assess the therapeutic potential of anti-ferroptosis drugs, highlighting promising directions for future research and clinical applications.
Collapse
Affiliation(s)
- Minghao Zhou
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Keyang Xu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jianxian Ge
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Xingnian Luo
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Mengyao Wu
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Ning Wang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jianfeng Zeng
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| |
Collapse
|
9
|
Fang K. Modulation of the central nervous system immune response and neuroinflammation via Wnt signaling in health and neurodegenerative diseases. IBRAIN 2024; 10:462-476. [PMID: 39691422 PMCID: PMC11649390 DOI: 10.1002/ibra.12185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 12/19/2024]
Abstract
The immune response in the central nervous system (CNS) is a highly specialized and tightly regulated process essential for maintaining neural health and protecting against pathogens and injuries. The primary immune cells within the CNS include microglia, astrocytes, T cells, and B cells. They work together, continuously monitor the CNS environment for signs of infection, injury, or disease, and respond by phagocytosing debris, releasing cytokines, and recruiting other immune cells. In addition to providing neuroprotection, these immune responses must be carefully balanced to prevent excessive inflammation that can lead to neuronal damage and contribute to neurodegenerative diseases. Dysregulated immune responses in the CNS are implicated in various neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Wnt signaling is a crucial pathway in the CNS that regulates various cellular processes critical for brain development, function, and maintenance. Despite enhancing immune responses in the health CNS, dysregulated Wnt signaling exacerbates neuroinflammation in the neurodegenerative brains. This review summarized the role of Wnt signaling in regulating immune response under different conditions. We then examined the role of immune response in healthy brains and during the development of neurodegenerative diseases. We also discussed therapeutic intervention in various neurodegenerative diseases through the modulation of the Wnt signaling pathway and neuroinflammation and highlighted challenges and limitations in current clinical trials.
Collapse
Affiliation(s)
- Kevin Fang
- Living Systems InstituteUniversity ExeterExeterUK
| |
Collapse
|
10
|
Parekh P, Serra M, Allaw M, Perra M, Pinna A, Manconi M, Morelli M. Extract from Nasco pomace loaded in nutriosomes exerts anti-inflammatory effects in the MPTP mouse model of Parkinson's disease. Exp Neurol 2024; 382:114958. [PMID: 39303846 DOI: 10.1016/j.expneurol.2024.114958] [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: 05/08/2024] [Revised: 08/24/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Neuroinflammation has recently emerged as a key event in Parkinson's disease (PD) pathophysiology and as a potential target for disease-modifying therapies. Plant-derived extracts, rich in bioactive phytochemicals with antioxidant properties, have shown potential in this regard. Yet their clinical utility is hampered by poor systemic availability and rapid metabolism. Recently, our group demonstrated that intragastric delivery of Nasco pomace extract via nutriosomes (NN), a novel nanoliposome formulation, contrasts the degeneration of nigrostriatal dopaminergic neurons in a subacute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. In the present study, we investigated the impact of intragastric NN treatment on the reactivity of glial cells in the substantia nigra pars compacta (SNc) and caudate-putamen (CPu) of MPTP-treated mice. To this scope, in mice exposed to MPTP (20 mg/kg/day, × 4 days), we conducted immunohistochemistry analyses of glial fibrillary acidic protein (GFAP) and ionized calcium-binding adapter molecule 1 (IBA1) to assess the responsiveness of astrocytes and microglial cells, respectively. Additionally, we studied the co-localization of the pro-inflammatory interleukin (IL)-1β and tumor necrosis factor (TNF)-α with IBA1 to obtain insights into microglial phenotype. Immunohistochemical results showed that NN administration significantly mitigated astrogliosis and microgliosis in the CPu and SNc of mice receiving subacute MPTP treatment, with region-specific variations in anti-inflammatory efficacy. Remarkably, the CPu showed a heightened response to NN treatment, including a pronounced decrease in microglial IL-1β and TNF-α production. Altogether, these findings underscore the anti-inflammatory effects of NN treatment and provide a potential mechanism underlying the neuroprotective effects previously observed in a subacute MPTP mouse model of PD.
Collapse
Affiliation(s)
- Pathik Parekh
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy; Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Marcello Serra
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy.
| | - Mohamad Allaw
- Department of Life and Environmental Sciences, University of Cagliari, Italy
| | - Matteo Perra
- Department of Life and Environmental Sciences, University of Cagliari, Italy
| | - Annalisa Pinna
- National Research Council of Italy, Institute of Neuroscience, Cagliari, Italy
| | - Maria Manconi
- Department of Life and Environmental Sciences, University of Cagliari, Italy.
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cagliari, Italy; National Research Council of Italy, Institute of Neuroscience, Cagliari, Italy
| |
Collapse
|
11
|
Sian-Hulsmann J, Riederer P. Virus-induced brain pathology and the neuroinflammation-inflammation continuum: the neurochemists view. J Neural Transm (Vienna) 2024; 131:1429-1453. [PMID: 38261034 PMCID: PMC11608394 DOI: 10.1007/s00702-023-02723-5] [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: 09/21/2023] [Accepted: 11/18/2023] [Indexed: 01/24/2024]
Abstract
Fascinatingly, an abundance of recent studies has subscribed to the importance of cytotoxic immune mechanisms that appear to increase the risk/trigger for many progressive neurodegenerative disorders, including Parkinson's disease (PD), Alzheimer's disease (AD), amyotrophic lateral sclerosis, and multiple sclerosis. Events associated with the neuroinflammatory cascades, such as ageing, immunologic dysfunction, and eventually disruption of the blood-brain barrier and the "cytokine storm", appear to be orchestrated mainly through the activation of microglial cells and communication with the neurons. The inflammatory processes prompt cellular protein dyshomeostasis. Parkinson's and Alzheimer's disease share a common feature marked by characteristic pathological hallmarks of abnormal neuronal protein accumulation. These Lewy bodies contain misfolded α-synuclein aggregates in PD or in the case of AD, they are Aβ deposits and tau-containing neurofibrillary tangles. Subsequently, these abnormal protein aggregates further elicit neurotoxic processes and events which contribute to the onset of neurodegeneration and to its progression including aggravation of neuroinflammation. However, there is a caveat for exclusively linking neuroinflammation with neurodegeneration, since it's highly unlikely that immune dysregulation is the only factor that contributes to the manifestation of many of these neurodegenerative disorders. It is unquestionably a complex interaction with other factors such as genetics, age, and environment. This endorses the "multiple hit hypothesis". Consequently, if the host has a genetic susceptibility coupled to an age-related weakened immune system, this makes them more susceptible to the virus/bacteria-related infection. This may trigger the onset of chronic cytotoxic neuroinflammatory processes leading to protein dyshomeostasis and accumulation, and finally, these events lead to neuronal destruction. Here, we differentiate "neuroinflammation" and "inflammation" with regard to the involvement of the blood-brain barrier, which seems to be intact in the case of neuroinflammation but defect in the case of inflammation. There is a neuroinflammation-inflammation continuum with regard to virus-induced brain affection. Therefore, we propose a staging of this process, which might be further developed by adding blood- and CSF parameters, their stage-dependent composition and stage-dependent severeness grade. If so, this might be suitable to optimise therapeutic strategies to fight brain neuroinflammation in its beginning and avoid inflammation at all.
Collapse
Affiliation(s)
- Jeswinder Sian-Hulsmann
- Department of Human Anatomy and Medical Physiology, University of Nairobi, P.O. Box 30197, Nairobi, 00100, Kenya
| | - Peter Riederer
- University Hospital Wuerzburg, Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy Margarete-Höppel-Platz 1, 97080, Würzburg, Germany.
- Department of Psychiatry, University of Southern Denmark, Winslows Vey 18, 5000, Odense, J.B, Denmark.
| |
Collapse
|
12
|
Harackiewicz O, Grembecka B. The Role of Microglia and Astrocytes in the Pathomechanism of Neuroinflammation in Parkinson's Disease-Focus on Alpha-Synuclein. J Integr Neurosci 2024; 23:203. [PMID: 39613467 DOI: 10.31083/j.jin2311203] [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: 04/29/2024] [Revised: 07/04/2024] [Accepted: 07/31/2024] [Indexed: 12/01/2024] Open
Abstract
Glial cells, including astrocytes and microglia, are pivotal in maintaining central nervous system (CNS) homeostasis and responding to pathological insults. This review elucidates the complex immunomodulatory functions of glial cells, with a particular focus on their involvement in inflammation cascades initiated by the accumulation of alpha-synuclein (α-syn), a hallmark of Parkinson's disease (PD). Deriving insights from studies on both sporadic and familial forms of PD, as well as animal models of PD, we explore how glial cells contribute to the progression of inflammation triggered by α-syn aggregation. Additionally, we analyze the interplay between glial cells and the blood-brain barrier (BBB), highlighting the role of these cells in maintaining BBB integrity and permeability in the context of PD pathology. Furthermore, we delve into the potential activation of repair and neuroprotective mechanisms mediated by glial cells amidst α-syn-induced neuroinflammation. By integrating information on sporadic and familial PD, as well as BBB dynamics, this review aims to deepen our understanding of the multifaceted interactions between glial cells, α-syn pathology, and CNS inflammation, thereby offering valuable insights into therapeutic strategies for PD and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Oliwia Harackiewicz
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdańsk, 80-308 Gdańsk, Poland
| | - Beata Grembecka
- Department of Animal and Human Physiology, Faculty of Biology, University of Gdańsk, 80-308 Gdańsk, Poland
| |
Collapse
|
13
|
Verma A, Goyal A. Beyond insulin: The Intriguing role of GLP-1 in Parkinson's disease. Eur J Pharmacol 2024; 982:176936. [PMID: 39182542 DOI: 10.1016/j.ejphar.2024.176936] [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: 04/09/2024] [Revised: 08/12/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
GLP-1 (Glucagon-like peptide 1) serves as both a peptide hormone and a growth factor, is released upon nutrient intake and contributes to insulin secretion stimulated by glucose levels. Also, GLP-1 is synthesized within several brain areas and plays a vital function in providing neuroprotection and reducing inflammation through the activation of the GLP-1 receptor. Parkinson's Disease (PD) is a neurodegenerative illness that worsens with time and is defined by considerable morbidity. Presently, there are few pharmaceutical choices available, and none of the existing therapies are capable of modifying the course of the disease. There is a suggestion that type 2 diabetes mellitus (T2DM) could increase the risk of PD, and the presence of both conditions concurrently might exacerbate PD symptoms and hasten neurodegeneration. GLP-1 receptor (GLP-1R) agonists exhibit numerous implications like enhancement of glucose-dependent insulin release and biosynthesis, suppression of glucagon secretion and gastric emptying. Also, some GLP-1R agonists have received clinical approval for the management of T2DM. Moreover, the use of GLP-1R agonists has demonstrated counter-inflammatory, neurotrophic, and neuroprotective actions in various preclinical models of neurodegenerative disorders. Considering the significant amount of evidence backing the potential of GLP-1R agonists to protect the nervous system across different research settings, this article delves into examining the hopeful prospect of GLP-1R agonists as a treatment option for PD. This review sheds light on combined neuroprotective benefits of GLP-1R agonists and the possible mechanisms driving the protective effects on the PD brain, through the collection of data from various preclinical and clinical investigations.
Collapse
Affiliation(s)
- Aanchal Verma
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India.
| |
Collapse
|
14
|
Hahn KR, Kwon HJ, Kim DW, Hwang IK, Yoon YS. Therapeutic Options of Crystallin Mu and Protein Disulfide Isomerase A3 for Cuprizone-Induced Demyelination in Mouse Hippocampus. Neurochem Res 2024; 49:3078-3093. [PMID: 39164609 PMCID: PMC11449959 DOI: 10.1007/s11064-024-04227-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024]
Abstract
This study investigates the changes in hippocampal proteomic profiles during demyelination and remyelination using the cuprizone model. Employing two-dimensional gel electrophoresis and liquid chromatography-tandem mass spectrometry for protein profiling, we observed significant alterations in the expression of ketimine reductase mu-crystallin (CRYM) and protein disulfide isomerase A3 precursor (PDIA3) following exposure to and subsequent withdrawal from cuprizone. Immunohistochemical staining validated these protein expression patterns in the hippocampus, revealing that both PDIA3 and CRYM were downregulated in the hippocampal CA1 region during demyelination and upregulated during remyelination. Additionally, we explored the potential protective effects of CRYM and PDIA3 against cuprizone-induced demyelination by synthesizing cell-permeable Tat peptide-fusion proteins (Tat-CRYM and Tat-PDIA3) to facilitate their crossing through the blood-brain barrier. Our results indicated that administering Tat-CRYM and Tat-PDIA3 mitigated the reduction in proliferating cell and differentiated neuroblast counts compared to the group receiving cuprizone alone. Notably, Tat-PDIA3 demonstrated significant effects in enhancing myelin basic protein expression alongside phosphorylation of CREB in the hippocampus, suggesting its potential therapeutic role in the prevention or treatment of demyelination, and by extension, in conditions such as multiple sclerosis.
