1
|
Wu Y, Wu Y, Yu J, Zhang Y, Dai X, Chen J, Sun Y, Yang Y, Zhao K, Xiao Q. Irisin alters D-galactose-induced apoptosis by increasing caveolin-1 expression in C2C12 myoblasts and skeletal muscle fibroblasts. Mol Cell Biochem 2025; 480:577-588. [PMID: 38581552 DOI: 10.1007/s11010-024-04990-6] [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: 12/04/2023] [Accepted: 03/15/2024] [Indexed: 04/08/2024]
Abstract
Muscle atrophy and skeletal muscle fibrosis are significant pathological manifestations of primary sarcopenia. The regulation of C2C12 myoblast and skeletal muscle fibroblast apoptosis is associated with these pathological changes. Previous studies have indicated that irisin, the cleaved form of fibronectin type III domain-containing protein 5 (FNDC5), can alleviate primary sarcopenia. However, the mechanisms of the effect of irisin in age-related apoptosis remain unknown. Our present research aimed to explore the effect of irisin and the underlying mechanism of D-galactose (D-gal)-induced apoptosis in skeletal muscle fibroblasts and C2C12 myoblasts. We found the opposite effects of D-gal on C2C12 myoblasts and fibroblasts. We also found that irisin suppressed C2C12 cell apoptosis and promoted fibroblast apoptosis. Mechanistically, irisin altered D-gal-induced apoptosis by increasing caveolin-1 expression. Taken together, these findings further demonstrated that irisin is a potential agent that can treat aged-relative muscle atrophy and fibrosis.
Collapse
Affiliation(s)
- Yaoxuan Wu
- Geriatric Diseases Institute of Chengdu, Department of Geriatrics, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611137, China
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China
| | - Yongxin Wu
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China
| | - Jing Yu
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China
| | - Yingxiao Zhang
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China
| | - Xin Dai
- Department of General Practice, Yongchuan Hospital of Chongqing Medical University, Chongqing Medical University, Chongqing, 400010, China
| | - Jinliang Chen
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310001, China
| | - Yue Sun
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China
| | - Yongxue Yang
- Geriatric Diseases Institute of Chengdu, Department of Geriatrics, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, 611137, China.
| | - Kexiang Zhao
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China.
| | - Qian Xiao
- Department of Geriatrics, The First Affiliated Hospital of Chongqing Medical University, 1St You Yi Road, Yu Zhong District, Chongqing, 400010, China
| |
Collapse
|
2
|
Zhang C, Zheng J, Yu X, Kuang B, Dai X, Zheng L, Yu W, Teng W, Cao H, Li M, Yao J, Liu X, Zou W. "Baihui" (DU20)-penetrating "Qubin" (GB7) acupuncture on blood-brain barrier integrity in rat intracerebral hemorrhage models via the RhoA/ROCK II/MLC 2 signaling pathway. Animal Model Exp Med 2024; 7:740-757. [PMID: 38379356 PMCID: PMC11528382 DOI: 10.1002/ame2.12374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 11/21/2023] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Blocking the RhoA/ROCK II/MLC 2 (Ras homolog gene family member A/Rho kinase II/myosin light chain 2) signaling pathway can initiate neuroprotective mechanisms against neurological diseases such as stroke, cerebral ischemia, and subarachnoid hemorrhage. Nevertheless, it is not clear whether and how disrupting the RhoA/ROCK II/MLC 2 signaling pathway changes the pathogenic processes of the blood-brain barrier (BBB) after intracerebral hemorrhage (ICH). The present investigation included the injection of rat caudal vein blood into the basal ganglia area to replicate the pathophysiological conditions caused by ICH. METHODS Scalp acupuncture (SA) therapy was performed on rats with ICH at the acupuncture point "Baihui"-penetrating "Qubin," and the ROCK selective inhibitor fasudil was used as a positive control to evaluate the inhibitory effect of acupuncture on the RhoA/ROCK II/MLC 2 signaling pathway. Post-assessments included neurological deficits, brain edema, Evans blue extravasation, Western blot, quantitative polymerase chain reaction, and transmission electron microscope imaging. RESULTS We found that ROCK II acts as a promoter of the RhoA/ROCK II/MLC 2 signaling pathway, and its expression increased at 6 h after ICH, peaked at 3 days, and then decreased at 7 days after ICH, but was still higher than the pre-intervention level. According to some experimental results, although 3 days is the peak, 7 days is the best time point for acupuncture treatment. Starting from 6 h after ICH, the neurovascular structure and endothelial cell morphology around the hematoma began to change. Based on the changes in the promoter ROCK II, a 7-day time point was selected as the breakthrough point for treating ICH model rats in the main experiment. The results of this experiment showed that both SA at "Baihui"-penetrating "Qubin" and treatment with fasudil could improve the expression of endothelial-related proteins by inhibiting the RhoA/ROCK II/MLC 2 signaling pathway and reduce neurological dysfunction, brain edema, and BBB permeability in rats. CONCLUSION This study found that these experimental data indicated that SA at "Baihui"-penetrating "Qubin" could preserve BBB integrity and neurological function recovery after ICH by inhibiting RhoA/ROCK II/MLC 2 signaling pathway activation and by regulating endothelial cell-related proteins.
