Nanowired delivery of dl-3-n-butylphthalide with antibodies to alpha synuclein potentiated neuroprotection in Parkinson’s disease with emotional stress

International Review of Neurobiology - Tập 171 - Trang 47-82 - 2023
Lianyuan Feng1, Aruna Sharma2, Zhenguo Wang3, Dafin F. Muresanu4,5, Z. Ryan Tian6, José Vicente Lafuente7, Anca D. Buzoianu8, Ala Nozari9, Cong Li10, Ziquiang Zhang10, Chen Lin11, Hongyun Huang12, Igor Manzhulo13, Lars Wiklund2, Hari Shanker Sharma2
1Department of Neurology, Bethune International Peace Hospital, Zhongshan Road (West), Shijiazhuang, Hebei Province, P.R. China
2International Experimental Central Nervous System Injury & Repair (IECNSIR), Dept. of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
3Shijiazhuang Pharma Group NBP Pharmaceutical Co. Ltd., Economic and Technological Development Zone, Shijiazhuang City, Hebei Province, P.R. China
4Dept. Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania
5”RoNeuro” Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
6Dept. Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
7LaNCE, Dept. Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
8Department of Clinical Pharmacology and Toxicology, “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
9Department of Anesthesiology, Boston University, Albany str, Boston, MA, United States
10Department of Neurosurgery, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province Hospital of Chinese Medical, Guangzhou, Guangdong, P.R. China
11Department of Neurosurgery, Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing, P.R. China
12Beijing Hongtianji Neuroscience Academy, Beijing, P.R. China
13Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia

Tài liệu tham khảo

Arslan, 2020, The cerebral blood flow deficits in Parkinson’s disease with mild cognitive impairment using arterial spin labeling MRI, Journal of Neural Transmission (Vienna), 127, 1285, 10.1007/s00702-020-02227-6 Badanjak, 2021, The contribution of microglia to neuroinflammation in Parkinson’s disease, International Journal of Molecular Sciences, 22, 4676, 10.3390/ijms22094676 Belova, 1982, Blood-brain barrier permeability and immobilization stress, Acta Physiologica Scandinavica, 116, 21, 10.1111/j.1748-1716.1982.tb10594.x Biju, 2020, Reduced cerebral blood flow in an α-synuclein transgenic mouse model of Parkinson’s disease, Journal of Cerebral Blood Flow and Metabolism: Official Journal of the International Society of Cerebral Blood Flow and Metabolism, 40, 2441, 10.1177/0271678X19895432 Bini, 2022, Stress-level glucocorticoids increase fasting hunger and decrease cerebral blood flow in regions regulating eating, NeuroImage: Clinical, 36 Blakemore, 2018, Stress-evoking emotional stimuli exaggerate deficits in motor function in Parkinson’s disease, Neuropsychologia, 112, 66, 10.1016/j.neuropsychologia.2018.03.006 Blandini, 2012, Animal models of Parkinson’s disease, The FEBS Journal, 279, 1156, 10.1111/j.1742-4658.2012.08491.x Burke, 2003, 3,4-dihydroxyphenylacetaldehyde: a potential target for neuroprotective therapy in Parkinson’s disease, Current Drug Targets. CNS and Neurological Disorders, 2, 143, 10.2174/1568007033482913 Burtscher, 2019, Chronic corticosterone aggravates behavioral and neuronal symptomatology in a mouse model of alpha-synuclein pathology, Neurobiology of Aging, 83, 11, 10.1016/j.neurobiolaging.2019.08.007 Chen, 2021, Cerebral edema formation after stroke: Emphasis on blood-brain barrier and the lymphatic drainage system of the brain, Frontiers in Cellular Neuroscience, 15, 10.3389/fncel.2021.716825 