Collapse
Affiliation(s)
- Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
- Department of Biomedical Sciences, and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
15
|
Tang H, Lu Y, Okun MS, Donahoo WT, Ramirez-Zamora A, Wang F, Huang Y, Armstrong M, Svensson M, Virnig BA, DeKosky ST, Bian J, Guo J. Glucagon-Like Peptide-1 Receptor Agonists and Risk of Parkinson's Disease in Patients with Type 2 Diabetes: A Population-Based Cohort Study. Mov Disord 2024; 39:1960-1970. [PMID: 39189078 PMCID: PMC11568939 DOI: 10.1002/mds.29992] [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: 04/29/2024] [Revised: 07/31/2024] [Accepted: 08/07/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND Previous studies have suggested that glucagon-like peptide-1 receptor agonists (GLP-1RAs) may have a disease-modifying effect in the development of Parkinson's disease (PD), but population studies yielded inconsistent results. OBJECTIVE The aim was to compare the risk of PD associated with GLP-1RAs compared to dipeptidyl peptidase 4 inhibitors (DPP4i) among older adults with type 2 diabetes (T2D). METHODS Using U.S. Medicare administrative data from 2016 to 2020, we conducted a population-based cohort study comparing the new use of GLP-1RA with the new use of DPP4i among adults aged ≥66 years with T2D. The primary endpoint was a new diagnosis of PD. A stabilized inverse probability of treatment weighting (sIPTW)-adjusted Cox proportional hazards regression model was employed to estimate the hazard ratio (HR) and 95% confidence intervals (CI) for PD between GLP-1RA and DPP4i users. RESULTS This study included 89,074 Medicare beneficiaries who initiated either GLP-1RA (n = 30,091) or DPP4i (n = 58,983). The crude incidence rate of PD was lower among GLP-1RA users than DPP4i users (2.85 vs. 3.92 patients per 1000 person-years). An sIPTW-adjusted Cox model showed that GLP-1RA users were associated with a 23% lower risk of PD than DPP4i users (HR, 0.77; 95% CI, 0.63-0.95). Our findings were largely consistent across different subgroup analyses such as sex, race, and molecular structure of GLP-1RA. CONCLUSION Among Medicare beneficiaries with T2D, the new use of GLP-1RAs was significantly associated with a decreased risk of PD compared to the new use of DPP4i. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Huilin Tang
- Department of Pharmaceutical Outcomes and Policy, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | - Ying Lu
- Department of Pharmaceutical Outcomes and Policy, University of Florida College of Pharmacy, Gainesville, Florida, USA
| | - Michael S Okun
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, Florida, USA
| | - William T Donahoo
- Division of Endocrinology, Diabetes and Metabolism, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Adolfo Ramirez-Zamora
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, New York, USA
| | - Yu Huang
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Melissa Armstrong
- Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Mikael Svensson
- Department of Pharmaceutical Outcomes and Policy, University of Florida College of Pharmacy, Gainesville, Florida, USA
- Center for Drug Evaluation and Safety, University of Florida, Gainesville, Florida, USA
| | - Beth A Virnig
- College of Public Health and Health Professions Dean’s Office, University of Florida, Gainesville, Florida, USA
| | - Steven T. DeKosky
- Department of Neurology and McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, Florida USA
- 1Florida Alzheimer’s Disease Research Center (ADRC), University of Florida, Gainesville, Florida, USA
| | - Jiang Bian
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Jingchuan Guo
- Department of Pharmaceutical Outcomes and Policy, University of Florida College of Pharmacy, Gainesville, Florida, USA
- Center for Drug Evaluation and Safety, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
16
|
Wang MY, Zhou Y, Li WL, Zhu LQ, Liu D. Friend or foe: Lactate in neurodegenerative diseases. Ageing Res Rev 2024; 101:102452. [PMID: 39127445 DOI: 10.1016/j.arr.2024.102452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
Lactate, a byproduct of glycolysis, was considered as a metabolic waste until identified by studies on the Warburg effect. Increasing evidence elucidates that lactate functions as energy fuel, signaling molecule, and donor for protein lactylation. Altered lactate utilization is a common metabolic feature of the onset and progression of neurodegenerative diseases, such as Alzheimer's disease, multiple sclerosis, amyotrophic lateral sclerosis, Parkinson's disease and Huntington's disease. This review offers an overview of lactate metabolism from the perspective of production, transportation and clearance, and the role of lactate in neurodegenerative progression, as well as a summary of protein lactylation and the signaling function of lactate in neurodegenerative diseases. Besides, this review delves into the dual roles of changed lactate metabolism during neurodegeneration and explores prospective therapeutic methods targeting lactate. We propose that elucidating the correlation between lactate and neurodegeneration is pivotal for exploring innovative therapeutic interventions for neurodegenerative diseases.
Collapse
Affiliation(s)
- Ming-Yu Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Zhou
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Wen-Lian Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Dan Liu
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| |
Collapse
|
17
|
Yu SJ, Wang Y, Shen H, Bae EK, Li Y, Sambamurti K, Tones MA, Zaleska MM, Hoffer BJ, Greig NH. DPP-4 inhibitors sitagliptin and PF-00734,200 mitigate dopaminergic neurodegeneration, neuroinflammation and behavioral impairment in the rat 6-OHDA model of Parkinson's disease. GeroScience 2024; 46:4349-4371. [PMID: 38563864 PMCID: PMC11336009 DOI: 10.1007/s11357-024-01116-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Epidemiological studies report an elevated risk of Parkinson's disease (PD) in patients with type 2 diabetes mellitus (T2DM) that is mitigated in those prescribed dipeptidyl peptidase 4 (DPP-4) inhibitors. With an objective to characterize clinically translatable doses of DPP-4 inhibitors (gliptins) in a well-characterized PD rodent model, sitagliptin, PF-00734,200 or vehicle were orally administered to rats initiated either 7-days before or 7-days after unilateral medial forebrain bundle 6-hydroxydopamine (6-OHDA) lesioning. Measures of dopaminergic cell viability, dopamine content, neuroinflammation and neurogenesis were evaluated thereafter in ipsi- and contralateral brain. Plasma and brain incretin and DPP-4 activity levels were quantified. Furthermore, brain incretin receptor levels were age-dependently evaluated in rodents, in 6-OHDA challenged animals and human subjects with/without PD. Cellular studies evaluated neurotrophic/neuroprotective actions of combined incretin administration. Pre-treatment with oral sitagliptin or PF-00734,200 reduced methamphetamine (meth)-induced rotation post-lesioning and dopaminergic degeneration in lesioned substantia nigra pars compacta (SNc) and striatum. Direct intracerebroventricular gliptin administration lacked neuroprotective actions, indicating that systemic incretin-mediated mechanisms underpin gliptin-induced favorable brain effects. Post-treatment with a threefold higher oral gliptin dose, likewise, mitigated meth-induced rotation, dopaminergic neurodegeneration and neuroinflammation, and augmented neurogenesis. These gliptin-induced actions associated with 70-80% plasma and 20-30% brain DPP-4 inhibition, and elevated plasma and brain incretin levels. Brain incretin receptor protein levels were age-dependently maintained in rodents, preserved in rats challenged with 6-OHDA, and in humans with PD. Combined GLP-1 and GIP receptor activation in neuronal cultures resulted in neurotrophic/neuroprotective actions superior to single agonists alone. In conclusion, these studies support further evaluation of the repurposing of clinically approved gliptins as a treatment strategy for PD.
Collapse
Affiliation(s)
- Seong-Jin Yu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, 35053, Taiwan.
- National Institute On Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA.
| | - Hui Shen
- National Institute On Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Eun-Kyung Bae
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Yazhou Li
- National Institute On Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Kumar Sambamurti
- Department of Neurosciences, the Medical University of South Carolina, Charleston, SC, 29425, USA
| | | | | | - Barry J Hoffer
- Department of Neurosurgery, University Hospitals of Cleveland, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Nigel H Greig
- National Institute On Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
18
|
Li R, Miao Z, Liu Y, Chen X, Wang H, Su J, Chen J. The Brain-Gut-Bone Axis in Neurodegenerative Diseases: Insights, Challenges, and Future Prospects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307971. [PMID: 39120490 PMCID: PMC11481201 DOI: 10.1002/advs.202307971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 06/04/2024] [Indexed: 08/10/2024]
Abstract
Neurodegenerative diseases are global health challenges characterized by the progressive degeneration of nerve cells, leading to cognitive and motor impairments. The brain-gut-bone axis, a complex network that modulates multiple physiological systems, has gained increasing attention owing to its profound effects on the occurrence and development of neurodegenerative diseases. No comprehensive review has been conducted to clarify the triangular relationship involving the brain-gut-bone axis and its potential for innovative therapies for neurodegenerative disorders. In light of this, a new perspective is aimed to propose on the interplay between the brain, gut, and bone systems, highlighting the potential of their dynamic communication in neurodegenerative diseases, as they modulate multiple physiological systems, including the nervous, immune, endocrine, and metabolic systems. Therapeutic strategies for maintaining the balance of the axis, including brain health regulation, intestinal microbiota regulation, and improving skeletal health, are also explored. The intricate physiological interactions within the brain-gut-bone axis pose a challenge in the development of effective treatments that can comprehensively target this system. Furthermore, the safety of these treatments requires further evaluation. This review offers a novel insights and strategies for the prevention and treatment of neurodegenerative diseases, which have important implications for clinical practice and patient well-being.
Collapse
Affiliation(s)
- Rong Li
- Department of NeurosurgeryShanghai Changhai HospitalNaval Medical UniversityShanghai200433China
| | - Zong Miao
- Department of NeurosurgeryShanghai Changhai HospitalNaval Medical UniversityShanghai200433China
| | - Yu'e Liu
- Tongji University Cancer CenterShanghai Tenth People's Hospital of Tongji UniversitySchool of MedicineTongji UniversityShanghai200092China
| | - Xiao Chen
- Department of OrthopedicsXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092China
- Institute of Translational MedicineShanghai UniversityShanghai200444China
- Organoid Research CenterShanghai UniversityShanghai200444China
| | - Hongxiang Wang
- Department of NeurosurgeryShanghai Changhai HospitalNaval Medical UniversityShanghai200433China
| | - Jiacan Su
- Department of OrthopedicsXinhua HospitalShanghai Jiao Tong University School of MedicineShanghai200092China
- Institute of Translational MedicineShanghai UniversityShanghai200444China
- Organoid Research CenterShanghai UniversityShanghai200444China
| | - Juxiang Chen
- Department of NeurosurgeryShanghai Changhai HospitalNaval Medical UniversityShanghai200433China
| |
Collapse
|
19
|
Joers V, Murray BC, McLaughlin C, Oliver D, Staley HE, Coronado J, Achat-Mendes C, Golshani S, Kelly SD, Goodson M, Lee D, Manfredsson FP, Moore Ii BM, Tansey MG. Modulation of cannabinoid receptor 2 alters neuroinflammation and reduces formation of alpha-synuclein aggregates in a rat model of nigral synucleinopathy. J Neuroinflammation 2024; 21:240. [PMID: 39334169 PMCID: PMC11438102 DOI: 10.1186/s12974-024-03221-5] [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: 03/26/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Research into the disequilibrium of microglial phenotypes has become an area of intense focus in neurodegenerative disease as a potential mechanism that contributes to chronic neuroinflammation and neuronal loss in Parkinson's disease (PD). There is growing evidence that neuroinflammation accompanies and may promote progression of alpha-synuclein (Asyn)-induced nigral dopaminergic (DA) degeneration. From a therapeutic perspective, development of immunomodulatory strategies that dampen overproduction of pro-inflammatory cytokines from chronically activated immune cells and induce a pro-phagocytic phenotype is expected to promote Asyn removal and protect vulnerable neurons. Cannabinoid receptor-2 (CB2) is highly expressed on activated microglia and peripheral immune cells, is upregulated in the substantia nigra of individuals with PD and in mouse models of nigral degeneration. Furthermore, modulation of CB2 protects against rotenone-induced nigral degeneration; however, CB2 has not been pharmacologically and selectively targeted in an Asyn model of PD. Here, we report that 7 weeks of peripheral administration of CB2 inverse agonist SMM-189 reduced phosphorylated (pSer129) Asyn in the substantia nigra compared to vehicle treatment. Additionally, SMM-189 delayed Asyn-induced immune cell infiltration into the brain as determined by flow cytometry, increased CD68 protein expression, and elevated wound-healing-immune-mediator gene expression. Additionally, peripheral immune cells increased wound-healing non-classical monocytes and decreased pro-inflammatory classical monocytes. In vitro analysis of RAW264.7 macrophages treated with lipopolysaccharide (LPS) and SMM-189 revealed increased phagocytosis as measured by the uptake of fluorescence of pHrodo E. coli bioparticles. Together, results suggest that targeting CB2 with SMM-189 skews immune cell function toward a phagocytic phenotype and reduces toxic aggregated species of Asyn. Our novel findings demonstrate that CB2 may be a target to modulate inflammatory and immune responses in proteinopathies.
