1
|
Pande S. Comparative potential of fenugreek (Trigonella foenum-graecum L.) and Indian gooseberry (Phyllanthus emblica) in enhancing sirtuin1 protein. Food Chem 2025; 477:143596. [PMID: 40023951 DOI: 10.1016/j.foodchem.2025.143596] [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: 03/06/2024] [Revised: 02/11/2025] [Accepted: 02/23/2025] [Indexed: 03/04/2025]
Abstract
Sirtuin1 (SIRT1) protein is NAD+-dependent with significant health benefits. The effect of fenugreek (F) and Indian gooseberry (IG) on SIRT1 protein remains obscure. The present study explored the effect of F and IG individually and in combination on SIRT1 protein. Results indicated that a combination of F + IG produced a more significant increase in SIRT1 protein in serum (45 %), heart (24 %), liver (38 %), and kidneys (27.8 %) than individually. The individual potential of IG (14.5 %, 17.1 %, 35.5 %, and 23 % in serum, heart, liver, and kidneys respectively) in enhancing SIRT1 was more pronounced than F (19.2 %, 27 %, 15 %, and 18 % in serum, heart, liver, and kidneys respectively). These findings were consistent with obtained NAD+/NADH ratio. F + IG produced synergistic effect (2-fold and 4-fold increase in catalase and superoxide dismutase respectively) and an additive effect in glutathione peroxidase levels in serum. Glutathione S transferase activity was enhanced by 2.6-fold, 2-fold, and 2.3-fold by F, IG, and F + IG respectively in serum. The study concluded that a combination of F and IG together may be advocated for enhancing SIRT1 protein.
Collapse
Affiliation(s)
- Shubhra Pande
- Centre for Rural Development and Technology, Indian Institute of Technology Delhi, New Delhi, India; Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi, India.
| |
Collapse
|
2
|
Pawlik K, Ciapała K, Ciechanowska A, Makuch W, Mika J. Pharmacological modulation of neutrophils, in contrast to that of macrophages/microglia, is sex independent and delays the development of morphine tolerance in a mouse model of neuropathic pain. Biomed Pharmacother 2025; 187:118149. [PMID: 40349556 DOI: 10.1016/j.biopha.2025.118149] [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: 03/10/2025] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025] Open
Abstract
Identifying sex-specific mechanisms underlying neuropathic pain, as well as its therapy, remains a challenge. Recent studies suggest important roles for neutrophils, macrophages and microglia in the development of hypersensitivity and the morphine effectiveness. Therefore, the aim of this study was to test whether substances that inhibit the activation/influx of neutrophils (4-aminobenzoic hydrazide) and microglia/macrophages (minocycline) can help achieve pain relief in male/female mice and improve the analgesic effectiveness of morphine in neuropathy in both sexes. Our behavioral studies performed using chronic constriction injury of the sciatic nerve indicate that repeated twice-daily administrations of 4-aminobenzoic hydrazide (in both sexes) and minocycline (only in males) cause analgesia and delay morphine tolerance development. Observations in female include the absence of alleviation of tactile hypersensitivity (von Frey test) following minocycline, and even an increase in thermal hypersensitivity (cold plate test), which may be of clinical importance. This may be explained by a lack of impact on IBA-1 protein level after repeated administration of minocycline in females, along with increased levels of pronociceptive factors such as CCL2, CXCL2, and TNFα. Notably, the repeated twice-daily administration of 4-aminobenzoic hydrazide has beneficial analgesic effects and delays morphine tolerance development in both sexes. Its influence on male mice is likely caused by its impact on spinal neutrophil activation/influx and on the level of anti-(IL-4)/pro-(CCL2) nociceptive cytokines. Our studies provide the first evidence that the inhibition of neutrophil activation/influx during long-term morphine treatment can improve its efficacy and delay the development of opioid tolerance in neuropathy in both sexes.
Collapse
Affiliation(s)
- Katarzyna Pawlik
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Street, Krakow 31-343, Poland
| | - Katarzyna Ciapała
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Street, Krakow 31-343, Poland
| | - Agata Ciechanowska
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Street, Krakow 31-343, Poland
| | - Wioletta Makuch
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Street, Krakow 31-343, Poland
| | - Joanna Mika
- Maj Institute of Pharmacology Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Street, Krakow 31-343, Poland.
| |
Collapse
|
3
|
Mok VCT, Cai Y, Markus HS. Vascular cognitive impairment and dementia: Mechanisms, treatment, and future directions. Int J Stroke 2024; 19:838-856. [PMID: 39283037 PMCID: PMC11490097 DOI: 10.1177/17474930241279888] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 08/17/2024] [Indexed: 10/21/2024]
Abstract
Worldwide, around 50 million people live with dementia, and this number is projected to triple by 2050. It has been estimated that 20% of all dementia cases have a predominant cerebrovascular pathology, while perhaps another 20% of vascular diseases contribute to a mixed dementia picture. Therefore, the vascular contribution to dementia affects 20 million people currently and will increase markedly in the next few decades, particularly in lower- and middle-income countries.In this review, we discuss the mechanisms of vascular cognitive impairment (VCI) and review management. VCI refers to the spectrum of cerebrovascular pathologies that contribute to any degree of cognitive impairment, ranging from subjective cognitive decline, to mild cognitive impairment, to dementia. While acute cognitive decline occurring soon after a stroke is the most recognized form of VCI, chronic cerebrovascular disease, in particular cerebral small-vessel disease, can cause insidious cognitive decline in the absence of stroke. Moreover, cerebrovascular disease not only commonly co-occurs with Alzheimer's disease (AD) and increases the probability that AD pathology will result in clinical dementia, but may also contribute etiologically to the development of AD pathologies.Despite its enormous health and economic impact, VCI has been a neglected research area, with few adequately powered trials of therapies, resulting in few proven treatments. Current management of VCI emphasizes prevention and treatment of stroke and vascular risk factors, with most evidence for intensive hypertension control. Reperfusion therapies in acute stroke may attenuate the risk of VCI. Associated behavioral symptoms such as apathy and poststroke emotionalism are common. We also highlight novel treatment strategies that will hopefully lead to new disease course-modifying therapies. Finally, we highlight the importance of including cognitive endpoints in large cardiovascular prevention trials and the need for an increased research focus and funding for this important area.
Collapse
Affiliation(s)
- Vincent Chung Tong Mok
- Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Therese Pei Fong Chow Research Centre for Prevention of Dementia, Lui Che Woo Institute of Innovative Medicine, Gerald Choa Neuroscience Institute, Li Ka Shing Institute of Health Science, Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Yuan Cai
- Lau Tat-chuen Research Centre of Brain Degenerative Diseases in Chinese, Therese Pei Fong Chow Research Centre for Prevention of Dementia, Lui Che Woo Institute of Innovative Medicine, Gerald Choa Neuroscience Institute, Li Ka Shing Institute of Health Science, Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Hugh S Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
4
|
Ip BYM, Ko H, Lam BYK, Au LWC, Lau AYL, Huang J, Kwok AJ, Leng X, Cai Y, Leung TWH, Mok VCT. Current and Future Treatments of Vascular Cognitive Impairment. Stroke 2024; 55:822-839. [PMID: 38527144 DOI: 10.1161/strokeaha.123.044174] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Affiliation(s)
- Bonaventure Yiu Ming Ip
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
- Kwok Tak Seng Centre for Stroke Research and Intervention, Hong Kong SAR, China (B.Y.M.I., X.L., T.W.H.L.)
| | - Ho Ko
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Bonnie Yin Ka Lam
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Lisa Wing Chi Au
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Alexander Yuk Lun Lau
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
| | - Junzhe Huang
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Andrew John Kwok
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Xinyi Leng
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Kwok Tak Seng Centre for Stroke Research and Intervention, Hong Kong SAR, China (B.Y.M.I., X.L., T.W.H.L.)
| | - Yuan Cai
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| | - Thomas Wai Hong Leung
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Kwok Tak Seng Centre for Stroke Research and Intervention, Hong Kong SAR, China (B.Y.M.I., X.L., T.W.H.L.)
| | - Vincent Chung Tong Mok
- Division of Neurology, Department of Medicine and Therapeutics (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Li Ka Shing Institute of Health Sciences (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., X.L., C.Y., T.W.H.L., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Margaret K. L. Cheung Research Centre for Management of Parkinsonism (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Lau Tat-Chuen Research Centre of Brain Degenerative Diseases in Chinese (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., A.Y.L.L., J.H., A.J.K., C.Y., V.C.T.M.), Faculty of Medicine, The Chinese University of Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong (B.Y.M.I., H.K., B.Y.K.L., L.W.C.A., J.H., A.J.K., C.Y., V.C.T.M.)
| |
Collapse
|
5
|
Bower WA, Yu Y, Person MK, Parker CM, Kennedy JL, Sue D, Hesse EM, Cook R, Bradley J, Bulitta JB, Karchmer AW, Ward RM, Cato SG, Stephens KC, Hendricks KA. CDC Guidelines for the Prevention and Treatment of Anthrax, 2023. MMWR Recomm Rep 2023; 72:1-47. [PMID: 37963097 PMCID: PMC10651316 DOI: 10.15585/mmwr.rr7206a1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023] Open
Abstract
This report updates previous CDC guidelines and recommendations on preferred prevention and treatment regimens regarding naturally occurring anthrax. Also provided are a wide range of alternative regimens to first-line antimicrobial drugs for use if patients have contraindications or intolerances or after a wide-area aerosol release of Bacillus anthracis spores if resources become limited or a multidrug-resistant B. anthracis strain is used (Hendricks KA, Wright ME, Shadomy SV, et al.; Workgroup on Anthrax Clinical Guidelines. Centers for Disease Control and Prevention expert panel meetings on prevention and treatment of anthrax in adults. Emerg Infect Dis 2014;20:e130687; Meaney-Delman D, Rasmussen SA, Beigi RH, et al. Prophylaxis and treatment of anthrax in pregnant women. Obstet Gynecol 2013;122:885-900; Bradley JS, Peacock G, Krug SE, et al. Pediatric anthrax clinical management. Pediatrics 2014;133:e1411-36). Specifically, this report updates antimicrobial drug and antitoxin use for both postexposure prophylaxis (PEP) and treatment from these previous guidelines best practices and is based on systematic reviews of the literature regarding 1) in vitro antimicrobial drug activity against B. anthracis; 2) in vivo antimicrobial drug efficacy for PEP and treatment; 3) in vivo and human antitoxin efficacy for PEP, treatment, or both; and 4) human survival after antimicrobial drug PEP and treatment of localized anthrax, systemic anthrax, and anthrax meningitis. Changes from previous CDC guidelines and recommendations include an expanded list of alternative antimicrobial drugs to use when first-line antimicrobial drugs are contraindicated or not tolerated or after a bioterrorism event when first-line antimicrobial drugs are depleted or ineffective against a genetically engineered resistant B. anthracis strain. In addition, these updated guidelines include new recommendations regarding special considerations for the diagnosis and treatment of anthrax meningitis, including comorbid, social, and clinical predictors of anthrax meningitis. The previously published CDC guidelines and recommendations described potentially beneficial critical care measures and clinical assessment tools and procedures for persons with anthrax, which have not changed and are not addressed in this update. In addition, no changes were made to the Advisory Committee on Immunization Practices recommendations for use of anthrax vaccine (Bower WA, Schiffer J, Atmar RL, et al. Use of anthrax vaccine in the United States: recommendations of the Advisory Committee on Immunization Practices, 2019. MMWR Recomm Rep 2019;68[No. RR-4]:1-14). The updated guidelines in this report can be used by health care providers to prevent and treat anthrax and guide emergency preparedness officials and planners as they develop and update plans for a wide-area aerosol release of B. anthracis.