Chen, 2008, Caffeine protects against MPTP-induced blood-brain barrier dysfunction in mouse striatum, Journal of Neurochemistry, 107, 1147, 10.1111/j.1471-4159.2008.05697.x Chen, 2019, Application and prospects of butylphthalide for the treatment of neurologic diseases, Chinese Medical Journal (Engl), 132, 1467, 10.1097/CM9.0000000000000289 Cunha, 2017, MPP+-lesioned mice: An experimental model of motor, emotional, memory/learning, and striatal neurochemical dysfunctions, Molecular Neurobiology, 54, 6356, 10.1007/s12035-016-0147-1 Das, 2000, Immobilization stress-induced changes in brain acetylcholinesterase activity and cognitive function in mice, Pharmacological Research: The Official Journal of the Italian Pharmacological Society, 42, 213, 10.1006/phrs.2000.0678 Date, 2009, Neurological disorders and neural regeneration, with special reference to Parkinson’s disease and cerebral ischemia, Journal of Artificial Organs: The Official Journal of the Japanese Society for Artificial Organs, 12, 11, 10.1007/s10047-008-0441-4 Datta, 2022, Post-stroke impairment of the blood-brain barrier and perifocal vasogenic edema is alleviated by endovascular mesenchymal stem cell administration: Modulation of the PKCδ/MMP9/AQP4-Mediated pathway, Molecular Neurobiology, 59, 2758, 10.1007/s12035-022-02761-2 Dionísio, 2021, Oxidative stress and regulated cell death in Parkinson’s disease, Ageing Research Reviews, 67, 10.1016/j.arr.2021.101263 Djamshidian, 2014, Can stress trigger Parkinson’s disease?, Journal of Neurology, Neurosurgery, and Psychiatry, 85, 878, 10.1136/jnnp-2013-305911 Dong, 2021, dl-3-n-butylphthalide pretreatment attenuates renal ischemia/reperfusion injury, Biochemical and Biophysical Research Communications, 557, 166, 10.1016/j.bbrc.2021.04.006 Dvorská, 1992, On the blood-brain barrier to peptides: Effects of immobilization stress on regional blood supply and accumulation of labelled peptides in the rat brain, Endocrine Regulations, 26, 77 Elliott, 1949, Measurement of experimentally induced brain swelling and shrinkage, The American Journal of Physiology, 157, 122, 10.1152/ajplegacy.1949.157.1.122 Esposito, 2002, Corticotropin-releasing hormone and brain mast cells regulate blood-brain-barrier permeability induced by acute stress, The Journal of Pharmacology and Experimental Therapeutics, 303, 1061, 10.1124/jpet.102.038497 Fahn, 2008, The history of dopamine and levodopa in the treatment of Parkinson’s disease, Movement Disorders: Official Journal of the Movement Disorder Society, 23, S497, 10.1002/mds.22028 Feng, 2018, TiO2-nanowired delivery of dl-3-n-butylphthalide (DL-NBP) attenuates blood-brain barrier disruption, brain edema formation, and neuronal damages following concussive head injury, Molecular Neurobiology, 55, 350, 10.1007/s12035-017-0746-5 Fongaro, 2022, 3,4-Dihydroxyphenylethanol and 3,4-dihydroxyphenylacetic acid affect the aggregation process of E46K variant of α-synuclein at different extent: Insights into the interplay between protein dynamics and catechol effect, Protein Science: A Publication of the Protein Society, 31, 10.1002/pro.4356 Golbidi, 2018, Oxidative stress: A unifying mechanism for cell damage induced by noise, (water-pipe) smoking, and emotional stress-therapeutic strategies targeting redox imbalance, Antioxidants & Redox Signaling, 28, 741, 10.1089/ars.2017.7257 Greenberg, 2014, Stress, PTSD, and dementia, Alzheimer’s & Dementia: The Journal of the Alzheimer’s Association, 10, S155 Gröger, 1990, Importance of protein content in the edema fluid for the resolution of brain