Collapse
Affiliation(s)
- Valerie Joers
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA.
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, USA.
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| | | | | | - Danielle Oliver
- Department of Physiology, Emory University, Atlanta, GA, USA
| | - Hannah E Staley
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jazmyn Coronado
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | | | - Sanam Golshani
- Department of Physiology, Emory University, Atlanta, GA, USA
| | - Sean D Kelly
- Department of Physiology, Emory University, Atlanta, GA, USA
| | - Matthew Goodson
- Department of Physiology, Emory University, Atlanta, GA, USA
| | - Danica Lee
- Department of Physiology, Emory University, Atlanta, GA, USA
| | - Fredric P Manfredsson
- Parkinson's Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Bob M Moore Ii
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Malú Gámez Tansey
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, FL, USA
- McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, USA
| |
Collapse
|
20
|
Minnella A, McCusker KP, Amagata A, Trias B, Weetall M, Latham JC, O'Neill S, Wyse RK, Klein MB, Trimmer JK. Targeting ferroptosis with the lipoxygenase inhibitor PTC-041 as a therapeutic strategy for the treatment of Parkinson's disease. PLoS One 2024; 19:e0309893. [PMID: 39292705 PMCID: PMC11410249 DOI: 10.1371/journal.pone.0309893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 08/18/2024] [Indexed: 09/20/2024] Open
Abstract
Parkinson's disease is the second most common neurodegenerative disorder, affecting nearly 10 million people worldwide. Ferroptosis, a recently identified form of regulated cell death characterized by 15-lipoxygenase-mediated hydroperoxidation of membrane lipids, has been implicated in neurodegenerative disorders including amyotrophic lateral sclerosis and Parkinson's disease. Pharmacological inhibition of 15 -lipoxygenase to prevent iron- and lipid peroxidation-associated ferroptotic cell death is a rational strategy for the treatment of Parkinson's disease. We report here the characterization of PTC-041 as an anti-ferroptotic reductive lipoxygenase inhibitor developed for the treatment of Parkinson's disease. In these studies, PTC-041 potently protects primary human Parkinson's disease patient-derived fibroblasts from lipid peroxidation and subsequent ferroptotic cell death and prevents ferroptosis-related neuronal loss and astrogliosis in primary rat neuronal cultures. Additionally, PTC-041 prevents ferroptotic-mediated α-synuclein protein aggregation and nitrosylation in vitro, suggesting a potential role for anti-ferroptotic lipoxygenase inhibitors in mitigating pathogenic aspects of synucleinopathies such as Parkinson's disease. We further found that PTC-041 protects against synucleinopathy in vivo, demonstrating that PTC-041 treatment of Line 61 transgenic mice protects against α-synuclein aggregation and phosphorylation as well as prevents associated neuronal and non-neuronal cell death. Finally, we show that. PTC-041 protects against 6-hydroxydopamine-induced motor deficits in a hemiparkinsonian rat model, further validating the potential therapeutic benefits of lipoxygenase inhibitors in the treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Angela Minnella
- PTC Therapeutics, Mountain View, California, United States of America
| | - Kevin P McCusker
- PTC Therapeutics, Mountain View, California, United States of America
| | - Akiko Amagata
- PTC Therapeutics, Mountain View, California, United States of America
| | - Beatrice Trias
- PTC Therapeutics, Warren, New Jersey, United States of America
| | - Marla Weetall
- PTC Therapeutics, Warren, New Jersey, United States of America
| | - Joey C Latham
- PTC Therapeutics, Mountain View, California, United States of America
| | - Sloane O'Neill
- PTC Therapeutics, Mountain View, California, United States of America
| | | | - Matthew B Klein
- PTC Therapeutics, Warren, New Jersey, United States of America
| | - Jeffrey K Trimmer
- PTC Therapeutics, Mountain View, California, United States of America
| |
Collapse
|
21
|
Miller MR, Landis HE, Miller RE, Tizabi Y. Intercellular Adhesion Molecule 1 (ICAM-1): An Inflammatory Regulator with Potential Implications in Ferroptosis and Parkinson's Disease. Cells 2024; 13:1554. [PMID: 39329738 PMCID: PMC11430830 DOI: 10.3390/cells13181554] [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: 08/09/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Intercellular adhesion molecule 1 (ICAM-1/CD54), a transmembrane glycoprotein, has been considered as one of the most important adhesion molecules during leukocyte recruitment. It is encoded by the ICAM1 gene and plays a central role in inflammation. Its crucial role in many inflammatory diseases such as ulcerative colitis and rheumatoid arthritis are well established. Given that neuroinflammation, underscored by microglial activation, is a key element in neurodegenerative diseases such as Parkinson's disease (PD), we investigated whether ICAM-1 has a role in this progressive neurological condition and, if so, to elucidate the underpinning mechanisms. Specifically, we were interested in the potential interaction between ICAM-1, glial cells, and ferroptosis, an iron-dependent form of cell death that has recently been implicated in PD. We conclude that there exist direct and indirect (via glial cells and T cells) influences of ICAM-1 on ferroptosis and that further elucidation of these interactions can suggest novel intervention for this devastating disease.
Collapse
Affiliation(s)
| | - Harold E. Landis
- Integrative Medicine Fellow, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | | | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA
| |
Collapse
|
22
|
Zhang F, Pan L, Lian C, Xu Z, Chen H, Lai W, Liang X, Liu Q, Wu H, Wang Y, Zhang P, Zhang G, Liu Z. ICAM-1 may promote the loss of dopaminergic neurons by regulating inflammation in MPTP-induced Parkinson's disease mouse models. Brain Res Bull 2024; 214:110989. [PMID: 38825252 DOI: 10.1016/j.brainresbull.2024.110989] [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: 03/16/2024] [Revised: 05/12/2024] [Accepted: 05/27/2024] [Indexed: 06/04/2024]
Abstract
Parkinson's disease (PD) is a chronic neurodegenerative disease with unclear pathogenesis that involves neuroinflammation and intestinal microbial dysbiosis. Intercellular adhesion molecule-1 (ICAM-1), an inflammatory marker, participates in neuroinflammation during dopaminergic neuronal damage. However, the explicit mechanisms of action of ICAM-1 in PD have not been elucidated. We established a subacute PD mouse model by the intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and observed motor symptoms and gastrointestinal dysfunction in mice. Immunofluorescence was used to examine the survival of dopaminergic neurons, expression of microglial and astrocyte markers, and intestinal tight junction-associated proteins. Then, we use 16 S rRNA sequencing to identify alterations in the microbiota. Our findings revealed that ICAM-1-specific antibody (Ab) treatment relieved behavioural defects, gastrointestinal dysfunction, and dopaminergic neuronal death in MPTP-induced PD mice. Further mechanistic investigations indicated that ICAM-1Ab might suppress neuroinflammation by inhibiting the activation of astrocytes and microglia in the substantia nigra and relieving colon barrier impairment and intestinal inflammation. Furthermore, 16 S rRNA sequencing revealed that the relative abundances of bacterial Firmicutes, Clostridia, and Lachnospiraceae were elevated in the PD mice. However, ICAM-1Ab treatment ameliorated the MPTP-induced disorders in the intestinal microbiota. Collectively, we concluded that the suppressing ICAM-1 might lead to the a significant decrease of inflammation and restore the gut microbial community, thus ameliorating the damage of DA neurons.
Collapse
Affiliation(s)
- Fen Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Rehabilitation Medicine, The First People's Hospital of Foshan, Foshan, Guangdong, China; Zunyi Medical University, Zunyi, Guizhou, China
| | - Lixin Pan
- Department of Neurology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Changlin Lian
- Department of Neurology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Zhifeng Xu
- Department of Neurology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Hongda Chen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenjie Lai
- Department of Neurology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, China
| | - Xiaojue Liang
- Department of Neurology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Qiyuan Liu
- Shantou University, Chaoshan, Guangdong, China
| | - Haomin Wu
- Department of Rehabilitation Medicine, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Yukai Wang
- Department of Neurology, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Pande Zhang
- Department of Rehabilitation Medicine, The First People's Hospital of Foshan, Foshan, Guangdong, China.
| | - Guohua Zhang
- Department of Neurology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, China.
| | - Zhen Liu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China; Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
23
|
Cai P, Li W, Xu Y, Wang H. Drp1 and neuroinflammation: Deciphering the interplay between mitochondrial dynamics imbalance and inflammation in neurodegenerative diseases. Neurobiol Dis 2024; 198:106561. [PMID: 38857809 DOI: 10.1016/j.nbd.2024.106561] [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: 03/17/2024] [Revised: 05/24/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024] Open
Abstract
Neuroinflammation and mitochondrial dysfunction are closely intertwined with the pathophysiology of neurological disorders. Recent studies have elucidated profound alterations in mitochondrial dynamics across a spectrum of neurological disorders. Dynamin-related protein 1 (DRP1) emerges as a pivotal regulator of mitochondrial fission, with its dysregulation disrupting mitochondrial homeostasis and fueling neuroinflammation, thereby exacerbating disease severity. In addition to its role in mitochondrial dynamics, DRP1 plays a crucial role in modulating inflammation-related pathways. This review synthesizes important functions of DRP1 in the central nervous system (CNS) and the impact of epigenetic modification on the progression of neurodegenerative diseases. The intricate interplay between neuroinflammation and DRP1 in microglia and astrocytes, central contributors to neuroinflammation, is expounded upon. Furthermore, the use of DRP1 inhibitors to influence the activation of microglia and astrocytes, as well as their involvement in processes such as mitophagy, mitochondrial oxidative stress, and calcium ion transport in CNS-mediated neuroinflammation, is scrutinized. The modulation of microglia to astrocyte crosstalk by DRP1 and its role in inflammatory neurodegeneration is also highlighted. Overall, targeting DRP1 presents a promising avenue for ameliorating neuroinflammation and enhancing the therapeutic management of neurological disorders.
Collapse
Affiliation(s)
- Peiyang Cai
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Wuhao Li
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Ye Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China
| | - Hui Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, PR China..
| |
Collapse
|
24
|
Folke J, Skougaard M, Korsholm TL, Laursen ALS, Salvesen L, Hejl AM, Bech S, Løkkegaard A, Brudek T, Ditlev SB, Aznar S. Assessing serum anti-nuclear antibodies HEp-2 patterns in synucleinopathies. Immun Ageing 2024; 21:49. [PMID: 39026277 PMCID: PMC11256463 DOI: 10.1186/s12979-024-00453-0] [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: 04/26/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
This study investigates the presence of antinuclear antibodies (ANA) in three primary synucleinopathies - Parkinson's disease (PD), multiple system atrophy (MSA), and dementia with Lewy bodies (DLB), compared to healthy controls. Autoinflammatory disorders typically involve the immune system mistakenly attacking the body's own cells and start producing ANA. There is an increasing body of evidence that immune-mediated inflammation is a pathological feature linked to synucleinopathies. To investigate whether this could be autoimmune mediated we analyzed for ANA in the plasma of 25 MSA, 25 PD, and 17 DLB patients, along with 25 healthy controls, using the ANA HEp-2 indirect immunofluorescence antibody assay (ANA HEp-2 IFA). Contrary to initial expectations, results showed ANA HEp-2 positivity in 12% of PD, 8% of MSA patients, 18% of DLB patients, and 17% of healthy controls, indicating no increased prevalence of ANA in synucleinopathies compared to age-matched healthy individuals. Various ANA HEp-2 patterns were identified, but no specific pattern was associated with individual synucleinopathies. We conclude hereby that synucleinopathies are not associated with detectable presence of ANA in plasma.