Collapse
|
6
|
Narne P, Phanithi PB. Role of NAD + and FAD in Ischemic Stroke Pathophysiology: An Epigenetic Nexus and Expanding Therapeutic Repertoire. Cell Mol Neurobiol 2023; 43:1719-1768. [PMID: 36180651 PMCID: PMC11412205 DOI: 10.1007/s10571-022-01287-4] [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: 04/03/2022] [Accepted: 09/15/2022] [Indexed: 11/03/2022]
Abstract
The redox coenzymes viz., oxidized β-nicotinamide adenine dinucleotide (NAD+) and flavin adenine dinucleotide (FAD) by way of generation of optimal reducing power and cellular energy currency (ATP), control a staggering array of metabolic reactions. The prominent cellular contenders for NAD+ utilization, inter alia, are sirtuins (SIRTs) and poly(ADP-ribose) polymerase (PARP-1), which have been significantly implicated in ischemic stroke (IS) pathogenesis. NAD+ and FAD are also two crucial epigenetic enzyme-required metabolites mediating histone deacetylation and poly(ADP-ribosyl)ation through SIRTs and PARP-1 respectively, and demethylation through FAD-mediated lysine specific demethylase activity. These enzymes and post-translational modifications impinge on the components of neurovascular unit, primarily neurons, and elicit diverse functional upshots in an ischemic brain. These could be circumstantially linked with attendant cognitive deficits and behavioral outcomes in post-stroke epoch. Parsing out the contribution of NAD+/FAD-synthesizing and utilizing enzymes towards epigenetic remodeling in IS setting, together with their cognitive and behavioral associations, combined with possible therapeutic implications will form the crux of this review.
Collapse
Affiliation(s)
- Parimala Narne
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana State, 500046, India.
| | - Prakash Babu Phanithi
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana State, 500046, India.
| |
Collapse
|
7
|
Tang Z, Meng S, Song Z, Yang X, Li X, Guo H, Du M, Chen J, Zhu YZ, Wang X. Neutrophil membrane fusogenic nanoliposomal leonurine for targeted ischemic stroke therapy via remodeling cerebral niche and restoring blood-brain barrier integrity. Mater Today Bio 2023; 20:100674. [PMID: 37273794 PMCID: PMC10238753 DOI: 10.1016/j.mtbio.2023.100674] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/19/2023] [Accepted: 05/18/2023] [Indexed: 06/06/2023] Open
Abstract
Ischemic stroke (IS) constitutes the leading cause of global morbidity and mortality. Neuroprotectants are essential to ameliorate the clinical prognosis, but their therapeutic outcomes are tremendously compromised by insufficient delivery to the ischemic lesion and intricate pathogenesis associated with neuronal damage, oxidative stress, inflammation responses, blood-brain barrier (BBB) dysfunction, etc. Herein, a biomimetic nanosystem (Leo@NM-Lipo) composed of neutrophil membrane-fused nanoliposomal leonurine (Leo) is constructed, which can not only efficiently penetrate and repair the disrupted BBB but also robustly remodel the harsh cerebral microenvironment to reverse ischemia-reperfusion (I/R) injury. More specifically, the neutrophil membrane inherits the BBB penetrating, infarct core targeting, inflammation neutralization, and immune evasion properties of neutrophils, while Leo, a naturally occurring neuroprotectant, exerts pleiotropic effects to attenuate brain damage. Remarkably, comprehensive investigations disclose the critical factors influencing the targetability and therapeutic performances of biomimetic nanosystems. Leo@NM-Lipo with a low membrane protein-to-lipid ratio of 1:10 efficiently targets the ischemic lesion and rescues the injured brain by alleviating neuronal apoptosis, oxidative stress, neuroinflammation, and restoring BBB integrity in transient middle cerebral artery occlusion (tMCAO) rats. Taken together, our study provides a neutrophil-mimetic nanoplatform for targeted IS therapy and sheds light on the rational design of biomimetic nanosystems favoring wide medical applications.
Collapse
Affiliation(s)
- Zhuang Tang
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Shiyu Meng
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Zhiling Song
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Xiaoxue Yang
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Xinzhi Li
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Hui Guo
- School of Chemical Engineering and Technology, Sun Yat-sen University, Zhuhai 519082, China
| | - Meirong Du
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Jun Chen
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yi Zhun Zhu
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| | - Xiaolin Wang
- School of Pharmacy and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao 999078, China
| |
Collapse
|
8
|
Loss of Blood-Brain Barrier Integrity in an In Vitro Model Subjected to Intermittent Hypoxia: Is Reversion Possible with a HIF-1α Pathway Inhibitor? Int J Mol Sci 2023; 24:ijms24055062. [PMID: 36902491 PMCID: PMC10003655 DOI: 10.3390/ijms24055062] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/11/2023] [Accepted: 02/27/2023] [Indexed: 03/09/2023] Open
Abstract
Several sleep-related breathing disorders provoke repeated hypoxia stresses, which potentially lead to neurological diseases, such as cognitive impairment. Nevertheless, consequences of repeated intermittent hypoxia on the blood-brain barrier (BBB) are less recognized. This study compared two methods of intermittent hypoxia induction on the cerebral endothelium of the BBB: one using hydralazine and the other using a hypoxia chamber. These cycles were performed on an endothelial cell and astrocyte coculture model. Na-Fl permeability, tight junction protein, and ABC transporters (P-gp and MRP-1) content were evaluated with or without HIF-1 inhibitors YC-1. Our results demonstrated that hydralazine as well as intermittent physical hypoxia progressively altered BBB integrity, as shown by an increase in Na-Fl permeability. This alteration was accompanied by a decrease in concentration of tight junction proteins ZO-1 and claudin-5. In turn, microvascular endothelial cells up-regulated the expression of P-gp and MRP-1. An alteration was also found under hydralazine after the third cycle. On the other hand, the third intermittent hypoxia exposure showed a preservation of BBB characteristics. Furthermore, inhibition of HIF-1α with YC-1 prevented BBB dysfunction after hydralazine treatment. In the case of physical intermittent hypoxia, we observed an incomplete reversion suggesting that other biological mechanisms may be involved in BBB dysfunction. In conclusion, intermittent hypoxia led to an alteration of the BBB model with an adaptation observed after the third cycle.
Collapse
|
9
|
Vaseghi S, Mostafavijabbari A, Alizadeh MS, Ghaffarzadegan R, Kholghi G, Zarrindast MR. Intricate role of sleep deprivation in modulating depression: focusing on BDNF, VEGF, serotonin, cortisol, and TNF-α. Metab Brain Dis 2023; 38:195-219. [PMID: 36399239 DOI: 10.1007/s11011-022-01124-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 11/06/2022] [Indexed: 11/19/2022]
Abstract
In this review article, we aimed to discuss intricate roles of SD in modulating depression in preclinical and clinical studies. Decades of research have shown the inconsistent effects of SD on depression, focusing on SD duration. However, inconsistent role of SD seems to be more complicated, and SD duration cannot be the only one factor. Regarding this issue, we chose some important factors involved in the effects of SD on cognitive functions and mood including brain-derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF), serotonin, cortisol, and tumor necrosis factor-alpha (TNF-α). It was concluded that SD has a wide-range of inconsistent effects on BDNF, VEGF, serotonin, and cortisol levels. It was noted that BDNF diurnal rhythm is significantly involved in the modulatory role of SD in depression. Furthermore, the important role of VEGF in blood-brain barrier permeability which is involved in modulating depression was discussed. It was also noted that there is a negative correlation between cortisol and BDNF that modulates depression. Eventually, it was concluded that TNF-α regulates sleep/wake cycle and is involved in the vulnerability to cognitive and behavioral impairments following SD. TNF-α also increases the permeability of the blood-brain barrier which is accompanied by depressive behavior. In sum, it was suggested that future studies should focus on these mechanisms/factors to better investigate the reasons behind intricate roles of SD in modulating depression.
Collapse
Affiliation(s)
- Salar Vaseghi
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran.
| | | | - Mohammad-Sadegh Alizadeh
- Department of Cognitive Neuroscience, Institute for Cognitive Science Studies (ICSS), Tehran, Iran
- Department of Cellular and Molecular Sciences, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Reza Ghaffarzadegan
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran
| | - Gita Kholghi
- Department of Psychology, Faculty of Human Sciences, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Mohammad-Reza Zarrindast
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Monsour M, Gordon J, Lockard G, Alayli A, Elsayed B, Connolly J, Borlongan CV. Minor Changes for a Major Impact: A Review of Epigenetic Modifications in Cell-Based Therapies for Stroke. Int J Mol Sci 2022; 23:13106. [PMID: 36361891 PMCID: PMC9656972 DOI: 10.3390/ijms232113106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 08/16/2024] Open
Abstract
Epigenetic changes in stroke may revolutionize cell-based therapies aimed at reducing ischemic stroke risk and damage. Epigenetic changes are a novel therapeutic target due to their specificity and potential for reversal. Possible targets for epigenetic modification include DNA methylation and demethylation, post-translational histone modification, and the actions of non-coding RNAs such as microRNAs. Many of these epigenetic modifications have been reported to modulate atherosclerosis development and progression, ultimately contributing to stroke pathogenesis. Furthermore, epigenetics may play a major role in inflammatory responses following stroke. Stem cells for stroke have demonstrated safety in clinical trials for stroke and show therapeutic benefit in pre-clinical studies. The efficacy of these cell-based interventions may be amplified with adjunctive epigenetic modifications. This review advances the role of epigenetics in atherosclerosis and inflammation in the context of stroke, followed by a discussion on current stem cell studies modulating epigenetics to ameliorate stroke damage.
Collapse
Affiliation(s)
- Molly Monsour
- University of South Florida Morsani College of Medicine, Tampa, FL 33602, USA
| | - Jonah Gordon
- University of South Florida Morsani College of Medicine, Tampa, FL 33602, USA
| | - Gavin Lockard
- University of South Florida Morsani College of Medicine, Tampa, FL 33602, USA
| | - Adam Alayli
- University of South Florida Morsani College of Medicine, Tampa, FL 33602, USA
| | - Bassel Elsayed
- University of South Florida Morsani College of Medicine, Tampa, FL 33602, USA
| | - Jacob Connolly
- University of South Florida Morsani College of Medicine, Tampa, FL 33602, USA
| | - Cesar V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
11
|
Hao K, Wang H, Zhang Y, Xie X, Huang H, Chen C, Xu S, Xu R, Shu C, Liu Z, Zhou Y, Reynolds GP, Wang G. Nicotinamide reverses deficits in puberty-born neurons and cognitive function after maternal separation. J Neuroinflammation 2022; 19:232. [PMID: 36131290 PMCID: PMC9494869 DOI: 10.1186/s12974-022-02591-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 09/04/2022] [Indexed: 01/09/2023] Open
Abstract
Background Early life stress (ELS) is associated with the development of schizophrenia later in life. The hippocampus develops significantly during childhood and is extremely reactive to stress. In rodent models, ELS can induce neuroinflammation, hippocampal neuronal loss, and schizophrenia-like behavior. While nicotinamide (NAM) can inhibit microglial inflammation, it is unknown whether NAM treatment during adolescence reduces hippocampal neuronal loss and abnormal behaviors induced by ELS. Methods Twenty-four hours of maternal separation (MS) of Wistar rat pups on post-natal day (PND)9 was used as an ELS. On PND35, animals received a single intraperitoneal injection of BrdU to label dividing neurons and were given NAM from PND35 to PND65. Behavioral testing was performed. Western blotting and immunofluorescence staining were used to detect nicotinamide adenine dinucleotide (NAD+)/Sirtuin3 (Sirt3)/superoxide dismutase 2 (SOD2) pathway-related proteins. Results Compared with controls, only MS animals in the adult stage (PND56–65) but not the adolescent stage (PND31–40) exhibited pre-pulse inhibition deficits and cognitive impairments mimicking schizophrenia symptoms. MS decreased the survival and activity of puberty-born neurons and hippocampal NAD+ and Sirt3 expression in adulthood. These observations were related to an increase in acetylated SOD2, microglial activation, and significant increases in pro-inflammatory IL-1β, TNF-α, and IL-6 expression. All the effects of MS at PND9 were reversed by administering NAM in adolescence (PND35–65). Conclusions MS may lead to schizophrenia-like phenotypes and persistent hippocampal abnormalities. NAM may be a safe and effective treatment in adolescence to restore normal hippocampal function and prevent or ameliorate schizophrenia-like behavior. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02591-y.