Collapse
Affiliation(s)
- Ce Zhang
- Heilongjiang University of Chinese MedicineHarbinChina
| | - Jia Zheng
- Heilongjiang University of Chinese MedicineHarbinChina
| | - Xueping Yu
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Binglin Kuang
- Heilongjiang University of Chinese MedicineHarbinChina
| | - Xiaohong Dai
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Lei Zheng
- Clinical Key Laboratory of Integrated Traditional Chinese and Western Medicine of Heilongjiang University of Chinese MedicineHarbinChina
| | - Weiwei Yu
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Wei Teng
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Hongtao Cao
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Mingyue Li
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Jiayong Yao
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Xiaoying Liu
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| | - Wei Zou
- First Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| |
Collapse
|
3
|
Badaut J, Blochet C, Obenaus A, Hirt L. Physiological and pathological roles of caveolins in the central nervous system. Trends Neurosci 2024; 47:651-664. [PMID: 38972795 PMCID: PMC11324375 DOI: 10.1016/j.tins.2024.06.003] [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: 06/27/2023] [Revised: 05/14/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024]
Abstract
Caveolins are a family of transmembrane proteins located in caveolae, small lipid raft invaginations of the plasma membrane. The roles of caveolin-enriched lipid rafts are diverse, and include mechano-protection, lipid homeostasis, metabolism, transport, and cell signaling. Caveolin-1 (Cav-1) and other caveolins were described in endothelial cells and later in other cell types of the central nervous system (CNS), including neurons, astrocytes, oligodendrocytes, microglia, and pericytes. This pancellular presence of caveolins demands a better understanding of their functional roles in each cell type. In this review we describe the various functions of Cav-1 in the cells of normal and pathological brains. Several emerging preclinical findings suggest that Cav-1 could represent a potential therapeutic target in brain disorders.
Collapse
Affiliation(s)
- Jérôme Badaut
- CNRS UMR 5536 RMSB-University of Bordeaux, Bordeaux, France; Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Camille Blochet
- Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland; Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - André Obenaus
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA; Division of Biomedical Sciences, University of California Riverside, Riverside, CA, USA
| | - Lorenz Hirt
- Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland; Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
4
|
Guo P, Wang W, Liang Z, Li Y, Ou X, Li M, Wang B, Wei X, Huang L, Qi S. Disintegration of Cav-1/β-catenin complex attenuates neuronal death after ischemia-reperfusion injury by promoting β-catenin nuclear translocation. Mol Biol Rep 2024; 51:829. [PMID: 39037581 DOI: 10.1007/s11033-024-09798-7] [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: 01/10/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND The roles of Caveolin-1 (Cav-1) and the Wnt/β-catenin signaling pathways in cerebral ischemia-reperfusion (I/R) injury are well established. The translocation of β-catenin into the nucleus is critical for regulating neuronal apoptosis, repair, and neurogenesis within the ischemic brain. It has been reported that the scaffold domain of Caveolin-1 (Cav-1) (residues 95-98) interacts with β-catenin (residues 330-337). However, the specific contribution of the Cav-1/β-catenin complex to I/R injury remains unknown. METHODS AND RESULTS To investigate the mechanism underlying the involvement of the Cav-1/β-catenin complex in the subcellular translocation of β-catenin and its subsequent effects on cerebral I/R injury, we treated ischemic brains with ASON (Cav-1 antisense oligodeoxynucleotides) or FTVT (a competitive peptide antagonist of the Cav-1 and β-catenin interaction). Our study demonstrated that the binding of Cav-1 to β-catenin following I/R injury prevented the nuclear accumulation of β-catenin. Treatment with ASON or FTVT after I/R injury significantly increased the levels of nuclear β-catenin. Furthermore, ASON reduced the phosphorylation of β-catenin at Ser33, Ser37, and Thr41, which contributes to its proteasomal degradation, while FTVT increased phosphorylation at Tyr333, which is associated with its nuclear translocation. CONCLUSIONS The above results indicate that the formation of the Cav-1/β-catenin complex anchors β-catenin in the cytoplasm following I/R injury. Additionally, both ASON and FTVT treatments attenuated neuronal death in ischemic brains. Our study suggests that targeting the interaction between Cav-1 and β-catenin serve as a novel therapeutic strategy to protect against neuronal damage during cerebral injury.