edema, Advances in Neurology, 52, 215 Guo, 2022, Neurobiological Links between stress, brain injury, and disease, Oxidative Medicine and Cellular Longevity, 2022, 8111022, 10.1155/2022/8111022 Haavik, 1998, Tyrosine hydroxylase and Parkinson’s disease, Molecular Neurobiology, 16, 285, 10.1007/BF02741387 Hirsch, 2009, Neuroinflammation in Parkinson’s disease: A target for neuroprotection?, Lancet Neurology, 8, 382, 10.1016/S1474-4422(09)70062-6 Hosoi, 2019, Immobilization stress induces XBP1 splicing in the mouse brain, Biochemical and Biophysical Research Communications, 508, 516, 10.1016/j.bbrc.2018.11.167 Hu, 2023, dl-3-n-butylphthalide alleviates motor disturbance by suppressing ferroptosis in a rat model of Parkinson’s disease, Neural Regen Res, 18, 194, 10.4103/1673-5374.343892 Idrissi, 2023, Restraint stress exacerbates apoptosis in a 6-OHDA animal model of Parkinson disease, Neurotoxicity Research, 41, 166, 10.1007/s12640-022-00630-3 Issam, 2019, A simple approach to studying cerebral blood flow during psychological stress, Naunyn-Schmiedeberg’s Archives of Pharmacology, 392, 505, 10.1007/s00210-019-01638-x Jackson, 2014, Evaluating the role of Hans Selye in the modern history of stress Jackson-Lewis, 2007, Protocol for the MPTP mouse model of Parkinson’s disease, Nature Protocols, 2, 141, 10.1038/nprot.2006.342 Kang, 2021, Tumor necrosis factor inhibition and Parkinson disease: A Mendelian randomization study, Neurology, 96, e1672, 10.1212/WNL.0000000000011630 Kim, 2014, Alpha-synuclein interferes with cAMP/PKA-dependent upregulation of dopamine β-hydroxylase and is associated with abnormal adaptive responses to immobilization stress, Experimental Neurology, 252, 63, 10.1016/j.expneurol.2013.11.009 Kim, 2005, Immobilization stress causes increases in tetrahydrobiopterin, dopamine, and neuromelanin and oxidative damage in the nigrostriatal system, Journal of Neurochemistry, 95, 89, 10.1111/j.1471-4159.2005.03342.x Kiyatkin, 2019, Leakage of the blood-brain barrier followed by vasogenic edema as the ultimate cause of death induced by acute methamphetamine overdose, International Review of Neurobiology, 146, 189, 10.1016/bs.irn.2019.06.010 Klatzo, 1987, Blood-brain barrier and ischaemic brain oedema, Zeitschrift fur Kardiologie, 76, 67 Klatzo, 1987, Pathophysiological aspects of brain edema, Acta Neuropathologica, 72, 236, 10.1007/BF00691095 Kuroiwa, 1994, Biomechanical characteristics of brain edema: the difference between vasogenic-type and cytotoxic-type edema, Acta Neurochirurgica. Supplementum, 60, 158 Kurumaji, 2008, Effects of FG7142 and immobilization stress on the gene expression in the neocortex of mice, Neuroscience Research, 62, 155, 10.1016/j.neures.2008.08.001 Kvetnansky, 2016, Exaggerated phosphorylation of brain tau protein in CRH KO mice exposed to repeated immobilization stress, Stress (Amsterdam, Netherlands), 19, 395, 10.1080/10253890.2016.1183119 Lan, 2022, Astrocytic VEGFA: An essential mediator in blood-brain-barrier disruption in Parkinson’s disease, Glia, 70, 337, 10.1002/glia.24109 Lauretti, 2016, Chronic behavioral stress exaggerates motor deficit and neuroinflammation in the MPTP mouse model of Parkinson’s disease, Translational Psychiatry, 6, 10.1038/tp.2016.1 Li, 2022, CaMKIIα signaling is required for the neuroprotective effects of dl-3-n-butylphthalide in Alzheimer’s disease, Molecular Neurobiology, 59, 3370, 10.1007/s12035-022-02777-8 Liu, 2022, Biomarker of neuroinflammation in Parkinson’s disease, International Journal of Molecular Sciences, 23, 4148, 10.3390/ijms23084148 Liu, 