Collapse
Affiliation(s)
- Keke Hao
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Huiling Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China. .,Department of Psychiatry, Zhongxiang Hospital of Renmin Hospital of Wuhan University, Zhongxiang, 431900, China. .,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, 430071, China.
| | - Yuejin Zhang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430060, China
| | - Xinhui Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Huan Huang
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Cheng Chen
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Shilin Xu
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Rui Xu
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Chang Shu
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Yuan Zhou
- Institute of Psychology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Gavin P Reynolds
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China. .,Hubei Institute of Neurology and Psychiatry Research, Wuhan, 430060, China.
| |
Collapse
|
12
|
Qin C, Yang S, Chu YH, Zhang H, Pang XW, Chen L, Zhou LQ, Chen M, Tian DS, Wang W. Signaling pathways involved in ischemic stroke: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2022; 7:215. [PMID: 35794095 PMCID: PMC9259607 DOI: 10.1038/s41392-022-01064-1] [Citation(s) in RCA: 377] [Impact Index Per Article: 125.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/01/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023] Open
Abstract
Ischemic stroke is caused primarily by an interruption in cerebral blood flow, which induces severe neural injuries, and is one of the leading causes of death and disability worldwide. Thus, it is of great necessity to further detailly elucidate the mechanisms of ischemic stroke and find out new therapies against the disease. In recent years, efforts have been made to understand the pathophysiology of ischemic stroke, including cellular excitotoxicity, oxidative stress, cell death processes, and neuroinflammation. In the meantime, a plethora of signaling pathways, either detrimental or neuroprotective, are also highly involved in the forementioned pathophysiology. These pathways are closely intertwined and form a complex signaling network. Also, these signaling pathways reveal therapeutic potential, as targeting these signaling pathways could possibly serve as therapeutic approaches against ischemic stroke. In this review, we describe the signaling pathways involved in ischemic stroke and categorize them based on the pathophysiological processes they participate in. Therapeutic approaches targeting these signaling pathways, which are associated with the pathophysiology mentioned above, are also discussed. Meanwhile, clinical trials regarding ischemic stroke, which potentially target the pathophysiology and the signaling pathways involved, are summarized in details. Conclusively, this review elucidated potential molecular mechanisms and related signaling pathways underlying ischemic stroke, and summarize the therapeutic approaches targeted various pathophysiology, with particular reference to clinical trials and future prospects for treating ischemic stroke.
Collapse
Affiliation(s)
- Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yun-Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Wei Pang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lian Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
13
|
Brown RB, Tozer DJ, Loubière L, Hong YT, Fryer TD, Williams GB, Graves MJ, Aigbirhio FI, O’Brien JT, Markus HS. MINocyclinE to Reduce inflammation and blood brain barrier leakage in small Vessel diseAse (MINERVA) trial study protocol. Eur Stroke J 2022; 7:323-330. [PMID: 36082255 PMCID: PMC9445404 DOI: 10.1177/23969873221100338] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Cerebral small vessel disease (SVD) is a common cause of stroke and cognitive impairment. Recent data has implicated neuroinflammation and increased blood-brain barrier (BBB) permeability in its pathogenesis, but whether such processes are causal and can be therapeutically modified is uncertain. In a rodent model of SVD, minocycline was associated with reduced white matter lesions, inflammation and BBB permeability. Aims: To determine whether blood-brain barrier permeability (measured using dynamic contrast-enhanced MRI) and microglial activation (measured by positron emission tomography using the radioligand 11C-PK11195) can be modified in SVD. Design: Phase II randomised double blind, placebo-controlled trial of minocycline 100 mg twice daily for 3 months in 44 participants with moderate to severe SVD defined as a clinical lacunar stroke and confluent white matter hyperintensities. Outcomes: Primary outcome measures are volume and intensity of focal increases of blood-brain barrier permeability and microglial activation determined using PET-MRI imaging. Secondary outcome measures include inflammatory biomarkers in serum, and change in conventional MRI markers and cognitive performance over 1 year follow up. Discussion: The MINERVA trial aims to test whether minocycline can influence novel pathological processes thought to be involved in SVD progression, and will provide insights into whether central nervous system inflammation in SVD can be therapeutically modulated.
Collapse
Affiliation(s)
- Robin B Brown
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Daniel J Tozer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Laurence Loubière
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Young T Hong
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Tim D Fryer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Guy B Williams
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Martin J Graves
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Franklin I Aigbirhio
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - John T O’Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Hugh S Markus
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
14
|
Abstract
Sirtuins (SIRT) are unique posttranslational modification enzymes that utilize NAD + as co-substrate to remove acyl groups from lysine residues. SIRT act on variety of substrates and impact major metabolic process. All seven members of SIRT family are unique and targets wide range of cellular proteins in nucleus, cytoplasm, and mitochondria for post-translational modification by acetylation (SIRT1, 2, 3, and 5) or ADP-ribosylation (SIRT4 and 6). Each member of SIRT family is distinct. SIRT2 was first to be discovered that incited research on mammalian SIRT. Enzymatic activities of SIRT 4 are yet to be elucidated while only SIRT7 is localized in nucleoli that govern the transcription of RNA polymerase I. SIRT 5 and 6 exhibit weakest deacetylase activity. Out of all SIRT analogs, SIRT1 is identified as nutrient sensor. Increased expression of only SIRT3 is linked with longevity in humans. Since SIRT is regulated by the bioenergetic state of the cell, nutrition impacts it but very few studies about diet-mediated effect on SIRT are reported. The present review elaborates distribution, specific biological role and prominent effect of all SIRT on vital human tissue along with highlighting need to trace molecular mechanisms and identifying foods that may augment it beneficially.
Collapse
Affiliation(s)
- Shubhra Pande
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Sheikh Raisuddin
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| |
Collapse
|
15
|
Shin HS, Zouboulis CC, Kim M, Lee DH, Chung JH. Minocycline Suppresses Lipogenesis via inhibition of p300 Histone Acetyltransferase Activity in Human SZ95 Sebocytes. J Eur Acad Dermatol Venereol 2022; 36:1325-1333. [DOI: 10.1111/jdv.18079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/15/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Hye Sun Shin
- Department of Dermatology Seoul National University College of Medicine Seoul Korea
- Department of Biomedical Sciences Seoul National University Graduate School Seoul Korea
- Institute of Human‐Environment Interface Biology Medical Research Center Seoul National University Seoul Korea
| | - Christos C. Zouboulis
- Departments of Dermatology, Venereology, Allergology and Immunology Dessau Medical Center Brandenburg Medical School Theodor Fontane and Faculty of Health Sciences Brandenburg Dessau Germany
| | - Min‐Kyoung Kim
- Department of Dermatology Seoul National University College of Medicine Seoul Korea
- Institute of Human‐Environment Interface Biology Medical Research Center Seoul National University Seoul Korea
| | - Dong Hun Lee
- Department of Dermatology Seoul National University College of Medicine Seoul Korea
- Institute of Human‐Environment Interface Biology Medical Research Center Seoul National University Seoul Korea
| | - Jin Ho Chung
- Department of Dermatology Seoul National University College of Medicine Seoul Korea
- Department of Biomedical Sciences Seoul National University Graduate School Seoul Korea
- Institute of Human‐Environment Interface Biology Medical Research Center Seoul National University Seoul Korea
- Institute on Aging Seoul National University Seoul Korea
| |
Collapse
|
16
|
Lu Q, Xiong J, Yuan Y, Ruan Z, Zhang Y, Chai B, Li L, Cai S, Xiao J, Wu Y, Huang P, Zhang H. Minocycline improves the functional recovery after traumatic brain injury via inhibition of aquaporin-4. Int J Biol Sci 2022; 18:441-458. [PMID: 34975343 PMCID: PMC8692149 DOI: 10.7150/ijbs.64187] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/04/2021] [Indexed: 12/28/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the main concerns worldwide as there is still no comprehensive therapeutic intervention. Astrocytic water channel aquaporin-4 (AQP-4) system is closely related to the brain edema, water transport at blood-brain barrier (BBB) and astrocyte function in the central nervous system (CNS). Minocycline, a broad-spectrum semisynthetic tetracycline antibiotic, has shown anti-inflammation, anti-apoptotic, vascular protection and neuroprotective effects on TBI models. Here, we tried to further explore the underlying mechanism of minocycline treatment for TBI, especially the relationship of minocycline and AQP4 during TBI treatment. In present study, we observed that minocycline efficaciously reduces the elevation of AQP4 in TBI mice. Furthermore, minocycline significantly reduced neuronal apoptosis, ameliorated brain edema and BBB disruption after TBI. In addition, the expressions of tight junction protein and astrocyte morphology alteration were optimized by minocycline administration. Similar results were found after treating with TGN-020 (an inhibitor of AQP4) in TBI mice. Moreover, these effects were reversed by cyanamide (CYA) treatment, which notably upregulated AQP4 expression level in vivo. In primary cultured astrocytes, small-interfering RNA (siRNA) AQP4 treatment prevented glutamate-induced astrocyte swelling. To sum up, our study suggests that minocycline improves the functional recovery of TBI through reducing AQP4 level to optimize BBB integrity and astrocyte function, and highlights that the AQP4 may be an important therapeutic target during minocycline treating for TBI.
Collapse
Affiliation(s)
- Qi Lu
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Jun Xiong
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Yuan Yuan
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China.,Department of pharmacy, Hangzhou Red Cross Hospital, Zhejiang Province Hospital of Integrated Traditional Chinese and Western Medicine, 310003, Hangzhou, Zhejiang, China
| | - Zhanwei Ruan
- Department of Emergency, Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Yu Zhang
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Bo Chai
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Lei Li
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Shufang Cai
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, 325035, Wenzhou, Zhejiang, China
| | - Jian Xiao
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Yanqing Wu
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, 325035, Wenzhou, Zhejiang, China
| | - Peng Huang
- Department of Pharmacy, Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, 325200, Wenzhou, Zhejiang, China
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China.,Department of Pharmacy, Zhuji People's Hospital, The Affiliated Hospital of Wenzhou Medical University, 311899, Shaoxing, Zhejiang, China
| |
Collapse
|
17
|
He Q, Ma Y, Liu J, Zhang D, Ren J, Zhao R, Chang J, Guo ZN, Yang Y. Biological Functions and Regulatory Mechanisms of Hypoxia-Inducible Factor-1α in Ischemic Stroke. Front Immunol 2021; 12:801985. [PMID: 34966392 PMCID: PMC8710457 DOI: 10.3389/fimmu.2021.801985] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/26/2021] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke is caused by insufficient cerebrovascular blood and oxygen supply. It is a major contributor to death or disability worldwide and has become a heavy societal and clinical burden. To date, effective treatments for ischemic stroke are limited, and innovative therapeutic methods are urgently needed. Hypoxia inducible factor-1α (HIF-1α) is a sensitive regulator of oxygen homeostasis, and its expression is rapidly induced after hypoxia/ischemia. It plays an extensive role in the pathophysiology of stroke, including neuronal survival, neuroinflammation, angiogenesis, glucose metabolism, and blood brain barrier regulation. In addition, the spatiotemporal expression profile of HIF-1α in the brain shifts with the progression of ischemic stroke; this has led to contradictory findings regarding its function in previous studies. Therefore, unveiling the Janus face of HIF-1α and its target genes in different type of cells and exploring the role of HIF-1α in inflammatory responses after ischemia is of great importance for revealing the pathogenesis and identifying new therapeutic targets for ischemic stroke. Herein, we provide a succinct overview of the current approaches targeting HIF-1α and summarize novel findings concerning HIF-1α regulation in different types of cells within neurovascular units, including neurons, endothelial cells, astrocytes, and microglia, during the different stages of ischemic stroke. The current representative translational approaches focused on neuroprotection by targeting HIF-1α are also discussed.