Collapse
Affiliation(s)
- Peng Guo
- Department of Laboratory Medicine, Jinhu County People's Hospital, Huai'an, 211600, People's Republic of China
- Research Center for Biochemistry and Molecular Biology and Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Wan Wang
- Xuzhou Key Laboratory of Laboratory Diagnostics, Medical Technology School of Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Zhiyan Liang
- Xuzhou Key Laboratory of Laboratory Diagnostics, Medical Technology School of Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Yihang Li
- Research Center for Biochemistry and Molecular Biology and Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Xiangling Ou
- Affiliated Xuzhou Rehabilitation Hospital of Xuzhou Medical University, Xuzhou, 221002, People's Republic of China
| | - Ming Li
- Xuzhou Key Laboratory of Laboratory Diagnostics, Medical Technology School of Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Bin Wang
- Department of Laboratory Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, People's Republic of China
| | - Xuewen Wei
- Department of Laboratory Medicine, Affiliated Municipal First People's Hospital of Xuzhou Medical University, Xuzhou, 221002, People's Republic of China
| | - Linyan Huang
- Xuzhou Key Laboratory of Laboratory Diagnostics, Medical Technology School of Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Suhua Qi
- Department of Laboratory Medicine, Jinhu County People's Hospital, Huai'an, 211600, People's Republic of China.
- Xuzhou Key Laboratory of Laboratory Diagnostics, Medical Technology School of Xuzhou Medical University, Xuzhou, 221004, People's Republic of China.
- Research Center for Biochemistry and Molecular Biology and Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China.
| |
Collapse
|
5
|
Huang F, Mao F, Nong W, Gong Z, Lao D, Huang W. Inhibiting Caveolin-1-Related Akt/mTOR Signaling Pathway Protects Against N-methyl-D-Aspartate Receptor Activation-Mediated Dysfunction of Blood-Brain Barrier in vitro. Mol Neurobiol 2024; 61:4166-4177. [PMID: 38066401 PMCID: PMC11236913 DOI: 10.1007/s12035-023-03833-7] [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: 07/06/2023] [Accepted: 11/22/2023] [Indexed: 07/11/2024]
Abstract
BACKGROUND The aim of this study was to further explore the role of caveolin-1 (Cav-1) related Akt/mTOR signaling pathway in blood brain barrier (BBB) dysfunction caused by NMDAR activation. METHODS The cell localization of NMDAR GluN1 subunit and Cav-1 was observed on human brain microvascular HBEC-5i cells after immunofluorescence double staining. The transendothelial resistance (TEER) of BBB in vitro was measured by Millicell-ERS cell resistance meter. Sodium fluorescein (SF) was used to measure the permeability of BBB in vitro. A stable Cav-1-silenced HBEC-5i cell line was established by infecting the cells with a lentivirus encoding Cav-1 shRNA. The changes of the protein and mRNA of MMP9 and Occludin induced by NMDA were detected by Western blot (WB) and real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR), respectively. The phosphorylated proteins of Cav-1, Akt, and mTOR were detected by WB. RESULTS NMDAR GluN1 was expressed in the cytoplasm and part of the cell membrane of the HBEC-5i cell line. NMDAR activation decreased TEER and increased the SF of BBB in vitro. HBEC-5i cells incubated with NMDA enhanced the phosphorylation of Cav-1, Akt, and mTOR, also promoting the expression of MMP9 along with the degradation of Occludin. These effects could be reversed by pretreatment with NMDAR antagonist (MK801) or Cav-1 antagonist (Daidzein), or Akt antagonist (LY294002), respectively. Further silencing Cav-1 with LV-Cav-1-RNAi also played a similar protective effect. CONCLUSION Caveolin-1 (Cav-1) related Akt/mTOR signaling probably contributes to BBB dysfunction by activating NMDAR on human brain microvascular cells.