2019, Memantine differentially regulates tau phosphorylation induced by chronic restraint stress of varying duration in mice, Neural Plasticity, 2019, 4168472, 10.1155/2019/4168472 Lu, 2021, The evolution of the concept of stress and the framework of the stress system, Cell Stress, 5, 76, 10.15698/cst2021.06.250 Lupien, 2009, Effects of stress throughout the lifespan on the brain, behaviour and cognition, Nature Reviews. Neuroscience, 10, 434, 10.1038/nrn2639 Ma, 2007, Functional imaging of cerebral blood flow and glucose metabolism in Parkinson’s disease and Huntington’s disease, Molecular Imaging and Biology: MIB: The Official Publication of the Academy of Molecular Imaging, 9, 223, 10.1007/s11307-007-0085-4 Marmarou, 1980, Biomechanics of brain edema and effects on local cerebral blood flow, Advances in Neurology, 28, 345 McCrodden, 1990, The neurotoxicity of MPTP and the relevance to Parkinson’s disease, Pharmacology & Toxicology, 67, 8, 10.1111/j.1600-0773.1990.tb00773.x McEwen, 2007, Physiology and neurobiology of stress and adaptation: Central role of the brain, Physiological Reviews, 87, 873, 10.1152/physrev.00041.2006 Mitsumoto, 2018, Acute restraint stress augments 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine neurotoxicity via increased toxin uptake into the brain in C57BL/6 mice, Neuroscience Bulletin, 34, 849, 10.1007/s12264-018-0254-2 Muhammad, 2022, Antioxidative role of traditional Chinese medicine in Parkinson’s disease, Journal of Ethnopharmacology, 285, 10.1016/j.jep.2021.114821 Mustapha, 2021, MPTP-induced mouse model of Parkinson’s disease: A promising direction of therapeutic strategies, Basic Medical Sciences, 21, 422 National Research Council (US); Committee for the Update of the Guide for the Care and Use of Laboratory Animals. (2011). Guide for the care and use of laboratory animals (8th ed.). Washington (DC): National Academies Press (US). The National Academies Collection: Reports funded by National Institutes of Health. Bookshelf ID: NBK54050. DOI: 10.17226/12910; 〈https://nap.nationalacademies.org/catalog/12910/guide-for-the-care-and-use-of-laboratory-animals-eighth〉. Ng, 2022, Microvascular dysfunction in blood-brain barrier disruption and hypoperfusion within the infarct posttreatment are associated with cerebral edema. EXTEND-IA TNK part 1 and 2 investigators, Stroke; A Journal of Cerebral Circulation, 53, 1597, 10.1161/STROKEAHA.121.036104 Ngema, 2017, Post 6-OHDA lesion exposure to stress affects neurotrophic factor expression and aggravates motor impairment, Metabolic Brain Disease, 32, 1061, 10.1007/s11011-017-9988-1 Niu, 2019, Nanowired delivery of dl-3-n-butylphthalide induces superior neuroprotection in concussive head injury, Progress in Brain Research, 245, 89, 10.1016/bs.pbr.2019.03.008 Niu, 2020, Co-administration of TiO2-nanowired dl-3-n-butylphthalide (dl-NBP) and mesenchymal stem cells enhanced neuroprotection in Parkinson’s disease exacerbated by concussive head injury, Progress in Brain Research, 258, 101, 10.1016/bs.pbr.2020.09.011 Niu, 2021, Nanodelivery of oxiracetam enhances memory, functional recovery and induces neuroprotection following concussive head injury, Progress in Brain Research, 265, 139, 10.1016/bs.pbr.2021.06.004 Nzou, 2018, Human cortex spheroid with a functional blood brain barrier for high-throughput neurotoxicity screening and disease modeling, Sci Rep, 8, 7413, 10.1038/s41598-018-25603-5 Ong, 2017, Reconsidering the role of glial cells in chronic stress-induced dopaminergic neurons loss within the substantia nigra? Friend or