Collapse
Affiliation(s)
- Qianyan He
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yinzhong Ma
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jie Liu
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Dianhui Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Jiaxin Ren
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Ruoyu Zhao
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - JunLei Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhen-Ni Guo
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Yi Yang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
18
|
Lu Q, Xiang H, Zhu H, Chen Y, Lu X, Huang C. Intranasal lipopolysaccharide administration prevents chronic stress-induced depression- and anxiety-like behaviors in mice. Neuropharmacology 2021; 200:108816. [PMID: 34599975 DOI: 10.1016/j.neuropharm.2021.108816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/24/2021] [Accepted: 09/26/2021] [Indexed: 02/06/2023]
Abstract
We recently reported that intraperitoneal injection of a low dose of lipopolysaccharide (LPS) prevents chronic stress-induced depression-like behaviors in mice. In this study, we reported that a single intranasal LPS administration (10 μg/mouse) one day prior to stress exposure produced prophylactic effects on chronic social defeat stress (CSDS)-induced depression-like behaviors, which was indicated by the reduction in social interaction time in the social interaction test and the decrease in immobility time in the tail suspension test and forced swimming test. The single intranasal LPS administration prior to stress exposure was also found to prevent CSDS-induced anxiety-like behaviors, including prevention of CSDS-induced decrease in the time spent in open arms in the elevated plus maze test, decrease in the time spent in lit side in the light-dark test, and decrease in the time spent in central regions in the open field test, along with no changes in locomotor activity. Further analysis showed that the single intranasal LPS administration one day prior to stress exposure prevented CSDS-induced increase in levels of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and IL-1β mRNA in the hippocampus and prefrontal cortex. Inhibition of innate immune stimulation by minocycline pretreatment not only abrogated the preventive effect of intranasal LPS administration on CSDS-induced depression- and anxiety-like behaviors, but also abrogated the preventive effect of intranasal LPS administration on CSDS-induced neuroinflammatory responses in the hippocampus and prefrontal cortex. These results demonstrate that intranasal administration of innate immune stimulants could be a potential approach for the prevention of depression and anxiety.
Collapse
Affiliation(s)
- Qun Lu
- Department of Pharmacy, Nantong Third Hospital Affiliated to Nantong University, #60 Middle Qingnian Road, Nantong, 226006, Jiangsu, China
| | - Haitao Xiang
- Department of Neurosurgery, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, #118 Wansheng Street, Suzhou, 215028, Jiangsu, China
| | - Haojie Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Yifan Chen
- Department of Medical Imaging, School of Medicine, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
19
|
Takata F, Nakagawa S, Matsumoto J, Dohgu S. Blood-Brain Barrier Dysfunction Amplifies the Development of Neuroinflammation: Understanding of Cellular Events in Brain Microvascular Endothelial Cells for Prevention and Treatment of BBB Dysfunction. Front Cell Neurosci 2021; 15:661838. [PMID: 34588955 PMCID: PMC8475767 DOI: 10.3389/fncel.2021.661838] [Citation(s) in RCA: 273] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 08/09/2021] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is involved in the onset or progression of various neurodegenerative diseases. Initiation of neuroinflammation is triggered by endogenous substances (damage-associated molecular patterns) and/or exogenous pathogens. Activation of glial cells (microglia and astrocytes) is widely recognized as a hallmark of neuroinflammation and triggers the release of proinflammatory cytokines, leading to neurotoxicity and neuronal dysfunction. Another feature associated with neuroinflammatory diseases is impairment of the blood-brain barrier (BBB). The BBB, which is composed of brain endothelial cells connected by tight junctions, maintains brain homeostasis and protects neurons. Impairment of this barrier allows trafficking of immune cells or plasma proteins into the brain parenchyma and subsequent inflammatory processes in the brain. Besides neurons, activated glial cells also affect BBB integrity. Therefore, BBB dysfunction can amplify neuroinflammation and act as a key process in the development of neuroinflammation. BBB integrity is determined by the integration of multiple signaling pathways within brain endothelial cells through intercellular communication between brain endothelial cells and brain perivascular cells (pericytes, astrocytes, microglia, and oligodendrocytes). For prevention of BBB disruption, both cellular components, such as signaling molecules in brain endothelial cells, and non-cellular components, such as inflammatory mediators released by perivascular cells, should be considered. Thus, understanding of intracellular signaling pathways that disrupt the BBB can provide novel treatments for neurological diseases associated with neuroinflammation. In this review, we discuss current knowledge regarding the underlying mechanisms involved in BBB impairment by inflammatory mediators released by perivascular cells.
Collapse
Affiliation(s)
- Fuyuko Takata
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Shinsuke Nakagawa
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Junichi Matsumoto
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Shinya Dohgu
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
20
|
Yang X, Zhang Y, Geng K, Yang K, Shao J, Xia W. Sirt3 Protects Against Ischemic Stroke Injury by Regulating HIF-1α/VEGF Signaling and Blood-Brain Barrier Integrity. Cell Mol Neurobiol 2021; 41:1203-1215. [PMID: 32500353 PMCID: PMC11448646 DOI: 10.1007/s10571-020-00889-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022]
Abstract
Sirtuin 3 (Sirt3) is a member of the Sirtuin family proteins and known to regulate multiple physiological processes such as metabolism and aging. As stroke is an aging-related disease, in this work, we attempt to examine the role and potential mechanism of Sirt3 in regulating ischemic stroke by using a permanent middle cerebral artery occlusion (pMCAO) model in wild type (WT) and Sirt3 knockout (KO) mice, coupled with oxygen glucose deprivation (OGD) experiments in cultured primary astrocytes. Sirt3 deficiency aggravated neuronal cell apoptosis and neurological deficits after brain ischemia. In addition, Sirt3 KO mice showed more severe blood-brain barrier (BBB) disruption and inflammatory responses compared with WT group in the acute phase. Furthermore, specific overexpression of Sirt3 in astrocytes by injecting glial fibrillary acidic protein (GFAP)::Sirt3 virus in ischemic region showed protective effect against stroke-induced damage. Mechanistically, Sirt3 could regulate vascular endothelial growth factor (VEGF) expression by inhibiting hypoxia inducible factor-1α (HIF-1α) signaling after ischemia (OGD). Our results have shown that Sirt3 plays a protective role in ischemic stroke via regulating HIF-1α/VEGF signaling in astrocytes, and reversal of the Sirt3 expression at the acute phase could be a worthy direction for stroke therapy.
Collapse
Affiliation(s)
- Xiao Yang
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yanshuang Zhang
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Keyi Geng
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Ke Yang
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Jiaxiang Shao
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Weiliang Xia
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.
- Med-X Research Institute, Shanghai Jiao Tong University, Room 211, 1954 Huashan Road, Shanghai, 200030, China.
| |
Collapse
|
21
|
Tyagi A, Mirita C, Shah I, Reddy PH, Pugazhenthi S. Effects of Lipotoxicity in Brain Microvascular Endothelial Cells During Sirt3 Deficiency-Potential Role in Comorbid Alzheimer's Disease. Front Aging Neurosci 2021; 13:716616. [PMID: 34393764 PMCID: PMC8355826 DOI: 10.3389/fnagi.2021.716616] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/08/2021] [Indexed: 11/17/2022] Open
Abstract
Silence information regulator 3 (SIRT3) is an NAD+ dependent deacetylase enzyme that enhances the function of key mitochondrial proteins. We have earlier demonstrated that deletion of Sirt3 gene leads to downregulation of metabolic enzymes, mitochondrial dysfunction and neuroinflammation in the brain, the major causes of Alzheimer’s disease (AD). We also reported recently that Sirt3 gene deletion in Alzheimer’s transgenic mice leads to exacerbation of neuroinflammation, amyloid plaque deposition and microglial activation. AD often coexists with other brain lesions caused by comorbidities which can exert their deleterious effects through the neurovascular unit. This unit consists of brain microvascular endothelial cells (BMECs), end feet of astrocytes, and pericytes. BMECs are uniquely different from other vascular endothelial cells because they are glued together by tight-junction proteins. BMECs are in constant contact with circulating factors as they line the luminal side. Therefore, we hypothesized that vascular endothelial injury caused by comorbidities plays a significant role in neuroinflammation. Herein, we investigated the effects of lipotoxicity in BMECs and how Sirt3 deficiency facilitate the deleterious effects of lipotoxicity on them using in vivo and in vitro models. We observed decreases in the levels of SIRT3 and tight junction proteins in the brain samples of western diet-fed APP/PS1 mice. Similar observations were obtained with Alzheimer’s post-mortem samples. Exposure of BEND3 cells, mouse brain-derived Endothelial cells3, to a combination of high glucose and palmitic acid resulted in significant (P < 0.01-P < 0.001) decreases in the levels of SIRT3, claudin-5 and ZO-1. Induction of inflammatory mediators, including Cox-2, CXCL1, RANTES, and GADD45β was also observed in these treated cells. Interestingly, the induction was more with Sirt3-silenced BEND3 cells, suggesting that Sirt3 deficiency exacerbates inflammatory response. Palmitic acid was more potent in inducing the inflammatory mediators. Significant cytotoxicity and changes in microglial morphology were observed when cocultures of Sirt3-silenced BEND3 and Sirt3-silenced BV2 cells were exposed to palmitic acid. Transendothelial electrical resistance measurement with these cocultures suggested decreased barrier integrity. The findings of this study suggest that hyperlipidemia in comorbidities can compromise blood brain barrier integrity by inducing inflammatory mediators and decreasing tight junction proteins in the vascular endothelial cells of the AD brain, leading to activation of microglia.
Collapse
Affiliation(s)
- Alpna Tyagi
- Rocky Mountain Regional VA Medical Center, Aurora, CO, United States.,Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Carol Mirita
- Rocky Mountain Regional VA Medical Center, Aurora, CO, United States
| | - Iman Shah
- Rocky Mountain Regional VA Medical Center, Aurora, CO, United States
| | - P Hemachandra Reddy
- Internal Medicine Department and Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Subbiah Pugazhenthi
- Rocky Mountain Regional VA Medical Center, Aurora, CO, United States.,Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
22
|
Hamblin MH, Lee JP. Neural Stem Cells for Early Ischemic Stroke. Int J Mol Sci 2021; 22:ijms22147703. [PMID: 34299322 PMCID: PMC8306669 DOI: 10.3390/ijms22147703] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/11/2022] Open
Abstract
Clinical treatments for ischemic stroke are limited. Neural stem cell (NSC) transplantation can be a promising therapy. Clinically, ischemia and subsequent reperfusion lead to extensive neurovascular injury that involves inflammation, disruption of the blood-brain barrier, and brain cell death. NSCs exhibit multiple potentially therapeutic actions against neurovascular injury. Currently, tissue plasminogen activator (tPA) is the only FDA-approved clot-dissolving agent. While tPA’s thrombolytic role within the vasculature is beneficial, tPA’s non-thrombolytic deleterious effects aggravates neurovascular injury, restricting the treatment time window (time-sensitive) and tPA eligibility. Thus, new strategies are needed to mitigate tPA’s detrimental effects and quickly mediate vascular repair after stroke. Up to date, clinical trials focus on the impact of stem cell therapy on neuro-restoration by delivering cells during the chronic stroke stage. Also, NSCs secrete factors that stimulate endogenous repair mechanisms for early-stage ischemic stroke. This review will present an integrated view of the preclinical perspectives of NSC transplantation as a promising treatment for neurovascular injury, with an emphasis on early-stage ischemic stroke. Further, this will highlight the impact of early sub-acute NSC delivery on improving short-term and long-term stroke outcomes.