Collapse
Affiliation(s)
- Fang Huang
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Fengping Mao
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Weidong Nong
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Zhuowei Gong
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Dayuan Lao
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Wen Huang
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, #6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
6
|
Xi Y, Feng Z, Xia T, Hong Y, Wu J, Chen J, Ge Y, Xiao H. Caveolin-1 scaffolding domain-derived peptide enhances erectile function by regulating oxidative stress, mitochondrial dysfunction, and apoptosis of corpus cavernosum smooth muscle cells in rats with cavernous nerve injury. Life Sci 2024; 348:122694. [PMID: 38718855 DOI: 10.1016/j.lfs.2024.122694] [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: 03/07/2024] [Revised: 04/23/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024]
Abstract
AIM Increased corpus cavernosum smooth muscle cells (CCSMCs) apoptosis in the penis due to cavernous nerve injury (CNI) is a crucial contributor to erectile dysfunction (ED). Caveolin-1 scaffolding domain (CSD)-derived peptide has been found to exert potential antiapoptotic properties. However, whether CSD peptide can alleviate CCSMCs apoptosis and ED in CNI rats remains unknown. The study aimed to determine whether CSD peptide can improve bilateral CNI-induced ED (BCNI-ED) by enhancing the antiapoptotic processes of CCSMCs. MAIN METHODS Fifteen 10-week-old male Sprague-Dawley (SD) rats were randomly classified into three groups: sham surgery (Sham) group and BCNI groups that underwent saline or CSD peptide treatment respectively. At 3 weeks postoperatively, erectile function was assessed and the penis tissue was histologically examined. Furthermore, an in vitro model of CCSMCs apoptosis was established using transforming growth factor-beta 1 (TGF-β1) to investigate the mechanism of CSD peptide in treating BCNI-ED. KEY FINDINGS In BCNI rats, CSD peptide significantly prevented ED and decreased oxidative stress, the Bax/Bcl-2 ratio, and the levels of caspase3. TGF-β1-treated CCSMCs exhibited severe oxidative stress, mitochondrial dysfunction, and apoptosis. However, CSD peptide partially reversed these alterations. SIGNIFICANCE Exogenous CSD peptide could improve BCNI-ED by inhibiting oxidative stress, the Bax/Bcl-2 ratio, and caspase3 expression in penile tissue. The underlying mechanism might involve the regulatory effects of CSD peptide on oxidative stress, mitochondrial dysfunction, and apoptosis of CCSMCs following CNI. This study highlights CSD peptide as an effective therapy for post-radical prostatectomy ED (pRP-ED).
Collapse
Affiliation(s)
- Yuhang Xi
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510000, China
| | - Zejia Feng
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510000, China
| | - Tian Xia
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510000, China
| | - Yude Hong
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510000, China
| | - Jianjie Wu
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510000, China
| | - Jialiang Chen
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510000, China
| | - Yunlong Ge
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510000, China; Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230000, China.
| | - Hengjun Xiao
- Department of Urology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510000, China.