Collapse
Affiliation(s)
- Jonas Folke
- Centre for Neuroscience & Stereology, Department of Neurology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marie Skougaard
- Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Trine-Line Korsholm
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Anne-Line Strange Laursen
- Centre for Neuroscience & Stereology, Department of Neurology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lisette Salvesen
- Department of Neurology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen Ø, DK-2100, Denmark
| | - Anne-Mette Hejl
- Department of Neurology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen Ø, DK-2100, Denmark
| | - Sara Bech
- Department of Neurology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Annemette Løkkegaard
- Department of Neurology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen Ø, DK-2100, Denmark
| | - Tomasz Brudek
- Centre for Neuroscience & Stereology, Department of Neurology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sisse Bolm Ditlev
- Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Susana Aznar
- Centre for Neuroscience & Stereology, Department of Neurology, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark.
- Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark.
| |
Collapse
|
25
|
Chen YC, Liaw YC, Nfor ON, Hsiao CH, Zhong JH, Wu SL, Liaw YP. Epigenetic associations of GPNMB rs199347 variant with alcohol consumption in Parkinson's disease. Front Psychiatry 2024; 15:1377403. [PMID: 39091454 PMCID: PMC11293056 DOI: 10.3389/fpsyt.2024.1377403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/27/2024] [Indexed: 08/04/2024] Open
Abstract
Introduction Alcohol consumption can induce a neuroinflammatory response and contribute to the progression of neurodegeneration. However, its association with Parkinson's disease (PD), the second most common neurodegenerative disorder, remains undetermined. Recent studies suggest that the glycoprotein non-metastatic melanoma protein B (GPNMB) is a potential biomarker for PD. We evaluated the association of rs199347, a variant of the GPNMB gene, with alcohol consumption and methylation upstream of GPNMB. Methods We retrieved genetic and DNA methylation data obtained from participants enrolled in the Taiwan Biobank (TWB) between 2008 and 2016. After excluding individuals with incomplete or missing information about potential PD risk factors, we included 1,357 participants in our final analyses. We used multiple linear regression to assess the association of GPNMB rs199347 and chronic alcohol consumption (and other potential risk factors) with GPNMB cg17274742 methylation. Results There was no difference between the distribution of GPNMB rs199347 genotypes between chronic alcohol consumers and the other study participants. A significant interaction was observed between the GPNMB rs199347 variant and alcohol consumption (p = 0.0102) concerning cg17274742 methylation. Compared to non-chronic alcohol consumers with the AA genotype, alcohol drinkers with the rs199347 GG genotype had significantly lower levels (hypomethylation) of cg17274742 (p = 0.0187). Conclusion Alcohol consumption among individuals with the rs199347 GG genotype was associated with lower levels of cg17274742 methylation, which could increase expression of the GPNMB gene, an important neuroinflammatory-related risk gene for PD.
Collapse
Affiliation(s)
- Yen-Chung Chen
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung, Taiwan
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan
| | - Yi-Chia Liaw
- Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Oswald Ndi Nfor
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung, Taiwan
| | - Chih-Hsuan Hsiao
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung, Taiwan
| | - Ji-Han Zhong
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung, Taiwan
| | - Shey-Lin Wu
- Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan
- Department of Electrical Engineering, National Changhua University of Education, Changhua, Taiwan
| | - Yung-Po Liaw
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
26
|
Yan H, Wang W, Cui T, Shao Y, Li M, Fang L, Feng L. Advances in the Understanding of the Correlation Between Neuroinflammation and Microglia in Alzheimer's Disease. Immunotargets Ther 2024; 13:287-304. [PMID: 38881647 PMCID: PMC11180466 DOI: 10.2147/itt.s455881] [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: 03/14/2024] [Accepted: 06/05/2024] [Indexed: 06/18/2024] Open
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disease with a subtle and progressive onset and is the most common type of dementia. However, its etiology and pathogenesis have not yet been fully elucidated. The common pathological manifestations of AD include extraneuronal β-amyloid deposition (Aβ), intraneuronal tau protein phosphorylation leading to the formation of 'neurofibrillary tangles' (NFTs), neuroinflammation, progressive loss of brain neurons/synapses, and glucose metabolism disorders. Current treatment approaches for AD primarily focus on the 'Aβ cascade hypothesis and abnormal aggregation of hyperphosphorylation of tau proteins', but have shown limited efficacy. Therefore, there is an ongoing need to identify more effective treatment targets for AD. The central nervous system (CNS) inflammatory response plays a key role in the occurrence and development of AD. Neuroinflammation is an immune response activated by glial cells in the CNS that usually occurs in response to stimuli such as nerve injury, infection and toxins or in response to autoimmunity. Neuroinflammation ranks as the third most prominent pathological feature in AD, following Aβ and NFTs. In recent years, the focus on the role of neuroinflammation and microglia in AD has increased due to the advancements in genome-wide association studies (GWAS) and sequencing technology. Furthermore, research has validated the pivotal role of microglia-mediated neuroinflammation in the progression of AD. Therefore, this article reviews the latest research progress on the role of neuroinflammation triggered by microglia in AD in recent years, aiming to provide a new theoretical basis for further exploring the role of neuroinflammation in the process of AD occurrence and development.
Collapse
Affiliation(s)
- Huiying Yan
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Wei Wang
- Department of Intensive Care Unit, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Tingting Cui
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Yanxin Shao
- Department of Neurology, The Second Affiliated Hospital of Shandong First Medical University, Taian, People's Republic of China
| | - Mingquan Li
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Limei Fang
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| | - Lina Feng
- Department of Neurology, The Third Affiliated Clinical Hospital of the Changchun University of Chinese Medicine, Changchun, People's Republic of China
| |
Collapse
|
27
|
Li Y, Yu C, Jiang X, Fu J, Sun N, Zhang D. The mechanistic view of non-coding RNAs as a regulator of inflammatory pathogenesis of Parkinson's disease. Pathol Res Pract 2024; 258:155349. [PMID: 38772115 DOI: 10.1016/j.prp.2024.155349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/17/2024] [Accepted: 05/10/2024] [Indexed: 05/23/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta, leading to motor and non-motor symptoms. Emerging evidence suggests that inflammation plays a crucial role in the pathogenesis of PD, with the NLRP3 inflammasome implicated as a key mediator. Nfon-coding RNAs (ncRNAs), including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), have recently garnered attention for their regulatory roles in various biological processes, including inflammation. This review aims to provide a mechanistic insight into how ncRNAs function as regulators of inflammatory pathways in PD, with a specific focus on the NLRP3 inflammasome. We discuss the dysregulation of miRNAs and lncRNAs in PD pathogenesis and their impact on neuroinflammation through modulation of NLRP3 activation, cytokine production, and microglial activation. Additionally, we explore the crosstalk between ncRNAs, alpha-synuclein pathology, and mitochondrial dysfunction, further elucidating the intricate network underlying PD-associated inflammation. Understanding the mechanistic roles of ncRNAs in regulating inflammatory pathways may offer novel therapeutic targets for the treatment of PD and provide insights into the broader implications of ncRNA-mediated regulation in neuroinflammatory diseases.
Collapse
Affiliation(s)
- Yu'an Li
- Department of Neurosurgery, Jilin Province FAW General Hospital, Changchun 130000, China
| | - Chunlei Yu
- Department of Neurosurgery, Jilin Province FAW General Hospital, Changchun 130000, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Jilin Province FAW General Hospital, Changchun 130000, China
| | - Jia Fu
- Department of Neurosurgery, Jilin Province FAW General Hospital, Changchun 130000, China
| | - Ning Sun
- Department of Neurosurgery, Jilin Province FAW General Hospital, Changchun 130000, China
| | - Daquan Zhang
- Department of Neurosurgery, Jilin Province FAW General Hospital, Changchun 130000, China.
| |
Collapse
|
28
|
Hassani S, Esmaeili A. The neuroprotective effects of ferulic acid in toxin-induced models of Parkinson's disease: A review. Ageing Res Rev 2024; 97:102299. [PMID: 38604452 DOI: 10.1016/j.arr.2024.102299] [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: 12/16/2023] [Revised: 03/04/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
Parkinson's disease is predominantly caused by dopaminergic neuron loss in the substantia nigra pars compacta and the accumulation of alpha-synuclein protein. Though the general consensus is that several factors, such as aging, environmental factors, mitochondrial dysfunction, accumulations of neurotoxic alpha-synuclein, malfunctions of the lysosomal and proteasomal protein degradation systems, oxidative stress, and neuroinflammation, are involved in the neurodegeneration process of Parkinson's disease, the precise mechanism by which all of these factors are triggered remains unknown. Typically, neurotoxic compounds such as rotenone, 6-hydroxydopamine, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), 1-methyl 4-phenyl pyridinium (mpp+), paraquat, and maneb are used to Preclinical models of Parkinson's disease Ferulic acid is often referred to by its scientific name, 4-hydroxy-3-methoxycinnamic acid (C10H10O4), and is found naturally in cereals, fruits, vegetables, and bee products. This substance exhibits neuroprotective effects against Parkinson's disease because of its intriguing potential, which includes anti-inflammatory and antioxidant qualities. This review goes into additional detail about Parkinson's disease and the neuroprotective properties of ferulic acid that may help prevent the condition.
Collapse
Affiliation(s)
- Samira Hassani
- Department of Plant and Animal Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Abolghasem Esmaeili
- Department of Cell and Molecular Biology & Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
29
|
Song LJ, Sui RX, Wang J, Miao Q, He Y, Yin JJ, An J, Ding ZB, Han QX, Wang Q, Yu JZ, Xiao BG, Ma CG. Targeting the differentiation of astrocytes by Bilobalide in the treatment of Parkinson's disease model. Int J Neurosci 2024; 134:274-291. [PMID: 36037147 DOI: 10.1080/00207454.2022.2100778] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 12/27/2022]
Abstract
Background: The etiology of Parkinson's disease (PD), a chronic and progressive neurodegenerative disease, is multifactorial but not fully unknown. Until now, no drug has been proven to have neuroprotective or neuroregenerative effects in patients with PD. Objectives: To observe the therapeutic potential of Bilobalide (BB), a constituent of ginkgo biloba, in MPTP-induced PD model, and explore its possible mechanisms of action. Material and Methods: Mice were randomly divided into three groups: healthy group, MPTP group and MPTP + BB group. PD-related phenotypes were induced by intraperitoneal injection of MPTP into male C57BL/6 mice, and BB (40 mg/kg/day) was intraperitoneally given for 7 consecutive days at the end of modeling. The injection of saline was set up as the control in a similar manner. Results: BB induced M2 polarization of microglia, accompanied by inhibition of neuroinflammation in the brain. Simultaneously, BB promoted the expression of BDNF in astrocytes and neurons, and expression of GDNF in neurons. Most interestingly, BB enhanced the formation of GFAP+ astrocytes expressing nestin, Brn2 and Ki67, as well as the transformation of GFAP+ astrocytes expressing tyrosine hydroxylase around subventricular zone, providing experimental evidence that BB could promote the conversion of astrocytes into TH+ dopamine neurons in vivo and in vitro. Conclusions: These results suggest the natural product BB may utilize multiple pathways to modify degenerative process of TH+ neurons, revealing an exciting opportunity for novel neuroprotective therapeutics. However, its multi-target and important mechanisms need to be further explored.