foe?, Brain, Behavior, and Immunity, 60, 117, 10.1016/j.bbi.2016.10.001 Ozkizilcik, 2018, Timed release of cerebrolysin using drug-loaded titanate nanospheres reduces brain pathology and improves behavioral functions in Parkinson’s disease, Molecular Neurobiology, 55, 359, 10.1007/s12035-017-0747-4 Ozkizilcik, 2019, Nanodelivery of cerebrolysin reduces pathophysiology of Parkinson’s disease, Progress in Brain Research, 245, 201, 10.1016/bs.pbr.2019.03.014 Pajares, 2020, Inflammation in Parkinson’s disease: Mechanisms and therapeutic implications, Cells, 9, 1687, 10.3390/cells9071687 Patnaik, 2018, Histamine H3 inverse agonist BF 2649 or antagonist with partial H4 agonist activity clobenpropit reduces amyloid beta peptide-induced brain pathology in Alzheimer’s disease, Molecular Neurobiology, 55, 312, 10.1007/s12035-017-0743-8 Phan, 2006, Corticolimbic blood flow during nontraumatic emotional processing in posttraumatic stress disorder, Archives of General Psychiatry, 63, 184, 10.1001/archpsyc.63.2.184 Pineda-Pardo, 2022, Striatal blood-brain barrier opening in Parkinson’s disease dementia: A pilot exploratory study, Movement Disorders: Official Journal of the Movement Disorder Society, 37, 2057, 10.1002/mds.29134 Ramanzini, 2022, Inflammatory markers and depression in Parkinson’s disease: A systematic review, Neurological Sciences: Official Journal of the Italian Neurological Society and of the Italian Society of Clinical Neurophysiology, 43, 6707, 10.1007/s10072-022-06363-7 Ramirez de Noriega, 2018, A swine model of intracellular cerebral edema - Cerebral physiology and intracranial compliance, Journal of Clinical Neuroscience: Official Journal of the Neurosurgical Society of Australasia, 58, 192, 10.1016/j.jocn.2018.10.051 Rausch, 2022, From the tyrosine hydroxylase hypothesis of Parkinson’s disease to modern strategies: A short historical overview, Journal of Neural Transmission (Vienna), 129, 487, 10.1007/s00702-022-02488-3 Riachi, 1990, Effects of internal carotid administration of MPTP on rat brain and blood-brain barrier, Brain Research, 533, 6, 10.1016/0006-8993(90)91788-I Riachi, 1991, Correlation of MPTP neurotoxicity in vivo with oxidation of MPTP by the brain and blood-brain barrier in vitro in five rat strains, Brain Research, 555, 19, 10.1016/0006-8993(91)90854-O Rochette, 2023, Stress: Eight decades after its definition by Hans Selye: Stress is the spice of life, Brain Sciences, 13, 310, 10.3390/brainsci13020310 Rom, 2016, The stress reaction: A historical perspective, Advances in Experimental Medicine and Biology, 905, 1 Salim, 2016, Oxidative stress: A potential link between emotional wellbeing and immune response, Current Opinion in Pharmacology, 29, 70, 10.1016/j.coph.2016.06.006 Sandyk, 1989, The neurotoxicity of MPTP and its relationship to the pathophysiology of Parkinson’s disease, The International Journal of Neuroscience, 44, 165, 10.3109/00207458908986194 Seegal, 1981, Selective activation of tuberoinfundibular dopaminergic neurons in the chronically restrained rat, Brain Research, 216, 460, 10.1016/0006-8993(81)90150-5 Segura-Aguilar, 2014, Protective and toxic roles of dopamine in Parkinson’s disease, Journal of Neurochemistry, 129, 898, 10.1111/jnc.12686 Selye, 1936, A syndrome produced by diverse nocuous agents, Nature, 138, 32, 10.1038/138032a0 Selye, 1955, Stress and disease, Science (New York, N. Y.), 122, 625, 10.1126/science.122.3171.625 Selye, 1959, The physiopathology of stress, Postgraduate Medicine, 25, 660, 10.1080/00325481.1959.11712484 Selye, 1975, Stress and distress, Comprehensive