| |
Collapse
|
7
|
Wu D, Zheng K, Yin W, Hu B, Yu M, Yu Q, Wei X, Deng J, Zhang C. Enhanced osteochondral regeneration with a 3D-Printed biomimetic scaffold featuring a calcified interfacial layer. Bioact Mater 2024; 36:317-329. [PMID: 38496032 PMCID: PMC10940945 DOI: 10.1016/j.bioactmat.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024] Open
Abstract
The integrative regeneration of both articular cartilage and subchondral bone remains an unmet clinical need due to the difficulties of mimicking spatial complexity in native osteochondral tissues for artificial implants. Layer-by-layer fabrication strategies, such as 3D printing, have emerged as a promising technology replicating the stratified zonal architecture and varying microstructures and mechanical properties. However, the dynamic and circulating physiological environments, such as mass transportation or cell migration, usually distort the pre-confined biological properties in the layered implants, leading to undistinguished spatial variations and subsequently inefficient regenerations. This study introduced a biomimetic calcified interfacial layer into the scaffold as a compact barrier between a cartilage layer and a subchondral bone layer to facilitate osteogenic-chondrogenic repair. The calcified interfacial layer consisting of compact polycaprolactone (PCL), nano-hydroxyapatite, and tasquinimod (TA) can physically and biologically separate the cartilage layer (TA-mixed, chondrocytes-load gelatin methacrylate) from the subchondral bond layer (porous PCL). This introduction preserved the as-designed independent biological environment in each layer for both cartilage and bone regeneration, successfully inhibiting vascular invasion into the cartilage layer and preventing hyaluronic cartilage calcification owing to devascularization of TA. The improved integrative regeneration of cartilage and subchondral bone was validated through gross examination, micro-computed tomography (micro-CT), and histological and immunohistochemical analyses based on an in vivo rat model. Moreover, gene and protein expression studies identified a key role of Caveolin (CAV-1) in promoting angiogenesis through the Wnt/β-catenin pathway and indicated that TA in the calcified layer blocked angiogenesis by inhibiting CAV-1.
Collapse
Affiliation(s)
- Di Wu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.600 Yishan Road, Shanghai, 200233, China
| | - Kaiwen Zheng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.600 Yishan Road, Shanghai, 200233, China
| | - Wenjing Yin
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.600 Yishan Road, Shanghai, 200233, China
| | - Bin Hu
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.600 Yishan Road, Shanghai, 200233, China
| | - Mingzhao Yu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.600 Yishan Road, Shanghai, 200233, China
| | - Qingxiao Yu
- Shanghai Uniorlechnology Corporation, No. 258 Xinzhuan Road, Shanghai, 201612, China
| | - Xiaojuan Wei
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.600 Yishan Road, Shanghai, 200233, China
| | - Jue Deng
- Academy for Engineering & Technology, Fudan University, No. 220 Handan Road, Shanghai, 200433, China
| | - Changqing Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, No.600 Yishan Road, Shanghai, 200233, China
| |
Collapse
|
8
|
Chen AC, Lai SC, Lu CY, Chen KM. Exploration of the Molecular Mechanism by Which Caveolin-1 Regulates Changes in Blood-Brain Barrier Permeability Leading to Eosinophilic Meningoencephalitis. Trop Med Infect Dis 2024; 9:124. [PMID: 38922036 PMCID: PMC11209224 DOI: 10.3390/tropicalmed9060124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/27/2024] Open
Abstract
Angiostrongylus cantonensis, a zoonotic parasite, can invade the human central nervous system (CNS) and cause acute eosinophilic meningitis or eosinophilic meningoencephalitis. Mice infected with A. cantonensis show elevated levels of pro-inflammatory cytokines, plasminogen activators, and matrix metalloproteinase-9, resulting in disruption of the blood-brain barrier (BBB) and immune cell infiltration into the CNS. Caveolin-1 (Cav-1) regulates the permeability of the BBB, which affects immune cells and cerebrospinal fluid. This intricate interaction ultimately fuels the progression of brain damage and edema. This study aims to investigate the regulatory role of Cav-1 in the pathogenesis of meningoencephalitis induced by A. cantonensis infection. We investigated pathological alterations by triphenyl-tetrazolium chloride, brain water content, BBB permeability, Western blot analysis, and gelatin zymography in BALB/c mice after A. cantonensis. The study evaluates the critical role of Cav-1 regulation through the TLR4/MyD88 signaling pathway, modulates tight junction proteins, influences BBB permeability, and contributes to brain damage in A. cantonensis-induced meningoencephalitis.