Collapse
Affiliation(s)
- Milton H. Hamblin
- Department of Pharmacology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA 70112, USA
- Correspondence: (M.H.H.); (J.-P.L.)
| | - Jean-Pyo Lee
- Department of Physiology, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA 70112, USA
- Tulane Brain Institute, Tulane University, 1430 Tulane Ave, New Orleans, LA 70112, USA
- Correspondence: (M.H.H.); (J.-P.L.)
| |
Collapse
|
23
|
He T, Shang J, Gao C, Guan X, Chen Y, Zhu L, Zhang L, Zhang C, Zhang J, Pang T. A novel SIRT6 activator ameliorates neuroinflammation and ischemic brain injury via EZH2/FOXC1 axis. Acta Pharm Sin B 2021; 11:708-726. [PMID: 33777677 PMCID: PMC7982432 DOI: 10.1016/j.apsb.2020.11.002] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/26/2020] [Accepted: 09/07/2020] [Indexed: 02/08/2023] Open
Abstract
Ischemic stroke is the second leading cause of death worldwide with limited medications and neuroinflammation was recognized as a critical player in the progression of stroke, but how to control the overactive neuroinflammation is still a long-standing challenge. Here, we designed a novel SIRT6 activator MDL-811 which remarkably inhibited inflammatory response in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages and primary mouse microglia, which were abolished by silencing SIRT6. RNA-seq screening identified the forkhead box C1 (Foxc1) is a key gene evoked by MDL-811 stimulation and is required for the anti-inflammatory effects of MDL-811. We found MDL-811-activated SIRT6 directly interacted with enhancer of zeste homolog 2 (EZH2) and promoted deacetylation of EZH2 which could bind to the promoter of Foxc1 and upregulate its expression to modulate inflammation. Moreover, our data demonstrated that MDL-811 not only ameliorated sickness behaviors in neuroinflammatory mice induced by LPS, but also markedly reduced the brain injury in ischemic stroke mice in addition to promoting long-term functional recovery. Importantly, MDL-811 also exhibited strong anti-inflammatory effects in human monocytes isolated from ischemic stroke patients, underlying an interesting translational perspective. Taken together, MDL-811 could be an alternative therapeutic candidate for ischemic stroke and other brain disorders associated with neuroinflammation.
Collapse
Affiliation(s)
- Tailin He
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Jialin Shang
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chenglong Gao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Xin Guan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Yingyi Chen
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Liwen Zhu
- Department of Neurology of Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China
| | - Luyong Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| | - Cunjin Zhang
- Department of Neurology of Drum Tower Hospital, Medical School and the State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China
| | - Jian Zhang
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Tao Pang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
24
|
Yang Q, Zhou Y, Sun Y, Luo Y, Shen Y, Shao A. Will Sirtuins Be Promising Therapeutic Targets for TBI and Associated Neurodegenerative Diseases? Front Neurosci 2020; 14:791. [PMID: 32848564 PMCID: PMC7411228 DOI: 10.3389/fnins.2020.00791] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/06/2020] [Indexed: 12/17/2022] Open
Abstract
Traumatic brain injury (TBI), a leading cause of morbidity worldwide, induces mechanical, persistent structural, and metabolic abnormalities in neurons and other brain-resident cells. The key pathological features of TBI include neuroinflammation, oxidative stress, excitotoxicity, and mitochondrial dysfunction. These pathological processes persist for a period of time after TBIs. Sirtuins are evolutionarily conserved nicotinamide-adenine dinucleotide (NAD+)-dependent deacetylases and mono-ADP-ribosyl transferases. The mammalian sirtuin family has seven members, referred to as Sirtuin (SIRT) 1-7. Accumulating evidence suggests that SIRT1 and SIRT3 play a neuroprotective role in TBI. Although the evidence is scant, considering the involvement of SIRT2, 4-7 in other brain injury models, they may also intervene in similar pathophysiology in TBI. Neurodegenerative diseases are generally accepted sequelae of TBI. It was found that TBI and neurodegenerative diseases have many similarities and overlaps in pathological features. Besides, sirtuins play some unique roles in some neurodegenerative diseases. Therefore, we propose that sirtuins might be a promising therapeutic target for both TBI and associated neurodegenerative diseases. In this paper, we review the neuroprotective effects of sirtuins on TBI as well as related neurodegeneration and discuss the therapeutic potential of sirtuin modulators.
Collapse
Affiliation(s)
- Qianjie Yang
- Department of Ophthalmology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuting Sun
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Luo
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ye Shen
- Department of Ophthalmology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
25
|
Boese AC, Eckert A, Hamblin MH, Lee JP. Human neural stem cells improve early stage stroke outcome in delayed tissue plasminogen activator-treated aged stroke brains. Exp Neurol 2020; 329:113275. [PMID: 32147438 PMCID: PMC7609039 DOI: 10.1016/j.expneurol.2020.113275] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/26/2020] [Accepted: 03/03/2020] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Clinically, significant stroke injury results from ischemia-reperfusion (IR), which induces a deleterious biphasic opening of the blood-brain barrier (BBB). Tissue plasminogen activator (tPA) remains the sole pharmacological agent to treat ischemic stroke. However, major limitations of tPA treatment include a narrow effective therapeutic window of 4.5 h in most patients after initial stroke onset and off-target non-thrombolytic effects (e.g., the risk of increased IR injury). We hypothesized that ameliorating BBB damage with exogenous human neural stem cells (hNSCs) would improve stroke outcome to a greater extent than treatment with delayed tPA alone in aged stroke mice. METHODS We employed middle cerebral artery occlusion to produce focal ischemia with subsequent reperfusion (MCAO/R) in aged mice and administered tPA at a delayed time point (6 h post-stroke) via tail vein. We transplanted hNSCs intracranially in the subacute phase of stroke (24 h post-stroke). We assessed the outcomes of hNSC transplantation on pathophysiological markers of stroke 48 h post-stroke (24 h post-transplant). RESULTS Delayed tPA treatment resulted in more extensive BBB damage and inflammation relative to MCAO controls. Notably, transplantation of hNSCs ameliorated delayed tPA-induced escalated stroke damage; decreased expression of proinflammatory factors (tumor necrosis factor-alpha (TNF-α) and interleukin (IL)-6), decreased the level of matrix metalloprotease-9 (MMP-9), increased the level of brain-derived neurotrophic factor (BDNF), and reduced BBB damage. CONCLUSIONS Aged stroke mice that received delayed tPA treatment in combination with hNSC transplantation exhibited reduced stroke pathophysiology in comparison to non-transplanted stroke mice with delayed tPA. This suggests that hNSC transplantation may synergize with already existing stroke therapies to benefit a larger stroke patient population.
Collapse
Affiliation(s)
- Austin C Boese
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Auston Eckert
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jean-Pyo Lee
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA.
| |
Collapse
|
26
|
Wu X, Luo J, Liu H, Cui W, Feng D, Qu Y. SIRT3 protects against early brain injury following subarachnoid hemorrhage via promoting mitochondrial fusion in an AMPK dependent manner. Chin Neurosurg J 2020; 6:1. [PMID: 32922930 PMCID: PMC7398350 DOI: 10.1186/s41016-019-0182-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/21/2019] [Indexed: 12/17/2022] Open
Abstract
Background Subarachnoid hemorrhage (SAH), an acute cerebrovascular accident, features with its high death and disability rate. Sirtuin3 (SIRT3) is a NAD+ dependent deacetylase which mainly located in mitochondria. Reduced SIRT3 function was indicated to involve in many disorders of central nervous system. Herein, we aimed to explore the neuroprotective effects of SIRT3 on SAH and to furtherly explore the underlying mechanisms. Methods Adult C57BL/6 J male mice (8–10 weeks) were used to establish SAH models. The pharmacological agonist of SIRT3, Honokiol (HKL), was injected in an intraperitoneal manner (10 mg/kg) immediately after the operation. Brain edema and neurobehavioral score were assessed. Nissl staining and FJC staining were used to evaluate the extent of neuronal damage. The changes of mitochondria morphology were observed with transmission electron microscopy. Western blot was used for analyzing the protein level of SIRT3 and the downstream signaling molecules. Result SIRT3 was downregulated after SAH, and additional treatment of SIRT3 agonist HKL alleviated brain edema and neurobehavioral deficits after SAH. Additionally, electron microscopy showed that HKL significantly alleviated the morphological damage of mitochondria induced by SAH. Further studies showed that HKL could increase the level of mitochondrial fusion protein Mfn1 and Mfn2, thus maintaining (mitochondrial morphology), protecting mitochondrial function and promoting neural survival. While, additional Compound C (CC) treatment, a selective AMPK inhibitor, abolished these protective effects. Conclusions Activation of SIRT3 protects against SAH injury through improving mitochondrial fusion in an AMPK dependent manner.
Collapse
Affiliation(s)
- Xun Wu
- Department of Neurosurgery, Tangdu Hospital, the Fourth Military Medical University, Xi'an, 710038 Shaanxi China
| | - Jianing Luo
- Department of Neurosurgery, Tangdu Hospital, the Fourth Military Medical University, Xi'an, 710038 Shaanxi China
| | - Haixiao Liu
- Department of Neurosurgery, Tangdu Hospital, the Fourth Military Medical University, Xi'an, 710038 Shaanxi China
| | - Wenxing Cui
- Department of Neurosurgery, Tangdu Hospital, the Fourth Military Medical University, Xi'an, 710038 Shaanxi China
| | - Dayun Feng
- Department of Neurosurgery, Tangdu Hospital, the Fourth Military Medical University, Xi'an, 710038 Shaanxi China
| | - Yan Qu
- Department of Neurosurgery, Tangdu Hospital, the Fourth Military Medical University, Xi'an, 710038 Shaanxi China
| |
Collapse
|
27
|
Sapozhnikova T, Borisevich S, Kireeva D, Gabdrakhmanova S, Khisamutdinova R, Makara N, Gibadullina N, Khursan S, Zarudii F. Effects of novel hexahydropyrimidine derivatives as potential ligands of M1 muscarinic acetylcholine receptor on cognitive function, hypoxia-induced lethality, and oxidative stress in rodents. Behav Brain Res 2019; 373:112109. [DOI: 10.1016/j.bbr.2019.112109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 07/16/2019] [Accepted: 07/20/2019] [Indexed: 12/20/2022]
|
28
|
Stamatovic SM, Phillips CM, Martinez-Revollar G, Keep RF, Andjelkovic AV. Involvement of Epigenetic Mechanisms and Non-coding RNAs in Blood-Brain Barrier and Neurovascular Unit Injury and Recovery After Stroke. Front Neurosci 2019; 13:864. [PMID: 31543756 PMCID: PMC6732937 DOI: 10.3389/fnins.2019.00864] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/31/2019] [Indexed: 12/13/2022] Open
Abstract
Cessation of blood flow leads to a complex cascade of pathophysiological events at the blood-vascular-parenchymal interface which evolves over time and space, and results in damage to neural cells and edema formation. Cerebral ischemic injury evokes a profound and deleterious upregulation in inflammation and triggers multiple cell death pathways, but it also induces a series of the events associated with regenerative responses, including vascular remodeling, angiogenesis, and neurogenesis. Emerging evidence suggests that epigenetic reprograming could play a pivotal role in ongoing post-stroke neurovascular unit (NVU) changes and recovery. This review summarizes current knowledge about post-stroke recovery processes at the NVU, as well as epigenetic mechanisms and modifiers (e.g., DNA methylation, histone modifying enzymes and microRNAs) associated with stroke injury, and NVU repair. It also discusses novel drug targets and therapeutic strategies for enhancing post-stroke recovery.
Collapse
Affiliation(s)
- Svetlana M. Stamatovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Chelsea M. Phillips
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | | | - Richard F. Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Molecular Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Anuska V. Andjelkovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
29
|
Hou Y, Wang J, Feng J. The neuroprotective effects of curcumin are associated with the regulation of the reciprocal function between autophagy and HIF-1α in cerebral ischemia-reperfusion injury. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1135-1144. [PMID: 31040648 PMCID: PMC6461000 DOI: 10.2147/dddt.s194182] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Purpose The beneficial, neuroprotective effects of curcumin against ischemia-reperfusion injury have been demonstrated. In the present study, whether curcumin exerts neuroprotective effects associated with the inhibition of autophagy and hypoxia inducible factor-1α (HIF-1α) was investigated. Materials and methods PC12 cellular model of oxygen glucose deprivation/reperfusion (OGD/R) has been developed to mimic cerebral ischemia-reperfusion injury. Cell viability was evaluated using the CellTiter 96® AQueous One Solution Cell Proliferation Assay. Apoptosis was assessed using flow cytometry. The expression levels of HIF-1α and autophagy-associated proteins, LC3 and P62, were examined using Western blot. The autophagy flux was quantitatively estimated based on the number of autophagic compartments using fluorescence microscopy. In addition, 3-methyladenine (3-MA) was administered to PC12 cells to investigate how autophagy affects HIF-1α. Moreover, the inhibitory effects of HIF-1α on autophagy activation level were examined. Results In this study, curcumin decreased the death and apoptosis of cells, and inhibited autophagy and HIF-1α under OGD/R conditions, consistent with 3-MA treatment or HIF-1α downregulation. Moreover, inhibition of autophagy caused a decrease in HIF-1α, and the attenuation of HIF-1α induced autophagy suppression under OGD/R conditions. Conclusion The results of this study showed that curcumin exerts neuroprotective effects against ischemia-reperfusion, which is associated with the regulation of the reciprocal function between autophagy and HIF-1α.