Therapy, 1, 9 Selye, 1976 Shan, 2016, Aging as a precipitating factor in chronic restraint stress-induced tau aggregation pathology, and the protective effects of rosmarinic acid, Journal of Alzheimer’s Disease: JAD, 49, 829, 10.3233/JAD-150486 Sharma, 2012, Monoclonal antibodies as novel neurotherapeutic agents in CNS injury and repair, International Review of Neurobiology, 102, 23, 10.1016/B978-0-12-386986-9.00002-8 Sharma, 2017, Novel treatment strategies using TiO2-nanowired delivery of histaminergic drugs and antibodies to tau with cerebrolysin for superior neuroprotection in the pathophysiology of Alzheimer’s disease, International Review of Neurobiology, 137, 123, 10.1016/bs.irn.2017.09.002 Sharma, 2020, Mild traumatic brain injury exacerbates Parkinson’s disease induced hemeoxygenase-2 expression and brain pathology: Neuroprotective effects of co-administration of TiO2 nanowired mesenchymal stem cells and cerebrolysin, Progress in Brain Research, 258, 157, 10.1016/bs.pbr.2020.09.010 Sharma, 2021, Histamine H3 and H4 receptors modulate Parkinson’s disease induced brain pathology. Neuroprotective effects of nanowired BF-2649 and clobenpropit with anti-histamine-antibody therapy, Progress in Brain Research, 266, 1, 10.1016/bs.pbr.2021.06.003 Sharma, 2009, Blood–central nervous system barriers: The gateway to neurodegeneration, neuroprotection and neuroregeneration Sharma, 2010, A combination of tumor necrosis factor-alpha and neuronal nitric oxide synthase antibodies applied topically over the traumatized spinal cord enhances neuroprotection and functional recovery in the rat, Annals of the New York Academy of Sciences, 1199, 175, 10.1111/j.1749-6632.2009.05327.x Sharma, 1981, Impairment of blood-brain barrier (BBB) in rat by immobilization stress: Role of serotonin (5-HT), Indian Journal of Physiology and Pharmacology, 25, 111 Sharma, 1984, Role of 5-HT on increased permeability of blood-brain barrier under heat stress, Indian Journal of Physiology and Pharmacology, 28, 259 Sharma, 1986, Probable involvement of 5-hydroxytryptamine in increased permeability of blood-brain barrier under heat stress in young rats, Neuropharmacology, 25, 161, 10.1016/0028-3908(86)90037-7 Sharma, 1986, Influence of long-term immobilization stress on regional blood-brain barrier permeability, cerebral blood flow and 5-HT level in conscious normotensive young rats, Journal of the Neurological Sciences, 72, 61, 10.1016/0022-510X(86)90036-5 Sharma, 1987, Influence of long-term acute heat exposure on regional blood-brain barrier permeability, cerebral blood flow and 5-HT level in conscious normotensive young rats, Brain Research, 424, 153, 10.1016/0006-8993(87)91205-4 Sharma, 1988, EEG changes following increased blood-brain barrier permeability under long-term immobilization stress in young rats, Neuroscience Research, 5, 224, 10.1016/0168-0102(88)90051-X Sharma, 1990, Brain oedema and cellular changes induced by acute heat stress in young rats, Acta Neurochirurgica. Supplementum, 51, 383 Sharma, 2008, Antibodies as promising novel neuroprotective agents in the central nervous system injuries, Central Nervous System Agents in Medicinal Chemistry, 8, 143, 10.2174/187152408785699640 Sharma, 1991, Increased blood-brain barrier permeability following acute short-term swimming exercise in conscious normotensive young rats, Neuroscience Research, 10, 211, 10.1016/0168-0102(91)90058-7 Sharma, 1991, Acute heat exposure causes cellular alteration in cerebral cortex of young rats, Neuroreport, 2, 155, 10.1097/00001756-199103000-00012 Sharma, 1998, Pathophysiology of brain edema and cell changes following hyperthermic brain injury, Progress in Brain Research, 115, 351, 10.1016/S0079-6123(08)62043-9 Sharma H.S., Nyberg F., Gordh T., Alm P.