Collapse
Affiliation(s)
- An-Chih Chen
- Department of Neurology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan;
| | - Shih-Chan Lai
- Department of Parasitology, Chung Shan Medical University, Taichung 40201, Taiwan;
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Cheng-You Lu
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan;
| | - Ke-Min Chen
- Department of Parasitology, Chung Shan Medical University, Taichung 40201, Taiwan;
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| |
Collapse
|
9
|
Gu C, Liu Y, An X, Yin G, Sun C. Dysregulated SYVN1 promotes CAV1 protein ubiquitination and accentuates ischemic stroke. J Stroke Cerebrovasc Dis 2024; 33:107668. [PMID: 38423151 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Stroke is a major cause of death and severe disability, and there remains a substantial need for the development of therapeutic agents for neuroprotection in acute ischemic stroke (IS) to protect the brain against damage before and during recanalization. Caveolin-1 (CAV1), an integrated protein that is located at the caveolar membrane, has been reported to exert neuroprotective effects during IS. Nevertheless, the mechanism remains largely unknown. Here, we explored the upstream modifiers of CAV1 in IS. METHODS E3 ubiquitin ligases of CAV1 that are differentially expressed in IS were screened using multiple databases. The transcription factor responsible for the dysregulation of E3 ubiquitin-protein ligase synoviolin (SYVN1) in IS was predicted and verified. Genetic manipulations by lentiviral vectors were applied to investigate the effects of double-strand-break repair protein rad21 homolog (RAD21), SYVN1, and CAV1 in a middle cerebral artery occlusion (MCAO) mouse model and mouse HT22 hippocampal neurons induced by oxygen-glucose deprivation (OGD). RESULTS SYVN1 was highly expressed in mice with MCAO, and knockdown of SYVN1 alleviated IS injury in mice, as evidenced by limited infarction volume, the lower water content in the brain, and repressed apoptosis and inflammatory response. RAD21 inhibited the transcription of SYVN1, thereby reducing the ubiquitination modification of CAV1. Overexpression of RAD21 elicited a neuroprotective role as well in mice with MCAO and HT22 induced with OGD, which was overturned by SYVN1. CONCLUSION Transcriptional repression of SYVN1 by RAD21 alleviates IS in mice by reducing ubiquitination modification of CAV1.
Collapse
Affiliation(s)
- Chunjie Gu
- Department of Neurology, The First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang, China.
| | - Yang Liu
- Department of Rheumatology, The First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang, China.
| | - Xiuli An
- Department of Neurology, The Second Hospital of Harbin, Harbin 150056, Heilongjiang, China.
| | - Gang Yin
- Department of Neurology, The First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang, China.
| | - Chenghe Sun
- Department of Neurology, The First Hospital of Qiqihar, Qiqihar 161005, Heilongjiang, China.
| |
Collapse
|
10
|
Bindal P, Kumar V, Kapil L, Singh C, Singh A. Therapeutic management of ischemic stroke. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2651-2679. [PMID: 37966570 DOI: 10.1007/s00210-023-02804-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/19/2023] [Indexed: 11/16/2023]
Abstract
Stroke is the third leading cause of years lost due to disability and the second-largest cause of mortality worldwide. Most occurrences of stroke are brought on by the sudden occlusion of an artery (ischemic stroke), but sometimes they are brought on by bleeding into brain tissue after a blood vessel has ruptured (hemorrhagic stroke). Alteplase is the only therapy the American Food and Drug Administration has approved for ischemic stroke under the thrombolysis category. Current views as well as relevant clinical research on the diagnosis, assessment, and management of stroke are reviewed to suggest appropriate treatment strategies. We searched PubMed and Google Scholar for the available therapeutic regimes in the past, present, and future. With the advent of endovascular therapy in 2015 and intravenous thrombolysis in 1995, the therapeutic options for ischemic stroke have expanded significantly. A novel approach such as vagus nerve stimulation could be life-changing for many stroke patients. Therapeutic hypothermia, the process of cooling the body or brain to preserve organ integrity, is one of the most potent neuroprotectants in both clinical and preclinical contexts. The rapid intervention has been linked to more favorable clinical results. This study focuses on the pathogenesis of stroke, as well as its recent advancements, future prospects, and potential therapeutic targets in stroke therapy.