Collapse
Affiliation(s)
- Yang Hou
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, People's Republic of China,
| | - Jue Wang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, People's Republic of China,
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, People's Republic of China,
| |
Collapse
|
30
|
Ross AM, Mc Nulty D, O'Dwyer C, Grabrucker AM, Cronin P, Mulvihill JJ. Standardization of research methods employed in assessing the interaction between metallic-based nanoparticles and the blood-brain barrier: Present and future perspectives. J Control Release 2019; 296:202-224. [DOI: 10.1016/j.jconrel.2019.01.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 01/31/2023]
|
31
|
Sun Y, Zhu Y, Zhong X, Chen X, Wang J, Ying G. Crosstalk Between Autophagy and Cerebral Ischemia. Front Neurosci 2019; 12:1022. [PMID: 30692904 PMCID: PMC6339887 DOI: 10.3389/fnins.2018.01022] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 12/18/2018] [Indexed: 12/18/2022] Open
Abstract
With the use of advanced electron microscopy and molecular biology tools, several studies have shown that autophagy is involved in the development of ischemic stroke. A series of molecular mechanisms are involved in the regulation of autophagy. In this work, the possible molecular mechanisms involved in autophagy during ischemic stroke were reviewed and new potential targets for the study and treatment of ischemic stroke were provided.
Collapse
Affiliation(s)
- Yulin Sun
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Yuanhan Zhu
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Xiaojun Zhong
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Xinle Chen
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Jun Wang
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Guozheng Ying
- Department of Neurosurgery, Zhejiang Rongjun Hospital, Jiaxing, China
| |
Collapse
|
32
|
New progress in the approaches for blood–brain barrier protection in acute ischemic stroke. Brain Res Bull 2019; 144:46-57. [DOI: 10.1016/j.brainresbull.2018.11.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/10/2018] [Accepted: 11/13/2018] [Indexed: 02/06/2023]
|
33
|
Pu J, Zhu S, Zhou D, Zhao L, Yin M, Wang Z, Hong J. Propofol Alleviates Apoptosis Induced by Chronic High Glucose Exposure via Regulation of HIF-1 α in H9c2 Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4824035. [PMID: 31093315 PMCID: PMC6481038 DOI: 10.1155/2019/4824035] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 01/26/2019] [Accepted: 02/04/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND The sedative anesthetic, propofol, is a cardioprotective agent for hyperglycemia-induced myocardial hypertrophy and dysfunction in rats. However, the specific protective mechanism has not been clarified. METHODS AND RESULTS In this experiment, we used H9c2 cells subjected to 22 mM glucose lasting for 72 hours as an in vitro model of cardiomyocyte injury by hyperglycemia and investigated the potential mechanism of propofol against hyperglycemic stress in cells. Propofol (5, 10, or 20 μM) was added to the cell cultures before and during the high glucose culture phases. Cell viability and levels of ROS were measured. The levels of proinflammatory cytokines were tested by ELISA. The levels of SIRT3, SOD2, PHD2, HIF-1α, Bcl-2, P53, and cleaved caspase-3 proteins were detected by western blotting. Our data showed that propofol attenuated high glucose-induced cell apoptosis accompanied by a decrease in the level of reactive oxygen species (ROS) and proinflammatory cytokines. Meanwhile, propofol decreased the apoptosis of H9c2 cells via increasing the expression of Bcl-2, SIRT3, SOD2, and PHD2 proteins and decreasing the expression of cleaved caspase-3, P53, and HIF-1α. Real-time PCR analysis showed that propofol did not significantly change the HIF-1α but increase PHD2 at mRNA level. HIF-1α silence significantly decreased apoptosis and inflammation in H9c2 cell during high glucose stress. Pretreatment of IOX2 (the inhibitor of PHD2) inhibited cell viability until the concentration reached 200 μM during high glucose stress. However, 50 μM TYP (the inhibitor of SIRT3) significantly inhibited cell viability during high glucose stress. Delayed IOX2 treatment for 6 hours significantly inhibited cell viability during high glucose stress. CONCLUSIONS Propofol might alleviate cell apoptosis via SIRT3-HIF-1α axis during high glucose stress.
Collapse
Affiliation(s)
- Jinjun Pu
- 1Department of Internal and Emergency Medicine, Shanghai General Hospital (Originally Named Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
- 2Department of Emergency Medicine, Putuo Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shun Zhu
- 1Department of Internal and Emergency Medicine, Shanghai General Hospital (Originally Named Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dandan Zhou
- 1Department of Internal and Emergency Medicine, Shanghai General Hospital (Originally Named Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lidong Zhao
- 1Department of Internal and Emergency Medicine, Shanghai General Hospital (Originally Named Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming Yin
- 3School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Zejian Wang
- 3School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Jiang Hong
- 1Department of Internal and Emergency Medicine, Shanghai General Hospital (Originally Named Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Marcus JM, Andrabi SA. SIRT3 Regulation Under Cellular Stress: Making Sense of the Ups and Downs. Front Neurosci 2018; 12:799. [PMID: 30450031 PMCID: PMC6224517 DOI: 10.3389/fnins.2018.00799] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 10/15/2018] [Indexed: 12/17/2022] Open
Abstract
Sirtuin 3 (SIRT3) is an NAD+ dependent deacetylase that resides primarily in mitochondria and functions to maintain mitochondrial homeostasis under stress. SIRT3 expression has been observed to change under a number of different stresses in multiple tissues and model systems. Inconsistencies in the literature with regards to how and when SIRT3 protein levels change indicates that the mechanism of SIRT3 regulation is multi-faceted. Alterations in SIRT3 have been observed in experimental models of cellular stress, however, the effect these changes have on mitochondrial health remain unknown. Neurons are highly dependent on proper mitochondrial function for their survival. SIRT3 dynamics and function have been studied using models of genotoxic, metabolic, and oxidative stresses, although it remains unclear how SIRT3 is being regulated under these conditions. A closer look into SIRT3 regulation under stress conditions in various model systems will help incorporate the many SIRT3 regulatory mechanisms at play in disease states. In this review, we describe the observations that have been made about SIRT3 protein modulation under basic stress conditions. We then point out consistencies and contradictions in these observations and what they mean. Lastly, we present the observations made in the complicated neuronal stress of stroke. We hope that this review will help consolidate the ambiguous SIRT3 literature and provide a framework for investigation of SIRT3 regulation during stress response.
Collapse
Affiliation(s)
- Joshua M Marcus
- Departments of Pharmacology and Toxicology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Shaida A Andrabi
- Departments of Pharmacology and Toxicology, The University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
35
|
Mi Y, Wu Q, Yuan W, Chen F, Du D. Role of microglia M1/M2 polarisation in the paraventricular nucleus: New insight into the development of stress-induced hypertension in rats. Auton Neurosci 2018; 213:71-80. [DOI: 10.1016/j.autneu.2018.06.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/08/2018] [Accepted: 06/11/2018] [Indexed: 12/20/2022]
|
36
|
Boese AC, Le QSE, Pham D, Hamblin MH, Lee JP. Neural stem cell therapy for subacute and chronic ischemic stroke. Stem Cell Res Ther 2018; 9:154. [PMID: 29895321 PMCID: PMC5998588 DOI: 10.1186/s13287-018-0913-2] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Neural stem cells (NSCs) play vital roles in brain homeostasis and exhibit a broad repertoire of potentially therapeutic actions following neurovascular injury. One such injury is stroke, a worldwide leading cause of death and disability. Clinically, extensive injury from ischemic stroke results from ischemia-reperfusion (IR), which is accompanied by inflammation, blood-brain barrier (BBB) damage, neural cell death, and extensive tissue loss. Tissue plasminogen activator (tPA) is still the only US Food and Drug Administration-approved clot-lysing agent. Whereas the thrombolytic role of tPA within the vasculature is beneficial, the effects of tPA (in a non-thrombolytic role) within the brain parenchyma have been reported as harmful. Thus, new therapies are needed to reduce the deleterious side effects of tPA and quickly facilitate vascular repair following stroke. The Stroke Treatment Academic Industry Roundtable (STAIR) recommends that stroke therapies "focus on drugs/devices/treatments with multiple mechanisms of action and that target multiple pathways". Thus, based on multifactorial ischemic cascades in various stroke stages, effective stroke therapies need to focus on targeting and ameliorating early IR injury as well as facilitating angiogenesis, neurogenesis, and neurorestorative mechanisms following stroke. This review will discuss the preclinical perspectives of NSC transplantation as a promising treatment for neurovascular injury and will emphasize both the subacute and chronic phase of ischemic stroke.
Collapse
Affiliation(s)
- Austin C Boese
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Quan-Son Eric Le
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Dylan Pham
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Jean-Pyo Lee
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, 70112, USA. .,Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA.
| |
Collapse
|
37
|
Zhou Z, Lu J, Liu WW, Manaenko A, Hou X, Mei Q, Huang JL, Tang J, Zhang JH, Yao H, Hu Q. Advances in stroke pharmacology. Pharmacol Ther 2018; 191:23-42. [PMID: 29807056 DOI: 10.1016/j.pharmthera.2018.05.012] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Stroke occurs when a cerebral blood vessel is blocked or ruptured, and it is the major cause of death and adult disability worldwide. Various pharmacological agents have been developed for the treatment of stroke either through interrupting the molecular pathways leading to neuronal death or enhancing neuronal survival and regeneration. Except for rtPA, few of these agents have succeeded in clinical trials. Recently, with the understanding of the pathophysiological process of stroke, there is a resurrection of research on developing neuroprotective agents for stroke treatment, and novel molecular targets for neuroprotection and neurorestoration have been discovered to predict or offer clinical benefits. Here we review the latest major progress of pharmacological studies in stroke, especially in ischemic stroke; summarize emerging potential therapeutic mechanisms; and highlight recent clinical trials. The aim of this review is to provide a panorama of pharmacological interventions for stroke and bridge basic and translational research to guide the clinical management of stroke therapy.