Collapse
Affiliation(s)
- Li-Juan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Ruo-Xuan Sui
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Wang
- The Department of Neurology, Shanxi Medical University, Taiyuan, China
| | - Qiang Miao
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Yan He
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Jun-Jun Yin
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Jun An
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Zhi-Bin Ding
- The Department of Neurology, Shanxi Medical University, Taiyuan, China
| | - Qing-Xian Han
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Qing Wang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Jie-Zhong Yu
- The NO. 1 Affiliated Hospital/Institute of Brain Science, Shanxi Datong University, Datong, China
| | - Bao-Guo Xiao
- Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Cun-Gen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
- The Department of Neurology, Shanxi Medical University, Taiyuan, China
- The NO. 1 Affiliated Hospital/Institute of Brain Science, Shanxi Datong University, Datong, China
| |
Collapse
|
30
|
Rombaut A, Jovancevic D, Wong RCB, Nicol A, Brautaset R, Finkelstein DI, Nguyen CTO, Tribble JR, Williams PA. Intravitreal MPTP drives retinal ganglion cell loss with oral nicotinamide treatment providing robust neuroprotection. Acta Neuropathol Commun 2024; 12:79. [PMID: 38773545 PMCID: PMC11107037 DOI: 10.1186/s40478-024-01782-3] [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: 02/02/2024] [Accepted: 04/16/2024] [Indexed: 05/24/2024] Open
Abstract
Neurodegenerative diseases have common underlying pathological mechanisms including progressive neuronal dysfunction, axonal and dendritic retraction, and mitochondrial dysfunction resulting in neuronal death. The retina is often affected in common neurodegenerative diseases such as Parkinson's and Alzheimer's disease. Studies have demonstrated that the retina in patients with Parkinson's disease undergoes changes that parallel the dysfunction in the brain. These changes classically include decreased levels of dopamine, accumulation of alpha-synuclein in the brain and retina, and death of dopaminergic nigral neurons and retinal amacrine cells leading to gross neuronal loss. Exploring this disease's retinal phenotype and vision-related symptoms is an important window for elucidating its pathophysiology and progression, and identifying novel ways to diagnose and treat Parkinson's disease. 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) is commonly used to model Parkinson's disease in animal models. MPTP is a neurotoxin converted to its toxic form by astrocytes, transported to neurons through the dopamine transporter, where it causes mitochondrial Complex I inhibition and neuron degeneration. Systemic administration of MPTP induces retinal changes in different animal models. In this study, we assessed the effects of MPTP on the retina directly via intravitreal injection in mice (5 mg/mL and 50 mg/mL to 7, 14 and 21 days post-injection). MPTP treatment induced the reduction of retinal ganglion cells-a sensitive neuron in the retina-at all time points investigated. This occurred without a concomitant loss of dopaminergic amacrine cells or neuroinflammation at any of the time points or concentrations tested. The observed neurodegeneration which initially affected retinal ganglion cells indicated that this method of MPTP administration could yield a fast and straightforward model of retinal ganglion cell neurodegeneration. To assess whether this model could be amenable to neuroprotection, mice were treated orally with nicotinamide (a nicotinamide adenine dinucleotide precursor) which has been demonstrated to be neuroprotective in several retinal ganglion cell injury models. Nicotinamide was strongly protective following intravitreal MPTP administration, further supporting intravitreal MPTP use as a model of retinal ganglion cell injury. As such, this model could be utilized for testing neuroprotective treatments in the context of Parkinson's disease and retinal ganglion cell injury.
Collapse
Affiliation(s)
- Anne Rombaut
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Danica Jovancevic
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Raymond Ching-Bong Wong
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia
- Department of Surgery (Ophthalmology), The University of Melbourne, Melbourne, Australia
| | - Alan Nicol
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Rune Brautaset
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - David I Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Christine T O Nguyen
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Australia
| | - James R Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
31
|
Joers V, Murray BC, McLaughlin C, Oliver D, Staley H, Coronado J, Achat-Mendes C, Golshani S, Kelly SD, Goodson M, Lee D, Manfredsson FP, Moore BM, Tansey MG. Modulation of cannabinoid receptor 2 alters neuroinflammation and reduces formation of alpha-synuclein aggregates in a rat model of nigral synucleinopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.25.554814. [PMID: 38562842 PMCID: PMC10983852 DOI: 10.1101/2023.08.25.554814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Research into the disequilibrium of microglial phenotypes has become an area of intense focus in neurodegenerative disease as a potential mechanism that contributes to chronic neuroinflammation and neuronal loss in Parkinson's disease (PD). There is growing evidence that neuroinflammation accompanies and may promote progression of alpha-synuclein (Asyn)-induced nigral dopaminergic (DA) degeneration. From a therapeutic perspective, development of immunomodulatory strategies that dampen overproduction of pro-inflammatory cytokines from chronically activated immune cells and induce a pro-phagocytic phenotype is expected to promote Asyn removal and protect vulnerable neurons. Cannabinoid receptor-2 (CB2) is highly expressed on activated microglia and peripheral immune cells, is upregulated in the substantia nigra of individuals with PD and in mouse models of nigral degeneration. Furthermore, modulation of CB2 protects against rotenone-induced nigral degeneration; however, CB2 has not been pharmacologically and selectively targeted in an Asyn model of PD. Here, we report that 7 weeks of peripheral administration of CB2 inverse agonist SMM-189 reduced phosphorylated (pSer129) alpha-synuclein in the substantia nigra compared to vehicle treatment. Additionally, SMM-189 delayed Asyn-induced immune cell infiltration into the brain as determined by flow cytometry, increased CD68 protein expression, and elevated wound-healing-immune-mediator gene expression. Additionally, peripheral immune cells increased wound-healing non-classical monocytes and decreased pro-inflammatory classical monocytes. In vitro analysis of RAW264.7 macrophages treated with lipopolysaccharide (LPS) and SMM-189 revealed increased phagocytosis as measured by the uptake of fluorescence of pHrodo E. coli bioparticles. Together, results suggest that targeting CB2 with SMM-189 skews immune cell function toward a phagocytic phenotype and reduces toxic aggregated species of Asyn. Our novel findings demonstrate that CB2 may be a target to modulate inflammatory and immune responses in proteinopathies.
Collapse
Affiliation(s)
- Valerie Joers
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, Florida
- McKnight Brain Institute, University of Florida, Gainesville, Florida
| | | | | | - Danielle Oliver
- Department of Physiology, Emory University, Atlanta, Georgia
| | - Hannah Staley
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, Florida
- McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Jazmyn Coronado
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, Florida
- McKnight Brain Institute, University of Florida, Gainesville, Florida
| | | | - Sanam Golshani
- Department of Physiology, Emory University, Atlanta, Georgia
| | - Sean D Kelly
- Department of Physiology, Emory University, Atlanta, Georgia
| | - Matthew Goodson
- Department of Physiology, Emory University, Atlanta, Georgia
| | - Danica Lee
- Department of Physiology, Emory University, Atlanta, Georgia
| | - Fredric P Manfredsson
- Parkinson's Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, Arizona
| | - Bob M Moore
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Malú Gámez Tansey
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, Florida
- Center for Translational Research in Neurodegenerative Disease, University of Florida College of Medicine, Gainesville, Florida
- McKnight Brain Institute, University of Florida, Gainesville, Florida
- Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, Florida
| |
Collapse
|
32
|
Rao Y, Peng B. Allogenic microglia replacement: A novel therapeutic strategy for neurological disorders. FUNDAMENTAL RESEARCH 2024; 4:237-245. [PMID: 38933508 PMCID: PMC11197774 DOI: 10.1016/j.fmre.2023.02.025] [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: 09/17/2022] [Revised: 11/17/2022] [Accepted: 02/19/2023] [Indexed: 03/29/2023] Open
Abstract
Microglia are resident immune cells in the central nervous system (CNS) that play vital roles in CNS development, homeostasis and disease pathogenesis. Genetic defects in microglia lead to microglial dysfunction, which in turn leads to neurological disorders. The correction of the specific genetic defects in microglia in these disorders can lead to therapeutic effects. Traditional genetic defect correction approaches are dependent on viral vector-based genetic defect corrections. However, the viruses used in these approaches, including adeno-associated viruses, lentiviruses and retroviruses, do not primarily target microglia; therefore, viral vector-based genetic defect corrections are ineffective in microglia. Microglia replacement is a novel approach to correct microglial genetic defects via replacing microglia of genetic defects with allogenic healthy microglia. In this paper, we systematically review the history, rationale and therapeutic perspectives of microglia replacement, which would be a novel strategy for treating CNS disorders.
Collapse
Affiliation(s)
- Yanxia Rao
- Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Bo Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200000, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
33
|
Tewari M, Michalski S, Egan TM. Modulation of Microglial Function by ATP-Gated P2X7 Receptors: Studies in Rat, Mice and Human. Cells 2024; 13:161. [PMID: 38247852 PMCID: PMC10814008 DOI: 10.3390/cells13020161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
P2X receptors are a family of seven ATP-gated ion channels that trigger physiological and pathophysiological responses in a variety of cells. Five of the family members are sensitive to low concentrations of extracellular ATP, while the P2X6 receptor has an unknown affinity. The last subtype, the P2X7 receptor, is unique in requiring millimolar concentrations to fully activate in humans. This low sensitivity imparts the agonist with the ability to act as a damage-associated molecular pattern that triggers the innate immune response in response to the elevated levels of extracellular ATP that accompany inflammation and tissue damage. In this review, we focus on microglia because they are the primary immune cells of the central nervous system, and they activate in response to ATP or its synthetic analog, BzATP. We start by introducing purinergic receptors and then briefly consider the roles that microglia play in neurodevelopment and disease by referencing both original works and relevant reviews. Next, we move to the role of extracellular ATP and P2X receptors in initiating and/or modulating innate immunity in the central nervous system. While most of the data that we review involve work on mice and rats, we highlight human studies of P2X7R whenever possible.
Collapse
|
34
|
Miao Y, Meng H. The involvement of α-synucleinopathy in the disruption of microglial homeostasis contributes to the pathogenesis of Parkinson's disease. Cell Commun Signal 2024; 22:31. [PMID: 38216911 PMCID: PMC10785555 DOI: 10.1186/s12964-023-01402-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/18/2023] [Indexed: 01/14/2024] Open
Abstract
The intracellular deposition and intercellular transmission of α-synuclein (α-syn) are shared pathological characteristics among neurodegenerative disorders collectively known as α-synucleinopathies, including Parkinson's disease (PD). Although the precise triggers of α-synucleinopathies remain unclear, recent findings indicate that disruption of microglial homeostasis contributes to the pathogenesis of PD. Microglia play a crucial role in maintaining optimal neuronal function by ensuring a homeostatic environment, but this function is disrupted during the progression of α-syn pathology. The involvement of microglia in the accumulation, uptake, and clearance of aggregated proteins is critical for managing disease spread and progression caused by α-syn pathology. This review summarizes current knowledge on the interrelationships between microglia and α-synucleinopathies, focusing on the remarkable ability of microglia to recognize and internalize extracellular α-syn through diverse pathways. Microglia process α-syn intracellularly and intercellularly to facilitate the α-syn neuronal aggregation and cell-to-cell propagation. The conformational state of α-synuclein distinctly influences microglial inflammation, which can affect peripheral immune cells such as macrophages and lymphocytes and may regulate the pathogenesis of α-synucleinopathies. We also discuss ongoing research efforts to identify potential therapeutic approaches targeting both α-syn accumulation and inflammation in PD. Video Abstract.
Collapse
Affiliation(s)
- Yongzhen Miao
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Hongrui Meng
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China.
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
35
|
Neha, Wali Z, Pinky, Hattiwale SH, Jamal A, Parvez S. GLP-1/Sigma/RAGE receptors: An evolving picture of Alzheimer's disease pathology and treatment. Ageing Res Rev 2024; 93:102134. [PMID: 38008402 DOI: 10.1016/j.arr.2023.102134] [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: 09/03/2023] [Revised: 11/18/2023] [Accepted: 11/19/2023] [Indexed: 11/28/2023]
Abstract
According to the facts and figures 2023stated that 6.7 million Americans over the age of 65 have Alzheimer's disease (AD). The scenario of AD has reached up to the maximum, of 4.1 million individuals, 2/3rd are female patients, and approximately 1 in 9 adults over the age of 65 have dementia with AD dementia. The fact that there are now no viable treatments for AD indicates that the underlying disease mechanisms are not fully understood. The progressive neurodegenerative disease, AD is characterized by amyloid plaques and neurofibrillary tangles (NFTs) of abnormally hyperphosphorylated tau protein and senile plaques (SPs), which are brought on by the buildup of amyloid beta (Aβ). Numerous attempts have been made to produce compounds that interfere with these characteristics because of significant research efforts into the primary pathogenic hallmark of this disorder. Here, we summarize several research that highlights interesting therapy strategies and the neuroprotective effects of GLP-1, Sigma, and, AGE-RAGE receptors in pre-clinical and clinical AD models.