Collapse
Affiliation(s)
- Priya Bindal
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Vishal Kumar
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Lakshay Kapil
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India
| | - Charan Singh
- Department of Pharmaceutical Sciences, HNB Garhwal University (A Central University), Chauras Campus, Distt. Tehri Garhwal, Uttarakhand, 246174, India
| | - Arti Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga 142001, Affiliated to I.K Gujral Punjab Technical University, Jalandhar, Punjab, India.
| |
Collapse
|
11
|
Wang X, Yu Z, Dong F, Li J, Niu P, Ta Q, Kan J, Ma C, Han M, Yu J, Zhao D, Li J. Clarifying the mechanism of apigenin against blood-brain barrier disruption in ischemic stroke using systems pharmacology. Mol Divers 2024; 28:609-630. [PMID: 36949297 DOI: 10.1007/s11030-023-10607-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/12/2023] [Indexed: 03/24/2023]
Abstract
Currently, recombinant tissue plasminogen activator (rtPA) is an effective therapy for ischemic stroke (IS). However, blood-brain barrier (BBB) disruption is a serious side effect of rtPA therapy and may lead to patients' death. The natural polyphenol apigenin has a good therapeutic effect on IS. Apigenin has potential BBB protection, but the mechanism by which it protects the BBB integrity is not clear. In this study, we used network pharmacology, bioinformatics, molecular docking and molecular dynamics simulation to reveal the mechanisms by which apigenin protects the BBB. Among the 146 targets of apigenin for the treatment of IS, 20 proteins were identified as core targets (e.g., MMP-9, TLR4, STAT3). Apigenin protects BBB integrity by inhibiting the activity of MMPs through anti-inflammation and anti-oxidative stress. These mechanisms included JAK/STAT, the toll-like receptor signaling pathway, and Nitrogen metabolism signaling pathways. The findings of this study contribute to a more comprehensive understanding of the mechanism of apigenin in the treatment of BBB disruption and provide ideas for the development of drugs to treat IS.
Collapse
Affiliation(s)
- Xu Wang
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China
| | - ZiQiao Yu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Fuxiang Dong
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Jinjian Li
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Ping Niu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Qiyi Ta
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - JunMing Kan
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Chunyu Ma
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Moxuan Han
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Junchao Yu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China
| | - Dexi Zhao
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130117, Jilin, China.
| | - Jinhua Li
- School of Public Health, Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
12
|
Thomas RG, Kim JH, Kim JH, Yoon J, Choi KH, Jeong YY. Treatment of Ischemic Stroke by Atorvastatin-Loaded PEGylated Liposome. Transl Stroke Res 2024; 15:388-398. [PMID: 36639607 DOI: 10.1007/s12975-023-01125-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/16/2022] [Accepted: 01/04/2023] [Indexed: 01/15/2023]
Abstract
There is insufficient evidence on the effect of nanoparticles, particularly liposomes loaded with a statin, on acute ischemic stroke. We investigated the impact of atorvastatin-loaded PEG (polyethylene glycol) conjugated liposomes (LipoStatin) on the outcomes in rats with cerebral ischemia-reperfusion. PEGylated liposome loaded with atorvastatin was developed as a nanoparticle to specifically accumulate in an ischemic region and release the drug to ameliorate the harmful effects of the stroke. LipoStatin was administered to rats with transient middle cerebral artery occlusion through the tail vein immediately after reperfusion (LipoStatin group). LipoStatin efficiently accumulated at the cerebral ischemic injury site of the rat. The LipoStatin group showed a significantly reduced infarct volume (p < 0.01) in brain micro-MR imaging and improved neurological function recovery compared to the control group (p < 0.05). In addition, markedly improved brain metabolism using fluorine-18 fluorodeoxyglucose micro-PET/CT imaging was demonstrated in the LipoStatin group compared with the control group (p < 0.01). Mechanistically, as a result of evaluation through IL-1 beta, TNF-alpha, ICAM-1, and Iba-1 mRNA expression levels at 5 days after cerebral ischemia, LipoStatin showed significant anti-inflammatory effects. Protein expression of occludin, JAM-A, Caveolin-1, and eNOS by western blot at 3 days and fluorescent images at 7 days showed considerable recovery of blood-brain barrier breakdown and endothelial dysfunction. PEGylated LipoStatin can be more effectively delivered to the ischemic brain and may have significant neuroprotective effects. Thus, PEGylated LipoStatin can be further developed as a promising targeted therapy for ischemic stroke and other major vascular diseases.