Collapse
Affiliation(s)
- Zhenhua Zhou
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA; Department of Neurology, Southwest Hospital, the Third Military Medical University, Chongqing 400038, China
| | - Jianfei Lu
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wen-Wu Liu
- Department of Diving and Hyperbaric Medicine, the Second Military Medical University, Shanghai 200433, China
| | - Anatol Manaenko
- Department of Neurology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Xianhua Hou
- Department of Neurology, Southwest Hospital, the Third Military Medical University, Chongqing 400038, China
| | - Qiyong Mei
- Department of Neurosurgery, Changzheng Hospital, the Second Military Medical University, Shanghai 200003, China
| | - Jun-Long Huang
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Honghong Yao
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, China.
| | - Qin Hu
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
38
|
Wang X, Yang J, Xing Z, Zhang H, Wen Y, Qi F, Zuo Z, Xu J, Yao Z. IL-4 mediates the delayed neurobehavioral impairments induced by neonatal hepatitis B vaccination that involves the down-regulation of the IL-4 receptor in the hippocampus. Cytokine 2018; 110:137-149. [PMID: 29751176 DOI: 10.1016/j.cyto.2018.04.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 04/29/2018] [Accepted: 04/30/2018] [Indexed: 01/23/2023]
Abstract
We have previously verified that neonatal hepatitis B vaccination induced hippocampal neuroinflammation and behavior impairments in mice. However, the exact mechanism of these effects remain unclear. In this study, we observed that neonatal hepatitis B vaccination induced an anti-inflammatory cytokine response lasting for 4-5 weeks in both the serum and the hippocampus, primarily indicated by elevated IL-4 levels. Three weeks after the vaccination schedule, however, hepatitis B vaccine (HBV)-mice showed delayed hippocampal neuroinflammation. In periphery, IL-4 is the major cytokine induced by this vaccine. Correlation analyses showed a positive relationship in the IL-4 levels between serum and hippocampus in HBV-mice. Thus, we investigated whether neonatal over-exposure to systemic IL-4 influences brain and behavior. We observed that mice injected intraperitoneally with recombinant mouse IL-4 (mIL-4) during early life had similar neuroinflammation and cognition impairment similar to those induced by neonatal hepatitis B vaccination. Next, the mechanism underlying the effects of IL-4 on brain in mice was explored using a series of experiments. In brief, these experiments showed that IL-4 mediates the delayed neurobehavioral impairments induced by neonatal hepatitis B vaccination, which involves the permeability of neonatal blood-brain barrier and the down-regulation of IL-4 receptor. This finding suggests that clinical events concerning neonatal IL-4 over-exposure, including neonatal hepatitis B vaccination and allergic asthma in human infants, may have adverse implications for brain development and cognition.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Junhua Yang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Zhiwei Xing
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Hongyang Zhang
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Yaru Wen
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Fangfang Qi
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Zejie Zuo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Jie Xu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China
| | - Zhibin Yao
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, PR China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, PR China.
| |
Collapse
|
39
|
Khoury N, Koronowski KB, Young JI, Perez-Pinzon MA. The NAD +-Dependent Family of Sirtuins in Cerebral Ischemia and Preconditioning. Antioxid Redox Signal 2018; 28:691-710. [PMID: 28683567 PMCID: PMC5824497 DOI: 10.1089/ars.2017.7258] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 07/04/2017] [Indexed: 12/11/2022]
Abstract
SIGNIFICANCE Sirtuins are an evolutionarily conserved family of NAD+-dependent lysine deacylases and ADP ribosylases. Their requirement for NAD+ as a cosubstrate allows them to act as metabolic sensors that couple changes in the energy status of the cell to changes in cellular physiological processes. NAD+ levels are affected by several NAD+-producing and NAD+-consuming pathways as well as by cellular respiration. Thus their intracellular levels are highly dynamic and are misregulated in a spectrum of metabolic disorders including cerebral ischemia. This, in turn, compromises several NAD+-dependent processes that may ultimately lead to cell death. Recent Advances: A number of efforts have been made to replenish NAD+ in cerebral ischemic injuries as well as to understand the functions of one its important mediators, the sirtuin family of proteins through the use of pharmacological modulators or genetic manipulation approaches either before or after the insult. Critical Issues and Future Directions: The results of these studies have regarded the sirtuins as promising therapeutic targets for cerebral ischemia. Yet, additional efforts are needed to understand the role of some of the less characterized members and to address the sex-specific effects observed with some members. Sirtuins also exhibit cell-type-specific expression in the brain as well as distinct subcellular and regional localizations. As such, they are involved in diverse and sometimes opposing cellular processes that can either promote neuroprotection or further contribute to the injury; which also stresses the need for the development and use of sirtuin-specific pharmacological modulators. Antioxid. Redox Signal. 28, 691-710.
Collapse
Affiliation(s)
- Nathalie Khoury
- Department of Neurology; Cerebral Vascular Research Laboratories; and Neuroscience Program, Miller School of Medicine, University of Miami, Miami, Florida
| | - Kevin B. Koronowski
- Department of Neurology; Cerebral Vascular Research Laboratories; and Neuroscience Program, Miller School of Medicine, University of Miami, Miami, Florida
| | - Juan I. Young
- Dr. John T. Macdonald Foundation Department of Human Genetics; Hussman Institute for Human Genomics, and Neuroscience Program, Miller School of Medicine, University of Miami, Miami, Florida
| | - Miguel A. Perez-Pinzon
- Department of Neurology; Cerebral Vascular Research Laboratories; and Neuroscience Program, Miller School of Medicine, University of Miami, Miami, Florida
| |
Collapse
|
40
|
Gong G, Yin L, Yuan L, Sui D, Sun Y, Fu H, Chen L, Wang X. Ganglioside GM1 protects against high altitude cerebral edema in rats by suppressing the oxidative stress and inflammatory response via the PI3K/AKT-Nrf2 pathway. Mol Immunol 2018; 95:91-98. [PMID: 29428576 DOI: 10.1016/j.molimm.2018.02.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 02/07/2023]
Abstract
High altitude cerebral edema (HACE) is a severe type of acute mountain sickness (AMS) that occurs in response to a high altitude hypobaric hypoxic (HH) environment. GM1 monosialoganglioside can alleviate brain injury under adverse conditions including amyloid-β-peptide, ischemia and trauma. However, its role in HACE-induced brain damage remains poorly elucidated. In this study, GM1 supplementation dose-dependently attenuated increase in rat brain water content (BWC) induced by hypobaric chamber (7600 m) exposurefor 24 h. Compared with the HH-treated group, rats injected with GM1 exhibited less brain vascular leakage, lower aquaporin-4 and higher occludin expression, but they also showed increase in Na+/K+-ATPase pump activities. Importantly, HH-incurred consciousness impairment and coordination loss also were ameliorated following GM1 administration. Furthermore, the increased oxidative stress and decrease in anti-oxidant stress system under the HH condition were also reversely abrogated by GM1 treatment via suppressing accumulation of ROS, MDA and elevating the levels of SOD and GSH. Simultaneously, GM1 administration also counteracted the enhanced inflammation in HH-exposed rats by muting pro-inflammatory cytokines IL-1β, TNF-α, and IL-6 levels in serum and brain tissues. Subsequently, GM1 potentiated the activation of the PI3K/AKT-Nrf2 pathway. Cessation of this pathway by LY294002 reversed GM1-mediated inhibitory effects on oxidative stress and inflammation, and ultimately abrogated the protective role of GM1 in abating brain edema, cognitive and motor dysfunction. Overall, GM1 may afford a protective intervention in HACE by suppressing oxidative stress and inflammatory response via activating the PI3K/AKT-Nrf2 pathway, implying a promising agent for the treatment of HACE.
Collapse
Affiliation(s)
- Gu Gong
- Department of Anesthesiology, General Hospital of Chengdu Military Region of PLA, Chengdu, 610083, PR China
| | - Liang Yin
- Department of Anesthesiology, General Hospital of Chengdu Military Region of PLA, Chengdu, 610083, PR China
| | - Libang Yuan
- Department of Anesthesiology, General Hospital of Chengdu Military Region of PLA, Chengdu, 610083, PR China
| | - Daming Sui
- Department of Anesthesiology, General Hospital of Chengdu Military Region of PLA, Chengdu, 610083, PR China
| | - Yangyang Sun
- Department of Anesthesiology, General Hospital of Chengdu Military Region of PLA, Chengdu, 610083, PR China
| | - Haiyu Fu
- Department of Anesthesiology, General Hospital of Chengdu Military Region of PLA, Chengdu, 610083, PR China
| | - Liang Chen
- Department of Anesthesiology, General Hospital of Chengdu Military Region of PLA, Chengdu, 610083, PR China
| | - Xiaowu Wang
- Center of Cardiovascular Surgery, General Hospital of Guangzhou Military Command of PLA, Guangzhou, Guangdong 510010, PR China.
| |
Collapse
|
41
|
SIRT3: A New Regulator of Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7293861. [PMID: 29643974 PMCID: PMC5831850 DOI: 10.1155/2018/7293861] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/20/2017] [Accepted: 01/04/2018] [Indexed: 01/13/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death worldwide, and defects in mitochondrial function contribute largely to the occurrence of CVDs. Recent studies suggest that sirtuin 3 (SIRT3), the mitochondrial NAD+-dependent deacetylase, may regulate mitochondrial function and biosynthetic pathways such as glucose and fatty acid metabolism and the tricarboxylic acid (TCA) cycle, oxidative stress, and apoptosis by reversible protein lysine deacetylation. SIRT3 regulates glucose and lipid metabolism and maintains myocardial ATP levels, which protects the heart from metabolic disturbances. SIRT3 can also protect cardiomyocytes from oxidative stress-mediated cell damage and block the development of cardiac hypertrophy. Recent reports show that SIRT3 is involved in the protection of several heart diseases. This review discusses the progress in SIRT3-related research and the role of SIRT3 in the prevention and treatment of CVDs.
Collapse
|
42
|
Evans NR, Tarkin JM, Buscombe JR, Markus HS, Rudd JHF, Warburton EA. PET imaging of the neurovascular interface in cerebrovascular disease. Nat Rev Neurol 2017; 13:676-688. [PMID: 28984315 DOI: 10.1038/nrneurol.2017.129] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cerebrovascular disease encompasses a range of pathologies that affect different components of the cerebral vasculature and brain parenchyma. Large artery atherosclerosis, acute cerebral ischaemia, and intracerebral small vessel disease all demonstrate altered metabolic processes that are key to their pathogenesis. Although structural imaging techniques such as MRI are the mainstay of clinical care and research in cerebrovascular disease, they have limited ability to detect these pathophysiological processes in vivo. By contrast, PET can detect and quantify metabolic processes that are relevant to each facet of cerebrovascular disease. Information obtained from PET studies has helped to shape the understanding of key concepts in cerebrovascular medicine, including vulnerable atherosclerotic plaque, salvageable ischaemic penumbra, neuroinflammation and selective neuronal loss after ischaemic insult. PET has also helped to elucidate the relationships between chronic hypoxia, neuroinflammation, and amyloid-β deposition in cerebral small vessel disease. This Review describes how PET-based imaging of metabolic processes at the neurovascular interface has contributed to our understanding of cerebrovascular disease.
Collapse
Affiliation(s)
- Nicholas R Evans
- Department of Clinical Neurosciences, University of Cambridge, Box 83, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Jason M Tarkin
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Box 157, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - John R Buscombe
- Department of Nuclear Medicine, Box 219, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, UK
| | - Hugh S Markus
- Department of Clinical Neurosciences, University of Cambridge, Box 83, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - James H F Rudd
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Box 157, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Elizabeth A Warburton
- Department of Clinical Neurosciences, University of Cambridge, Box 83, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK
| |
Collapse
|
43
|
Wei CC, Kong YY, Hua X, Li GQ, Zheng SL, Cheng MH, Wang P, Miao CY. NAD replenishment with nicotinamide mononucleotide protects blood-brain barrier integrity and attenuates delayed tissue plasminogen activator-induced haemorrhagic transformation after cerebral ischaemia. Br J Pharmacol 2017; 174:3823-3836. [PMID: 28812311 DOI: 10.1111/bph.13979] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 07/25/2017] [Accepted: 07/31/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Tissue plasminogen activator (tPA) is the only approved pharmacological therapy for acute brain ischaemia; however, a major limitation of tPA is the haemorrhagic transformation that follows tPA treatment. Here, we determined whether nicotinamide mononucleotide (NMN), a key intermediate of nicotinamide adenine dinucleotide biosynthesis, affects tPA-induced haemorrhagic transformation. EXPERIMENTAL APPROACH Middle cerebral artery occlusion (MCAO) was achieved in CD1 mice by introducing a filament to the left MCA for 5 h. When the filament was removed for reperfusion, tPA was infused via the tail vein. A single dose of NMN was injected i.p. (300 mg·kg-1 ). Mice were killed at 24 h post ischaemia, and their brains were evaluated for brain infarction, oedema, haemoglobin content, apoptosis, neuroinflammation, blood-brain barrier (BBB) permeability, the expression of tight junction proteins (TJPs) and the activity/expression of MMPs. KEY RESULTS In the mice infused with tPA at 5 h post ischaemia, there were significant increases in mortality, brain infarction, brain oedema, brain haemoglobin level, neural apoptosis, Iba-1 staining (microglia activation) and myeloperoxidase staining (neutrophil infiltration). All these tPA-induced alterations were significantly prevented by NMN administration. Mechanistically, the delayed tPA treatment increased BBB permeability by down-regulating TJPs, including claudin-1, occludin and zonula occludens-1, and enhancing the activities and protein expression of MMP9 and MMP2. Similarly, NMN administration partly blocked these tPA-induced molecular changes. CONCLUSIONS AND IMPLICATIONS Our results demonstrate that NMN ameliorates tPA-induced haemorrhagic transformation in brain ischaemia by maintaining the integrity of the BBB.