Collapse
Affiliation(s)
- Neha
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| | - Zitin Wali
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Pinky
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| | - Shaheenkousar H Hattiwale
- Department of Basic Medical Sciences, College of Medicine, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Azfar Jamal
- Department of Biology, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia; Health and Basic Science Research Centre, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Suhel Parvez
- Department of Toxicology, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
36
|
Bailey HM, Cookson MR. How Parkinson's Disease-Linked LRRK2 Mutations Affect Different CNS Cell Types. JOURNAL OF PARKINSON'S DISEASE 2024; 14:1331-1352. [PMID: 38905056 PMCID: PMC11492021 DOI: 10.3233/jpd-230432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/12/2024] [Indexed: 06/23/2024]
Abstract
LRRK2 is a relatively common genetic risk factor for Parkinson's disease (PD), with six coding variants known to cause familial PD. Non-coding variation at the same locus is also associated with sporadic PD. LRRK2 plays a role in many different intracellular signaling cascades including those involved in endolysosomal function, cytoskeletal dynamics, and Ca2+ homeostasis. PD-causing LRRK2 mutations cause hyperactive LRRK2 kinase activity, resulting in altered cellular signaling. Importantly, LRRK2 is lowly expressed in neurons and prominently expressed in non-neuronal cells in the brain. In this review, we will summarize recent and novel findings on the effects of PD-causing LRRK2 mutations in different nervous system cell types. This review will also provide novel insight into future areas of research at the intersection of LRRK2 cell biology, cell type specificity, and PD.
Collapse
Affiliation(s)
- Hannah M. Bailey
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
37
|
Cui D, Chen Y, Ye B, Guo W, Wang D, He J. Natural products for the treatment of neurodegenerative diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155101. [PMID: 37778246 DOI: 10.1016/j.phymed.2023.155101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 08/29/2023] [Accepted: 09/17/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND Neurodegenerative diseases are among the most common diseases in older adults worldwide. Alzheimer's disease (AD) and Parkinson's disease (PD) are two of the most common neurodegenerative diseases, and are accompanied by cerebral cortical atrophy, neuronal loss, protein accumulation, and excessive accumulation of metal ions. Natural products exhibit outstanding performance in improving cerebral circulatory disorders, promoting cerebral haematoma absorption, repairing damaged nerve tissue, and improving damaged nerve function. In recent years, studies have shown that neuroinflammatory mechanisms and signalling pathways closely related to the occurrence and development of neurological diseases include microglial activation, nuclear factor-κB (NF-κB) pathway, mitogen activated protein kinases (MAPK) pathway, reactive oxygen pathway, nucleotide binding oligomerisation domain-like receptor protein3 (NLRP3) inflammasomes, toll-like receptor4 (TLR4) pathway, nuclear factor erythroid 2-related factor 2 (Nrf2)/hemeoxygenase-1 (HO-1) pathway, phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway, and intestinal flora. Therefore, this study considered the mechanism of neurological diseases as the starting point to review the mechanism of action of natural products in the prevention and treatment of AD and PD in recent years to provide a theoretical basis for clinical prevention and treatment. AIM Natural products are a promising source of novel lead structures that have long been used to treat various nervous system diseases. METHODOLOGY This review collected literature on neurological diseases and natural products from 2012 to 2022, which were mainly searched through databases such as ScienceDirect, Springer, PubMed, SciFinder, China National Knowledge Infrastructure (CNKI), Wanfang, Google Scholar, and Baidu Academic. The following keywords were searched: neurological disorders, natural products, signalling pathway, mechanism of action. RESULTS This review summarises the pathogenesis of degenerative neurological diseases, recent findings on natural products used in neurodegenerative diseases, and the molecular mechanisms underlying these effects.
Collapse
Affiliation(s)
- Donghan Cui
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University, West China Hospital, Chengdu 610041, China
| | - Yajuan Chen
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan 650500, China
| | - Bengui Ye
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China; Medical College of Tibet University, Lasa 850002, China
| | - Wenhao Guo
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center and State Key Laboratory of Biotherapy, Sichuan University, West China Hospital, Chengdu 610041, China.
| | - Dongdong Wang
- Centre for Metabolism, Obesity, and Diabetes Research, Department of Medicine, McMaster University, HSC 4N71, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada.
| | - Jun He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy Sichuan University, Chengdu 610041, China.
| |
Collapse
|
38
|
Zhang F, Yang D, Li J, Du C, Sun X, Li W, Liu F, Yang Y, Li Y, Fu L, Li R, Zhang CX. Synaptotagmin-11 regulates immune functions of microglia in vivo. J Neurochem 2023; 167:680-695. [PMID: 37924268 DOI: 10.1111/jnc.16003] [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: 04/11/2022] [Revised: 08/28/2023] [Accepted: 10/10/2023] [Indexed: 11/06/2023]
Abstract
Membrane trafficking pathways mediate key microglial activities such as cell migration, cytokine secretion, and phagocytosis. However, the underlying molecular mechanism remains poorly understood. Previously, we found that synaptotagmin-11 (Syt11), a non-Ca2+ -binding Syt associated with Parkinson's disease (PD) and schizophrenia, inhibits cytokine release and phagocytosis in primary microglia. Here we reported the in vivo function of Syt11 in microglial immune responses using an inducible microglia-specific Syt11-conditional-knockout (cKO) mouse strain. Syt11-cKO resulted in activation of microglia and elevated mRNA levels of IL-6, TNF-α, IL-1β, and iNOS in various brain regions under both resting state and LPS-induced acute inflammation state in adult mice. In a PD mouse model generated by microinjection of preformed α-synuclein fibrils into the striatum, a reduced number of microglia migrated toward the injection sites and an enhanced phagocytosis of α-synuclein fibrils by microglia were found in Syt11-cKO mice. To understand the molecular mechanism of Syt11 function, we identified its direct binding proteins vps10p-tail-interactor-1a (vti1a) and vti1b. The linker domain of Syt11 interacted with both proteins and a peptide derived from it competitively inhibited the interaction of Syt11 with vti1a/vti1b in vitro and in cells. Importantly, application of this peptide induced more cytokine secretion in wild-type microglia upon LPS treatment, phenocopying defects in Syt11 knockdown cells. Altogether, we propose that Syt11 inhibits microglial activation in vivo and regulates cytokine secretion through interactions with vti1a and vti1b.
Collapse
Affiliation(s)
- Feifan Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Dong Yang
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Jingchen Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Cuilian Du
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Xinran Sun
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Wanru Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Fengwei Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Yiwei Yang
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Yuhong Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Lei Fu
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Rena Li
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital and Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Claire Xi Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China
| |
Collapse
|
39
|
Bérard M, Martínez-Drudis L, Sheta R, El-Agnaf OMA, Oueslati A. Non-invasive systemic viral delivery of human alpha-synuclein mimics selective and progressive neuropathology of Parkinson's disease in rodent brains. Mol Neurodegener 2023; 18:91. [PMID: 38012703 PMCID: PMC10683293 DOI: 10.1186/s13024-023-00683-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Alpha-synuclein (α-syn) aggregation into proteinaceous intraneuronal inclusions, called Lewy bodies (LBs), is the neuropathological hallmark of Parkinson's disease (PD) and related synucleinopathies. However, the exact role of α-syn inclusions in PD pathogenesis remains elusive. This lack of knowledge is mainly due to the absence of optimal α-syn-based animal models that recapitulate the different stages of neurodegeneration. METHODS Here we describe a novel approach for a systemic delivery of viral particles carrying human α-syn allowing for a large-scale overexpression of this protein in the mouse brain. This approach is based on the use of a new generation of adeno-associated virus (AAV), AAV-PHP.eB, with an increased capacity to cross the blood-brain barrier, thus offering a viable tool for a non-invasive and large-scale gene delivery in the central nervous system. RESULTS Using this model, we report that widespread overexpression of human α-syn induced selective degeneration of dopaminergic (DA) neurons, an exacerbated neuroinflammatory response in the substantia nigra and a progressive manifestation of PD-like motor impairments. Interestingly, biochemical analysis revealed the presence of insoluble α-syn oligomers in the midbrain. Together, our data demonstrate that a single non-invasive systemic delivery of viral particles overexpressing α-syn prompted selective and progressive neuropathology resembling the early stages of PD. CONCLUSIONS Our new in vivo model represents a valuable tool to study the role of α-syn in PD pathogenesis and in the selective vulnerability of nigral DA neurons; and offers the opportunity to test new strategies targeting α-syn toxicity for the development of disease-modifying therapies for PD and related disorders.
Collapse
Affiliation(s)
- Morgan Bérard
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Laura Martínez-Drudis
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Razan Sheta
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Omar M A El-Agnaf
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, 34110, Qatar
| | - Abid Oueslati
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, Canada.
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada.
| |
Collapse
|
40
|
Wang Q, Ruan Z, Jing L, Guo Z, Zhang X, Liu J, Tian L, Sun W, Song S, Hong JS, Shih YYI, Hou L, Wang Q. Complement receptor 3-mediated neurotoxic glial activation contributes to rotenone-induced cognitive decline in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 266:115550. [PMID: 37832486 PMCID: PMC10807506 DOI: 10.1016/j.ecoenv.2023.115550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 10/15/2023]
Abstract
Microglia-mediated chronic neuroinflammation has been associated with cognitive decline induced by rotenone, a well-known neurotoxic pesticide used in agriculture. However, the mechanisms remain unclear. This work aimed to elucidate the role of complement receptor 3 (CR3), a highly expressed receptor in microglia, in cognitive deficits induced by rotenone. Rotenone up-regulated the expression of CR3 in the hippocampus and cortex area of mice. CR3 deficiency markedly ameliorated rotenone-induced cognitive impairments, neurodegeneration and phosphorylation (Ser129) of α-synuclein in mice. CR3 deficiency also attenuated rotenone-stimulated microglial M1 activation. In microglial cells, siRNA-mediated knockdown of CR3 impeded, while CR3 activation induced by LL-37 exacerbated, rotenone-induced microglial M1 activation. Mechanistically, CR3 deficiency blocked rotenone-induced activation of nuclear factor κB (NF-κB), signal transducer and activator of transcription 1 (STAT1) and STAT3 signaling pathways. Pharmacological inhibition of NF-κB or STAT3 but not STAT1 was confirmed to suppress microglial M1 activation elicited by rotenone. Further study revealed that CR3 deficiency or knockdown also reduced rotenone-induced expression of C3, an A1 astrocyte marker, and production of microglial C1q, TNFα and IL-1α, a cocktail for activated microglia to induce neurotoxic A1 astrocytes, via NF-κB and STAT3 pathways. Finally, a small molecule modulator of CR3 efficiently mitigated rotenone-elicited cognitive deficits in mice even administered after the establishment of cognitive dysfunction. Taken together, our findings demonstrated that CR3 is a key factor in mediating neurotoxic glial activation and subsequent cognitive impairments in rotenone-treated mice, giving novel insights into the immunopathogenesis of cognitive impairments in pesticide-related Parkinsonism.
Collapse
Affiliation(s)
- Qinghui Wang
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
- Department of Anesthesiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116023, China
| | - Zhengzheng Ruan
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Lu Jing
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Ziyang Guo
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Xiaomeng Zhang
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Jianing Liu
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Lu Tian
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Wei Sun
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Sheng Song
- Biomedical Research Imaging Center, University of North Caroline at Chapel Hill, Chapel Hill, NC, USA
| | - Jau-Shyong Hong
- Neuropharmacology Section, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Yen-Yu Ian Shih
- Biomedical Research Imaging Center, University of North Caroline at Chapel Hill, Chapel Hill, NC, USA
| | - Liyan Hou
- Dalian Medical University Library, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
| | - Qingshan Wang
- School of Public Health, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian 116044, China
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
41
|
Fu D, Liang X, Jiang Y, Liu J, Lin X, Yang Q, Chen X, Huang P, Wang W, Wu W. Iron blocks autophagic flux and induces autophagosomes accumulation in microglia. Food Chem Toxicol 2023; 181:114054. [PMID: 37777083 DOI: 10.1016/j.fct.2023.114054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/07/2023] [Accepted: 09/18/2023] [Indexed: 10/02/2023]
Abstract
Iron is an essential dietary micronutrient for maintaining physiological homeostasis. However, disruption of cerebral iron regulation with the accumulation of iron in different brain structures appears to have a role in the pathogenesis of various neurodegenerative disorders. Studies have reported that autophagy induction could potentially mitigate progression in neurodegenerative diseases with iron deposition, but the relationship between autophagy and iron remains poorly understood. Meanwhile, abnormal autophagy in microglia is closely related to the occurrence of neurodegenerative diseases. Therefore, the effect of iron on microglia autophagy needs to be elaborated. In the present study, we found that iron induces autophagosome accumulation but inhibits its initiation in an Akt-mTOR pathway independent manner. Meanwhile, it caused autophagy flux defects and dysfunction of lysosomes. We also found that iron overload reduced the expression of Rab7, which is an essential protein for the fusion of autophagosomes and lysosomes. These results suggest that iron induces the accumulation of autophagosome in microglia and disrupts the autophagic flux in late stage of autophagy. Therefore, our work provides new insights into the molecular mechanisms of iron neurotoxicity.