(n.d.). Topical application of dynorphin A (1–17) antibodies attenuates neuronal nitric oxide synthase up-regulation, edema formation, and cell injury following focal trauma to the rat spinal cord. Sharma, 1992, Acute systemic heat stress increases glial fibrillary acidic protein immunoreactivity in brain: Experimental observations in conscious normotensive young rats, Neuroscience, 48, 889, 10.1016/0306-4522(92)90277-9 Sharma, 1996, Involvement of nitric oxide in acute spinal cord injury: An immunocytochemical study using light and electron microscopy in the rat, Neuroscience Research, 24, 373, 10.1016/0168-0102(95)01015-7 Sharma, 2012, The blood-brain barrier in Alzheimer’s disease: Novel therapeutic targets and nanodrug delivery, International Review of Neurobiology, 102, 47, 10.1016/B978-0-12-386986-9.00003-X Sharma, 2013, Exacerbation of brain pathology after partial restraint in hypertensive rats following SiO2 nanoparticles exposure at high ambient temperature, Molecular Neurobiology, 48, 368, 10.1007/s12035-013-8502-y Sharma, 1998, Blood-brain barrier in stress: A gateway to various brain diseases, 259 Sharma, 2010, Influence of nanoparticles on blood-brain barrier permeability and brain edema formation in rats, Acta Neurochirurgica. Supplement, 106, 359, 10.1007/978-3-211-98811-4_65 Sharma, 1992, Age-related pathophysiology of the blood-brain barrier in heat stress, Progress in Brain Research, 91, 189, 10.1016/S0079-6123(08)62334-1 Sharma, 2003, Topical application of TNF-alpha antiserum attenuates spinal cord trauma induced edema formation, microvascular permeability disturbances and cell injury in the rat, Acta Neurochirurgica. Supplement, 86, 407 Sharma, 2019, Nanowired delivery of cerebrolysin with neprilysin and p-Tau antibodies induces superior neuroprotection in Alzheimer’s disease, Progress in Brain Research, 245, 145, 10.1016/bs.pbr.2019.03.009 Sharma, 2021, Alzheimer’s disease neuropathology is exacerbated following traumatic brain injury. Neuroprotection by co-administration of nanowired mesenchymal stem cells and cerebrolysin with monoclonal antibodies to amyloid beta peptide, Progress in Brain Research, 265, 1, 10.1016/bs.pbr.2021.04.008 Singer, 1990, Mechanism of the neurotoxicity of MPTP. An update, FEBS Letters, 274, 1 Skultétyová, 1998, Stress-induced increase in blood-brain barrier permeability in control and monosodium glutamate-treated rats, Brain Research Bulletin, 45, 175, 10.1016/S0361-9230(97)00335-3 Smith, 2008, Stress accelerates neural degeneration and exaggerates motor symptoms in a rat model of Parkinson’s disease, The European Journal of Neuroscience, 27, 2133, 10.1111/j.1460-9568.2008.06177.x Solar, 2022, Blood-brain barrier alterations and edema formation in different brain mass lesions, Frontiers in Cellular Neuroscience, 16, 10.3389/fncel.2022.922181 Son, 2019, A chronic immobilization stress protocol for inducing depression-like behavior in mice, Journal of Visualized Experiments, 147 Stefanova, 2022, Microglia in Parkinson’s disease, Journal of Parkinson’s Disease, 12, S105, 10.3233/JPD-223237 Subramaniam, 2013, Mitochondrial dysfunction and oxidative stress in Parkinson’s disease, Progress in Neurobiology, 106, 17, 10.1016/j.pneurobio.2013.04.004 Sugama, 2016, Loss of dopaminergic neurons occurs in the ventral tegmental area and hypothalamus of rats following chronic stress: Possible pathogenetic loci for depression involved in Parkinson’s disease, Neuroscience Research, 111, 48, 10.1016/j.neures.2016.04.008 Sugama, 2016, Chronic restraint stress triggers dopaminergic and noradrenergic neurodegeneration: Possible role of chronic stress in the onset of Parkinson’s disease, Brain, Behavior, and Immunity, 51, 39, 10.1016/j.bbi.2015.08.015 Sun, 2017, Effects of Hint1 deficiency on emotional-like behaviors in mice under chronic immobilization stress, Brain and Behavior, 7, 10.1002/brb3.831 Szabo, 2017, Stress is 80 years old: From Hans Selye original paper in 1936 to recent advances in GI ulceration, Current Pharmaceutical Design, 23, 4029, 10.2174/1381612823666170622110046 Tipton, 1993, Advances in our understanding of the mechanisms of the neurotoxicity of MPTP and related compounds, Journal of Neurochemistry, 61, 1191, 10.1111/j.1471-4159.1993.tb13610.x Torres, 2002, Effects of chronic restraint stress on feeding behavior and on monoamine levels in different brain structures in rats, Neurochemical Research, 27, 519, 10.1023/A:1019856821430 Trist, 2019, Oxidative stress in the aging substantia nigra and the etiology of Parkinson’s disease, Aging Cell, 18, 10.1111/acel.13031 Urakami, 1988, Increase of striatal dopamine turnover by stress in MPTP-treated mice, Clinical Neuropharmacology, 11, 360, 10.1097/00002826-198808000-00004 Valuskova, 2017, Brain region-specific effects of immobilization stress on cholinesterases in mice, Stress, 20, 36, 10.1080/10253890.2016.1263836 van der Heide, 2021, Stress and mindfulness in Parkinson’s disease: Clinical effects and potential underlying mechanisms, Movement Disorders: Official Journal of the Movement Disorder Society, 36, 64, 10.1002/mds.28345 Wang, 2019, dl-3-n-Butylphthalide inhibits NLRP3 inflammasome and mitigates Alzheimer’s-like pathology via Nrf2-TXNIP-TrX axis, Antioxidants & Redox Signaling, 30, 1411, 10.1089/ars.2017.7440 Wang, 2018, dl-3-n-Butylphthalide (NBP): A promising therapeutic agent for ischemic stroke, CNS & Neurological Disorders Drug Targets, 17, 338, 10.2174/1871527317666180612125843 Wang, 2022, Cerebral blood flow alterations specific to freezing of gait in Parkinson’s disease, Neurological Sciences: Official Journal of the Italian Neurological Society and of the Italian Society of Clinical Neurophysiology, 43, 5323, 10.1007/s10072-022-06205-6 Warren, 2017, Dopamine dysregulation syndrome in Parkinson’s disease: A systematic review of published cases, Journal of Neurology, Neurosurgery, and Psychiatry, 88, 1060, 10.1136/jnnp-2017-315985 Wei, 2021, dl-3-n-butylphthalide increases collateriogenesis and functional recovery after focal ischemic stroke in mice, Aging Dis, 12, 1835, 10.14336/AD.2020.1226 Weiss, 2022, Immune responses in the Parkinson’s disease brain, Neurobiology of Disease, 168, 10.1016/j.nbd.2022.105700 Zhang, 2018, RTP801 is a critical factor in the neurodegeneration process of A53T α-synuclein in a mouse model of Parkinson’s disease under chronic restraint stress, British Journal of Pharmacology, 175, 590, 10.1111/bph.14091 Zhao, 2007, TNF-alpha knockout and minocycline treatment attenuates blood-brain barrier leakage in MPTP-treated mice, Neurobiology of Disease, 26, 36, 10.1016/j.nbd.2006.11.012 Zhou, 2022, The role of tyrosine hydroxylase-dopamine pathway in Parkinson’s disease pathogenesis, Cellular and Molecular Life Sciences: CMLS, 79, 599, 10.1007/s00018-022-04574-x Zlokovic, 2008, The blood-brain barrier in health and chronic neurodegenerative disorders, Neuron, 57, 178, 10.1016/j.neuron.2008.01.003