Collapse
Affiliation(s)
- Chun-Chun Wei
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Yuan-Yuan Kong
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Xia Hua
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Guo-Qiang Li
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Si-Li Zheng
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Ming-He Cheng
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | - Pei Wang
- Department of Pharmacology, Second Military Medical University, Shanghai, China.,Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Yantai University, Yantai, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University, Shanghai, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
44
|
Venkat P, Shen Y, Chopp M, Chen J. Cell-based and pharmacological neurorestorative therapies for ischemic stroke. Neuropharmacology 2017; 134:310-322. [PMID: 28867364 DOI: 10.1016/j.neuropharm.2017.08.036] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/22/2017] [Accepted: 08/24/2017] [Indexed: 01/09/2023]
Abstract
Ischemic stroke remains one of most common causes of death and disability worldwide. Stroke triggers a cascade of events leading to rapid neuronal damage and death. Neuroprotective agents that showed promise in preclinical experiments have failed to translate to the clinic. Even after decades of research, tPA remains the only FDA approved drug for stroke treatment. However, tPA is effective when administered 3-4.5 h after stroke onset and the vast majority of stroke patients do not receive tPA therapy. Therefore, there is a pressing need for novel therapies for ischemic stroke. Since stroke induces rapid cell damage and death, neuroprotective strategies that aim to salvage or replace injured brain tissue are challenged by treatment time frames. To overcome the barriers of neuroprotective therapies, there is an increasing focus on neurorestorative therapies for stroke. In this review article, we provide an update on neurorestorative treatments for stroke using cell therapy such as bone marrow derived mesenchymal stromal cells (BMSCs), human umbilical cord blood cells (HUCBCs) and select pharmacological approaches including Minocycline and Candesartan that have been employed in clinical trials. This review article discusses the present understanding of mechanisms of neurorestorative therapies and summarizes ongoing clinical trials. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
Collapse
Affiliation(s)
- Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Yi Shen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA; Gerontology Institute, Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA; Department of Physics, Oakland University, Rochester, MI, 48309, USA
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA; Gerontology Institute, Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China.
| |
Collapse
|
45
|
Effect of Oral Minocycline on Clinical Recovery Process in Patients with Acute Ischemic Stroke: A Randomized Clinical Trial. Jundishapur J Nat Pharm Prod 2017. [DOI: 10.5812/jjnpp.63792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
46
|
Manso Y, Holland PR, Kitamura A, Szymkowiak S, Duncombe J, Hennessy E, Searcy JL, Marangoni M, Randall AD, Brown JT, McColl BW, Horsburgh K. Minocycline reduces microgliosis and improves subcortical white matter function in a model of cerebral vascular disease. Glia 2017; 66:34-46. [PMID: 28722234 DOI: 10.1002/glia.23190] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/26/2017] [Accepted: 06/27/2017] [Indexed: 12/19/2022]
Abstract
Chronic cerebral hypoperfusion is a key mechanism associated with white matter disruption in cerebral vascular disease and dementia. In a mouse model relevant to studying cerebral vascular disease, we have previously shown that cerebral hypoperfusion disrupts axon-glial integrity and the distribution of key paranodal and internodal proteins in subcortical myelinated axons. This disruption of myelinated axons is accompanied by increased microglia and cognitive decline. The aim of the present study was to investigate whether hypoperfusion impairs the functional integrity of white matter, its relation with axon-glial integrity and microglial number, and whether by targeting microglia these effects can be improved. We show that in response to increasing durations of hypoperfusion, the conduction velocity of myelinated fibres in the corpus callosum is progressively reduced and that paranodal and internodal axon-glial integrity is disrupted. The number of microglial cells increases in response to hypoperfusion and correlates with disrupted paranodal and internodal integrity and reduced conduction velocities. Further minocycline, a proposed anti-inflammatory and microglia inhibitor, restores white matter function related to a reduction in the number of microglia. The study suggests that microglial activation contributes to the structural and functional alterations of myelinated axons induced by cerebral hypoperfusion and that dampening microglia numbers/proliferation should be further investigated as potential therapeutic benefit in cerebral vascular disease.
Collapse
Affiliation(s)
- Yasmina Manso
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| | - Philip R Holland
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| | - Akihiro Kitamura
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| | - Stefan Szymkowiak
- University of Edinburgh, The Roslin Institute, Easter Bush, Edinburgh, EH25 9RG
| | - Jessica Duncombe
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| | - Edel Hennessy
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| | - James L Searcy
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| | - Martina Marangoni
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| | - Andrew D Randall
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Jon T Brown
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Barry W McColl
- University of Edinburgh, The Roslin Institute, Easter Bush, Edinburgh, EH25 9RG.,UK Dementia Research Institute, University of Edinburgh, Edinburgh Medical School, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Karen Horsburgh
- Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, United Kingdom
| |
Collapse
|
47
|
Yang Q, Guo M, Wang X, Zhao Y, Zhao Q, Ding H, Dong Q, Cui M. Ischemic preconditioning with a ketogenic diet improves brain ischemic tolerance through increased extracellular adenosine levels and hypoxia-inducible factors. Brain Res 2017; 1667:11-18. [DOI: 10.1016/j.brainres.2017.04.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 03/18/2017] [Accepted: 04/15/2017] [Indexed: 02/07/2023]
|
48
|
Machado-Pereira M, Santos T, Bernardino L, Ferreira R. Vascular inter-regulation of inflammation: molecular and cellular targets for CNS therapy. J Neurochem 2016; 140:692-702. [PMID: 27925201 DOI: 10.1111/jnc.13914] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 11/07/2016] [Accepted: 11/21/2016] [Indexed: 12/17/2022]
Abstract
Angiogenesis and inflammation are clearly interconnected and interdependent processes that are dysregulated in a series of systemic and brain pathologies. Herein, key aspects regarding endothelial cell function and tissue remodelling that are particularly affected or aggravated by inflammation are presented. Most importantly, the cellular and molecular mechanisms involved in the vascular regulation of the inflammatory processes occurring in several brain disorders and how they impact on disease/injury progression are detailed, highlighting potential targets for therapy. Finally, nanomedicine-based approaches designed to overcome limitations pertaining to low systemic bioavailability, light, pH and temperature sensitivity and/or rapid degradation of these targets, and to optimize their mode of action are discussed. Ultimately, we expect this review to provide new insight and to suggest novel approaches for the treatment of blood-brain barrier dysfunction per se or as a means to treat the injured or diseased central nervous system.
Collapse
Affiliation(s)
- Marta Machado-Pereira
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Rua Marquês d'Ávila e Bolama, Covilhã, Portugal
| | - Tiago Santos
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Rua Marquês d'Ávila e Bolama, Covilhã, Portugal
| | - Liliana Bernardino
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Rua Marquês d'Ávila e Bolama, Covilhã, Portugal
| | - Raquel Ferreira
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Rua Marquês d'Ávila e Bolama, Covilhã, Portugal
| |
Collapse
|
49
|
Liu X, Cai X, Hu B, Mei Z, Zhang D, Ouyang G, Wang J, Zhang W, Xiao W. Forkhead Transcription Factor 3a (FOXO3a) Modulates Hypoxia Signaling via Up-regulation of the von Hippel-Lindau Gene (VHL). J Biol Chem 2016; 291:25692-25705. [PMID: 27777301 DOI: 10.1074/jbc.m116.745471] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/20/2016] [Indexed: 01/26/2023] Open
Abstract
FOXO3a, a member of the forkhead homeobox type O (FOXO) family of transcriptional factors, regulates cell survival in response to DNA damage, caloric restriction, and oxidative stress. The von Hippel-Lindau (VHL) tumor suppressor gene encodes a component of the E3 ubiquitin ligase complex that mediates hypoxia-inducible factor α degradation under aerobic conditions, thus acting as one of the key regulators of hypoxia signaling. However, whether FOXO3a impacts cellular hypoxia stress remains unknown. Here we show that FOXO3a directly binds to the VHL promoter and up-regulates VHL expression. Using a zebrafish model, we confirmed the up-regulation of vhl by foxo3b, an ortholog of mammalian FOXO3a Furthermore, by employing the clustered regularly interspaced short palindromic repeats (CRISPR)-associated RNA-guided endonuclease Cas9 (CRISPR/Cas9) technology, we deleted foxo3b in zebrafish and determined that expression of hypoxia-inducible genes was affected under hypoxia. Moreover, foxo3b-null zebrafish exhibited impaired acute hypoxic tolerance, resulting in death. In conclusion, our findings suggest that, by modulating hypoxia-inducible factor activity via up-regulation of VHL, FOXO3a (foxo3b) plays an important role in survival in response to hypoxic stress.
Collapse
Affiliation(s)
- Xing Liu
- From the Key Laboratory of Aquatic Biodiversity and Conservation and
| | - Xiaolian Cai
- From the Key Laboratory of Aquatic Biodiversity and Conservation and
| | - Bo Hu
- From the Key Laboratory of Aquatic Biodiversity and Conservation and
| | - Zhichao Mei
- From the Key Laboratory of Aquatic Biodiversity and Conservation and
| | - Dawei Zhang
- From the Key Laboratory of Aquatic Biodiversity and Conservation and
| | - Gang Ouyang
- From the Key Laboratory of Aquatic Biodiversity and Conservation and
| | - Jing Wang
- From the Key Laboratory of Aquatic Biodiversity and Conservation and
| | - Wei Zhang
- From the Key Laboratory of Aquatic Biodiversity and Conservation and
| | - Wuhan Xiao
- From the Key Laboratory of Aquatic Biodiversity and Conservation and .,State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| |
Collapse
|
50
|
Xu B, Tian R, Wang X, Zhan S, Wang R, Guo Y, Ge W. Protein profile changes in the frontotemporal lobes in human severe traumatic brain injury. Brain Res 2016; 1642:344-352. [PMID: 27067185 DOI: 10.1016/j.brainres.2016.04.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 12/20/2022]
Abstract
Severe traumatic brain injury (sTBI) is a serious public health issue with high morbidity and mortality rates. Previous proteomic studies on sTBI have mainly focused on human cerebrospinal fluid and serum, as well as on brain protein changes in murine models. However, human proteomic data in sTBI brain is still scarce. We used proteomic and bioinformatic strategies to investigate variations in protein expression levels in human brains after sTBI, using samples from the Department of Neurosurgery, Affiliated Hospital of Hebei University (Hebei, China). Our proteomic data identified 4031 proteins, of which 160 proteins were overexpressed and 5 proteins were downregulated. Bioinformatics analysis showed significant changes in biological pathways including glial cell differentiation, complement activation and apolipoprotein catalysis in the statin pathway. Western blot verification of protein changes in a subset of the available tissue samples showed results that were consistent with the proteomic data. This study is one of the first to investigate the whole proteome of human sTBI brains, and provide a characteristic signature and overall landscape of the sTBI brain proteome.
Collapse
Affiliation(s)
- Benhong Xu
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 071000, China; National Key Laboratory of Medical Molecular Biology and Department of Immunology; Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Rui Tian
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xia Wang
- National Key Laboratory of Medical Molecular Biology and Department of Immunology; Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Shaohua Zhan
- National Key Laboratory of Medical Molecular Biology and Department of Immunology; Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yi Guo
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 071000, China; Department of Neurosurgery, Tsinghua Changgung Hospital, Beijing 102218, China.
| | - Wei Ge
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 071000, China; National Key Laboratory of Medical Molecular Biology and Department of Immunology; Institute of Basic Medical Sciences; Chinese Academy of Medical Sciences, Beijing 100005, China.
| |
Collapse
|