Collapse
Affiliation(s)
- Deqiang Fu
- Department of Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Xingyue Liang
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China; School of Advance Manufacture, Fuzhou University, Jinjiang, 362251, China
| | - Yuxuan Jiang
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China
| | - Jieping Liu
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China
| | - Xiaosi Lin
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China
| | - Quan Yang
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China
| | - Xue Chen
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China
| | - Ping Huang
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China
| | - Wei Wang
- Institute of Oceanography, Minjiang University, Fuzhou, 350108, China.
| | - Wenlin Wu
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 362000, China; School of Advance Manufacture, Fuzhou University, Jinjiang, 362251, China.
| |
Collapse
|
42
|
Yu H, Chang Q, Sun T, He X, Wen L, An J, Feng J, Zhao Y. Metabolic reprogramming and polarization of microglia in Parkinson's disease: Role of inflammasome and iron. Ageing Res Rev 2023; 90:102032. [PMID: 37572760 DOI: 10.1016/j.arr.2023.102032] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
Parkinson's disease (PD) is a slowly progressive neurodegenerative disease characterized by α-synuclein aggregation and dopaminergic neuronal death. Recent evidence suggests that neuroinflammation is an early event in the pathogenesis of PD. Microglia are resident immune cells in the central nervous system that can be activated into either pro-inflammatory M1 or anti-inflammatory M2 phenotypes as found in peripheral macrophages. To exert their immune functions, microglia respond to various stimuli, resulting in the flexible regulation of their metabolic pathways. Inflammasomes activation in microglia induces metabolic shift from oxidative phosphorylation to glycolysis, and leads to the polarization of microglia to pro-inflammatory M1 phenotype, finally causing neuroinflammation and neurodegeneration. In addition, iron accumulation induces microglia take an inflammatory and glycolytic phenotype. M2 phenotype microglia is more sensitive to ferroptosis, inhibition of which can attenuate neuroinflammation. Therefore, this review highlights the interplay between microglial polarization and metabolic reprogramming of microglia. Moreover, it will interpret how inflammasomes and iron regulate microglial metabolism and phenotypic shifts, which provides a promising therapeutic target to modulate neuroinflammation and neurodegeneration in PD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Haiyang Yu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China; Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Qing Chang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China; Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Tong Sun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xin He
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Lulu Wen
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Jing An
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China.
| | - Yuhong Zhao
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China; Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China; Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
43
|
Van Laar AD, Webb KR, Keeney MT, Van Laar VS, Zharikov A, Burton EA, Hastings TG, Glajch KE, Hirst WD, Greenamyre JT, Rocha EM. Transient exposure to rotenone causes degeneration and progressive parkinsonian motor deficits, neuroinflammation, and synucleinopathy. NPJ Parkinsons Dis 2023; 9:121. [PMID: 37567894 PMCID: PMC10421849 DOI: 10.1038/s41531-023-00561-6] [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: 10/25/2022] [Accepted: 07/17/2023] [Indexed: 08/13/2023] Open
Abstract
Individuals with Parkinson's disease (PD) typically receive a diagnosis once they have developed motor symptoms, at which point there is already significant loss of substantia nigra dopamine neurons, α-synuclein accumulation in surviving neurons, and neuroinflammation. Consequently, the point of clinical presentation may be too late to initiate disease-modifying therapy. In contrast to this clinical reality, animal models often involve acute neurodegeneration and potential therapies are tested concurrently or shortly after the pathogenic insult has begun rather than later when diagnostic clinical symptoms emerge. Therefore, we sought to develop a model that reflects the clinical situation more accurately. Middle-aged rats (7-9 months-old) received a single daily intraperitoneal injection of rotenone for 5 consecutive days and were observed over the next 8-9 months. Rotenone-treated rats showed transient motor slowing and postural instability during exposure but recovered within 9 days of rotenone cessation. Rats remained without behavioral deficits for 3-4 months, then developed progressive motor abnormalities over the ensuing months. As motor abnormalities began to emerge 3 months after rotenone exposure, there was significant loss of nigral dopaminergic neurons and significant microglial activation. There was delayed accumulation of α-synuclein in neurons of the substantia nigra and frontal cortex, which was maximal at 9 months post-rotenone. In summary, a brief temporally-remote exposure to rotenone causes delayed and progressive behavioral and neuropathological changes similar to Parkinson's disease. This model mimics the human clinical situation, in which pathogenesis is well-established by the time diagnostic motor deficits appear. As such, this model may provide a more relevant experimental system in which to test disease-modifying therapeutics.
Collapse
Affiliation(s)
- Amber D Van Laar
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Katherine R Webb
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew T Keeney
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Victor S Van Laar
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alevtina Zharikov
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Edward A Burton
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA
| | - Teresa G Hastings
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kelly E Glajch
- Neurodegenerative Diseases Research Unit, Biogen, Cambridge, MA, 02142, USA
| | - Warren D Hirst
- Neurodegenerative Diseases Research Unit, Biogen, Cambridge, MA, 02142, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Emily M Rocha
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
44
|
Nascimento GC, Santos BM, Pedrazzi JF, Silva-Amaral D, Bortolanza M, Harris GT, Del Bel E, Branco LG. Effects of hydrogen gas inhalation on L-DOPA-induced dyskinesia. Brain Behav Immun Health 2023; 30:100623. [PMID: 37096172 PMCID: PMC10121822 DOI: 10.1016/j.bbih.2023.100623] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/07/2023] [Accepted: 04/07/2023] [Indexed: 04/26/2023] Open
Abstract
L-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesia is a side effect of Parkinson's disease treatment and it is characterized by atypical involuntary movements. A link between neuroinflammation and L-DOPA-induced dyskinesia has been documented. Hydrogen gas (H2) has neuroprotective effects in Parkinson's disease models and has a major anti-inflammatory effect. Our objective is to test the hypothesis that H2 inhalation reduces L-DOPA-induced dyskinesia. 15 days after 6-hydroxydopamine lesions of dopaminergic neurons were made (microinjection into the medial forebrain bundle), chronic L-DOPA treatment (15 days) was performed. Rats were exposed to H2 (2% gas mixture, 1 h) or air (controls) before L-DOPA injection. Abnormal involuntary movements and locomotor activity were conducted. Striatal microglia and astrocyte was analyzed and striatal and plasma samples for cytokines evaluation were collected after the abnormal involuntary movements analysis. H2 inhalation attenuated L-DOPA-induced dyskinesia. The gas therapy did not impair the improvement of locomotor activity achieved by L-DOPA treatment. H2 inhalation reduced activated microglia in the lesioned striatum, which is consistent with the observed reduced pro-inflammatory cytokines levels. Display of abnormal involuntary movements was positively correlated with plasma IL-1β and striatal TNF-α levels and negatively correlated with striatal IL-10 levels. Prophylactic H2 inhalation decreases abnormal involuntary movements in a preclinical L-DOPA-induced dyskinesia model. The H2 antidyskinetic effect was associated with decreased striatal and peripheral inflammation. This finding has a translational importance to L-DOPA-treated parkinsonian patients' well-being.
Collapse
Affiliation(s)
- Glauce C. Nascimento
- Department of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
- Department of Basic and Oral Biology, Ribeirão Preto Dentistry Faculty, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Bruna M. Santos
- Department of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
- Thermoregulation and Systemic Inflammation Laboratory (FeverLab), St. Josephs' Hospital and Medical Center, Phoenix, AZ, USA
| | - João F. Pedrazzi
- Neuroscience Graduate Program, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Danyelle Silva-Amaral
- Department of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Mariza Bortolanza
- Department of Basic and Oral Biology, Ribeirão Preto Dentistry Faculty, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Grant T. Harris
- Thermoregulation and Systemic Inflammation Laboratory (FeverLab), St. Josephs' Hospital and Medical Center, Phoenix, AZ, USA
| | - Elaine Del Bel
- Department of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
- Department of Basic and Oral Biology, Ribeirão Preto Dentistry Faculty, University of São Paulo, Ribeirão Preto, SP, Brazil
- Neuroscience Graduate Program, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Luiz G.S. Branco
- Department of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
- Department of Basic and Oral Biology, Ribeirão Preto Dentistry Faculty, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
45
|
Wu Y, Dong Z, Jiang X, Qu L, Zhou W, Sun X, Hou J, Xu H, Cheng M. Gut Microbiota Taxon-Dependent Transformation of Microglial M1/M2 Phenotypes Underlying Mechanisms of Spatial Learning and Memory Impairment after Chronic Methamphetamine Exposure. Microbiol Spectr 2023; 11:e0030223. [PMID: 37212669 PMCID: PMC10269813 DOI: 10.1128/spectrum.00302-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/06/2023] [Indexed: 05/23/2023] Open
Abstract
Methamphetamine (METH) exposure may lead to cognitive impairment. Currently, evidence suggests that METH exposure alters the configuration of the gut microbiota. However, the role and mechanism of the gut microbiota in cognitive impairment after METH exposure are still largely unknown. Here, we investigated the impact of the gut microbiota on the phenotype status of microglia (microglial phenotypes M1 and microglial M2) and their secreting factors, the subsequent hippocampal neural processes, and the resulting influence on spatial learning and memory of chronically METH-exposed mice. We determined that gut microbiota perturbation triggered the transformation of microglial M2 to M1 and a subsequent change of pro-brain-derived neurotrophic factor (proBDNF)-p75NTR-mature BDNF (mBDNF)-TrkB signaling, which caused reduction of hippocampal neurogenesis and synaptic plasticity-related proteins (SYN, PSD95, and MAP2) and, consequently, deteriorated spatial learning and memory. More specifically, we found that Clostridia, Bacteroides, Lactobacillus, and Muribaculaceae might dramatically affect the homeostasis of microglial M1/M2 phenotypes and eventually contribute to spatial learning and memory decline after chronic METH exposure. Finally, we found that fecal microbial transplantation could protect against spatial learning and memory decline by restoring the microglial M1/M2 phenotype status and the subsequent proBDNF-p75NTR/mBDNF-TrkB signaling in the hippocampi of chronically METH-exposed mice. IMPORTANCE Our study indicated that the gut microbiota contributes to spatial learning and memory dysfunction after chronic METH exposure, in which microglial phenotype status plays an intermediary role. The elucidated "specific microbiota taxa-microglial M1/M2 phenotypes-spatial learning and memory impairment" pathway would provide a novel mechanism and elucidate potential gut microbiota taxon targets for the no-drug treatment of cognitive deterioration after chronic METH exposure.
Collapse
Affiliation(s)
- Yulong Wu
- Department of Pathogenic Biology, Binzhou Medical University, Yantai, China
| | - Zhouyan Dong
- Department of Pathogenic Biology, Binzhou Medical University, Yantai, China
| | - Xinze Jiang
- Department of Pathogenic Biology, Binzhou Medical University, Yantai, China
| | - Lei Qu
- Department of Pathogenic Biology, Binzhou Medical University, Yantai, China
| | - Wei Zhou
- Department of Health and Disease Management, Binzhou Medical University, Yantai, China
| | - Xu Sun
- Department of Health and Disease Management, Binzhou Medical University, Yantai, China
| | - Jiangshan Hou
- Department of Pathogenic Biology, Binzhou Medical University, Yantai, China
| | - Hongmei Xu
- Department of Health and Disease Management, Binzhou Medical University, Yantai, China
| | - Mei Cheng
- Department of Health and Disease Management, Binzhou Medical University, Yantai, China
| |
Collapse
|
46
|
Guan S, Sun L, Wang X, Huang X, Luo T. Propofol inhibits neuroinflammation and metabolic reprogramming in microglia in vitro and in vivo. Front Pharmacol 2023; 14:1161810. [PMID: 37383725 PMCID: PMC10293632 DOI: 10.3389/fphar.2023.1161810] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/12/2023] [Indexed: 06/30/2023] Open
Abstract
Microglial activation-induced neuroinflammation is closely related to the development of sepsis-associated encephalopathy. Accumulating evidence suggests that changes in the metabolic profile of microglia is crucial for their response to inflammation. Propofol is widely used for sedation in mechanically ventilated patients with sepsis. Here, we investigate the effect of propofol on lipopolysaccharide-induced neuroinflammation, neuronal injuries, microglia metabolic reprogramming as well as the underlying molecular mechanisms. The neuroprotective effects of propofol (80 mg/kg) in vivo were measured in the lipopolysaccharide (2 mg/kg)-induced sepsis in mice through behavioral tests, Western blot analysis and immunofluorescent staining. The anti-inflammatory effects of propofol (50 μM) in microglial cell cultures under lipopolysaccharide (10 ng/ml) challenge were examined with Seahorse XF Glycolysis Stress test, ROS assay, Western blot, and immunofluorescent staining. We showed that propofol treatment reduced microglia activation and neuroinflammation, inhibited neuronal apoptosis and improved lipopolysaccharide-induced cognitive dysfunction. Propofol also attenuated lipopolysaccharide-stimulated increases of inducible nitric oxide synthase, nitric oxide, tumor necrosis factor-α, interlukin-1β and COX-2 in cultured BV-2 cells. Propofol-treated microglia showed a remarkable suppression of lipopolysaccharide-induced HIF-1α, PFKFB3, HK2 expression and along with downregulation of the ROS/PI3K/Akt/mTOR signaling pathway. Moreover, propofol attenuated the enhancement of mitochondrial respiration and glycolysis induced by lipopolysaccharide. Together, our data suggest that propofol attenuated inflammatory response by inhibiting metabolic reprogramming, at least in part, through downregulation of the ROS/PI3K/Akt/mTOR/HIF-1α signaling pathway.
Collapse
|
47
|
Conte C, Ingrassia A, Breve J, Bol JJ, Timmermans-Huisman E, van Dam AM, Beccari T, van de Berg WDJ. Toll-like Receptor 4 Is Upregulated in Parkinson's Disease Patients and Co-Localizes with pSer129αSyn: A Possible Link with the Pathology. Cells 2023; 12:1368. [PMID: 37408202 DOI: 10.3390/cells12101368] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/03/2023] [Accepted: 05/10/2023] [Indexed: 07/07/2023] Open
Abstract
Growing evidence suggests a crucial role of neuroinflammation in the pathophysiology of Parkinson's disease (PD). Neuroinflammation is linked to the accumulation and aggregation of a-synuclein (αSyn), the primary pathological hallmark of PD. Toll-like receptors 4 (TLR4) can have implications in the development and progression of the pathology. In this study, we analyzed the expression of TLR4 in the substantia nigra (SN) and medial temporal gyrus (GTM) of well-characterized PD patients and age-matched controls. We also assessed the co-localization of TLR4 with pSer129 αSyn. Using qPCR, we observed an upregulation of TLR4 expression in the SN and GTM in PD patients compared to controls, which was accompanied by a reduction in αSyn expression likely due to the depletion of dopaminergic (DA) cells. Additionally, using immunofluorescence and confocal microscopy, we observed TLR4-positive staining and co-localization with pSer129-αSyn in Lewy bodies of DA neurons in the SN, as well as in pyramidal neurons in the GTM of PD donors. Furthermore, we observed a co-localization of TLR4 and Iba-1 in glial cells of both SN and GTM. Our findings provide evidence for the increased expression of TLR4 in the PD brain and suggest that the interaction between TLR4 and pSer129-αSyn could play a role in mediating the neuroinflammatory response in PD.
Collapse
Affiliation(s)
- Carmela Conte
- Department of Pharmaceutical Sciences, University of Perugia, 06100 Perugia, Italy
| | - Angela Ingrassia
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - John Breve
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - John J Bol
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Evelien Timmermans-Huisman
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Anne-Marie van Dam
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Tommaso Beccari
- Department of Pharmaceutical Sciences, University of Perugia, 06100 Perugia, Italy
| | - Wilma D J van de Berg
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
48
|
Sabari SS, Balasubramani K, Iyer M, Sureshbabu HW, Venkatesan D, Gopalakrishnan AV, Narayanaswamy A, Senthil Kumar N, Vellingiri B. Type 2 Diabetes (T2DM) and Parkinson's Disease (PD): a Mechanistic Approach. Mol Neurobiol 2023:10.1007/s12035-023-03359-y. [PMID: 37118323 PMCID: PMC10144908 DOI: 10.1007/s12035-023-03359-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/19/2023] [Indexed: 04/30/2023]
Abstract
Growing evidence suggest that there is a connection between Parkinson's disease (PD) and insulin dysregulation in the brain, whilst the connection between PD and type 2 diabetes mellitus (T2DM) is still up for debate. Insulin is widely recognised to play a crucial role in neuronal survival and brain function; any changes in insulin metabolism and signalling in the central nervous system (CNS) can lead to the development of various brain disorders. There is accumulating evidence linking T2DM to PD and other neurodegenerative diseases. In fact, they have a lot in common patho-physiologically, including insulin dysregulation, oxidative stress resulting in mitochondrial dysfunction, microglial activation, and inflammation. As a result, initial research should focus on the role of insulin and its molecular mechanism in order to develop therapeutic outcomes. In this current review, we will look into the link between T2DM and PD, the function of insulin in the brain, and studies related to impact of insulin in causing T2DM and PD. Further, we have also highlighted the role of various insulin signalling pathway in both T2DM and PD. We have also suggested that T2DM-targeting pharmacological strategies as potential therapeutic approach for individuals with cognitive impairment, and we have demonstrated the effectiveness of T2DM-prescribed drugs through current PD treatment trials. In conclusion, this investigation would fill a research gap in T2DM-associated Parkinson's disease (PD) with a potential therapy option.
Collapse
Affiliation(s)
- S Sri Sabari
- Department of Zoology, School of Basic Sciences, Stem Cell and Regenerative Medicine/Translational Research, Central University of Punjab (CUPB), Bathinda, 151401, Punjab, India
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Kiruthika Balasubramani
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Mahalaxmi Iyer
- Department of Biotechnology, Karpagam Academy of Higher Education (Deemed to Be University), Coimbatore, 641021, Tamil Nadu, India
| | - Harysh Winster Sureshbabu
- Department of Zoology, School of Basic Sciences, Stem Cell and Regenerative Medicine/Translational Research, Central University of Punjab (CUPB), Bathinda, 151401, Punjab, India
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Dhivya Venkatesan
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, 632 014, India
| | - Arul Narayanaswamy
- Department of Zoology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India
| | - Nachimuthu Senthil Kumar
- Department of Biotechnology, Mizoram University (A Central University), Aizawl, 796004, Mizoram, India
| | - Balachandar Vellingiri
- Department of Zoology, School of Basic Sciences, Stem Cell and Regenerative Medicine/Translational Research, Central University of Punjab (CUPB), Bathinda, 151401, Punjab, India.
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, 641 046, Tamil Nadu, India.
| |
Collapse
|
49
|
Kopp KO, Greer ME, Glotfelty EJ, Hsueh SC, Tweedie D, Kim DS, Reale M, Vargesson N, Greig NH. A New Generation of IMiDs as Treatments for Neuroinflammatory and Neurodegenerative Disorders. Biomolecules 2023; 13:747. [PMID: 37238617 PMCID: PMC10216254 DOI: 10.3390/biom13050747] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
The immunomodulatory imide drug (IMiD) class, which includes the founding drug member thalidomide and later generation drugs, lenalidomide and pomalidomide, has dramatically improved the clinical treatment of specific cancers, such as multiple myeloma, and it combines potent anticancer and anti-inflammatory actions. These actions, in large part, are mediated by IMiD binding to the human protein cereblon that forms a critical component of the E3 ubiquitin ligase complex. This complex ubiquitinates and thereby regulates the levels of multiple endogenous proteins. However, IMiD-cereblon binding modifies cereblon's normal targeted protein degradation towards a new set of neosubstrates that underlies the favorable pharmacological action of classical IMiDs, but also their adverse actions-in particular, their teratogenicity. The ability of classical IMiDs to reduce the synthesis of key proinflammatory cytokines, especially TNF-α levels, makes them potentially valuable to reposition as drugs to mitigate inflammatory-associated conditions and, particularly, neurological disorders driven by an excessive neuroinflammatory element, as occurs in traumatic brain injury, Alzheimer's and Parkinson's diseases, and ischemic stroke. The teratogenic and anticancer actions of classical IMiDs are substantial liabilities for effective drugs in these disorders and can theoretically be dialed out of the drug class. We review a select series of novel IMiDs designed to avoid binding with human cereblon and/or evade degradation of downstream neosubstrates considered to underpin the adverse actions of thalidomide-like drugs. These novel non-classical IMiDs hold potential as new medications for erythema nodosum leprosum (ENL), a painful inflammatory skin condition associated with Hansen's disease for which thalidomide remains widely used, and, in particular, as a new treatment strategy for neurodegenerative disorders in which neuroinflammation is a key component.
Collapse
Affiliation(s)
- Katherine O. Kopp
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
| | - Margaret E. Greer
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
- Faculty of Medicine, Georgetown University School of Medicine, Washington, DC 20007, USA
| | - Elliot J. Glotfelty
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Shih-Chang Hsueh
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
| | - Dong Seok Kim
- Aevisbio Inc., Gaithersburg, MD 20878, USA
- Aevis Bio Inc., Daejeon 34141, Republic of Korea
| | - Marcella Reale
- Department of Innovative Technologies in Medicine and Dentistry, G. d’Annunzio University of Chieti and Pescara, 66100 Chieti, Italy
| | - Neil Vargesson
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
| |
Collapse
|
50
|
Isik S, Yeman Kiyak B, Akbayir R, Seyhali R, Arpaci T. Microglia Mediated Neuroinflammation in Parkinson’s Disease. Cells 2023; 12:cells12071012. [PMID: 37048085 PMCID: PMC10093562 DOI: 10.3390/cells12071012] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Parkinson’s Disease (PD) is the second most common neurodegenerative disorder seen, especially in the elderly. Tremor, shaking, movement problems, and difficulty with balance and coordination are among the hallmarks, and dopaminergic neuronal loss in substantia nigra pars compacta of the brain and aggregation of intracellular protein α-synuclein are the pathological characterizations. Neuroinflammation has emerged as an involving mechanism at the initiation and development of PD. It is a complex network of interactions comprising immune and non-immune cells in addition to mediators of the immune response. Microglia, the resident macrophages in the CNS, take on the leading role in regulating neuroinflammation and maintaining homeostasis. Under normal physiological conditions, they exist as “homeostatic” but upon pathological stimuli, they switch to the “reactive state”. Pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes are used to classify microglial activity with each phenotype having its own markers and released mediators. When M1 microglia are persistent, they will contribute to various inflammatory diseases, including neurodegenerative diseases, such as PD. In this review, we focus on the role of microglia mediated neuroinflammation in PD and also signaling pathways, receptors, and mediators involved in the process, presenting the studies that associate microglia-mediated inflammation with PD. A better understanding of this complex network and interactions is important in seeking new therapies for PD and possibly other neurodegenerative diseases.
Collapse
Affiliation(s)
- Sevim Isik
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Uskudar University, Uskudar, Istanbul 34662, Turkey
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Correspondence: ; Tel.: +90-216-400-2222 (ext. 2462)
| | - Bercem Yeman Kiyak
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Medicine, Institute of Hamidiye Health Sciences, University of Health Sciences, Uskudar, Istanbul 34668, Turkey
| | - Rumeysa Akbayir
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Biology, Institute of Science, Uskudar University, Uskudar, Istanbul 34662, Turkey
| | - Rama Seyhali
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Biology, Institute of Science, Uskudar University, Uskudar, Istanbul 34662, Turkey
| | - Tahire Arpaci
- Stem Cell Research and Application Center (USKOKMER), Uskudar University, Uskudar, Istanbul 34662, Turkey
- Department of Molecular Biology, Institute of Science, Uskudar University, Uskudar, Istanbul 34662, Turkey
| |
Collapse
|