Collapse
Affiliation(s)
- Reju George Thomas
- Department of Radiology, Chonnam National University Medical School and Hwasun Hospital, 322 Seoyang-Ro, Hwasun-Eup, Hwasun-Gun, Jeollanam-Do, 58128, South Korea
| | - Ja-Hae Kim
- Department of Nuclear Medicine, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| | - Ji-Hye Kim
- Department of Neurology, Chonnam National University Medical School and Hwasun Hospital, 322 Seoyang-Ro, Hwasun-Eup, Hwasun-Gun, Jeollanam-Do, 58128, South Korea
| | - Jungwon Yoon
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Kang-Ho Choi
- Department of Neurology, Chonnam National University Medical School and Hwasun Hospital, 322 Seoyang-Ro, Hwasun-Eup, Hwasun-Gun, Jeollanam-Do, 58128, South Korea.
| | - Yong-Yeon Jeong
- Department of Radiology, Chonnam National University Medical School and Hwasun Hospital, 322 Seoyang-Ro, Hwasun-Eup, Hwasun-Gun, Jeollanam-Do, 58128, South Korea.
| |
Collapse
|
13
|
Li Y, Chen J, Quan X, Chen Y, Han Y, Chen J, Yang L, Xu Y, Shen X, Wang R, Zhao Y. Extracellular Vesicles Maintain Blood-Brain Barrier Integrity by the Suppression of Caveolin-1/CD147/VEGFR2/MMP Pathway After Ischemic Stroke. Int J Nanomedicine 2024; 19:1451-1467. [PMID: 38371456 PMCID: PMC10874237 DOI: 10.2147/ijn.s444009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 02/08/2024] [Indexed: 02/20/2024] Open
Abstract
Background Ischemic stroke (IS) causes tragic death and disability worldwide. However, effective therapeutic interventions are finite. After IS, blood-brain barrier (BBB) integrity is disrupted, resulting in deteriorating neurological function. As a novel therapeutic, extracellular vesicles (EVs) have shown ideal restorative effects on BBB integrity post-stroke; however, the definite mechanisms remain ambiguous. In the present study, we investigated the curative effects and the mechanisms of EVs derived from bone marrow mesenchymal stem cells and brain endothelial cells (BMSC-EVs and BEC-EVs) on BBB integrity after acute IS. Methods EVs were isolated from BMSCs and BECs, and we investigated the therapeutic effect in vitro oxygen-glucose deprivation (OGD) insulted BECs model and in vivo rat middle cerebral artery occlusion (MCAo) model. The cell monolayer leakage, tight junction expression, and metalloproteinase (MMP) activity were evaluated, and rat brain infarct volume and neurological function were also analyzed. Results The administration of two kinds of EVs not only enhanced ZO-1 and Occludin expressions but also reduced the permeability and the activity of MMP-2/9 in OGD-insulted BECs. The amelioration of the cerebral infarction, BBB leakage, neurological function deficits, and the increasing ZO-1 and Occludin levels, as well as MMP activity inhibition was observed in MCAo rats. Additionally, the increased levels of Caveolin-1, CD147, vascular endothelial growth factor receptor 2 (VEGFR2), and vascular endothelial growth factor A (VEGFA) in isolated brain microvessels were downregulated after EVs treatment. In vitro, the employment of Caveolin-1 and CD147 siRNA partly suppressed the expressions of VEGFR2, VEGFA and MMP-2/9 activity and reduced the leakage of OGD insulted BECs and enhanced ZO-1 and Occludin expressions. Conclusion Our study firstly demonstrates that BEC and BMSC-EVs administrations maintain BBB integrity via the suppression of Caveolin-1/CD147/VEGFR2/MMP pathway after IS, and the efficacy of BMSC-EVs is superior to that of BEC-EVs.
Collapse
Affiliation(s)
- Yiyang Li
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Jiali Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Xingping Quan
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Ying Chen
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Yan Han
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Jinfen Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Li Yang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Youhua Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, People’s Republic of China
| | - Xu Shen
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Ruibing Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Yonghua Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, People’s Republic of China
| |
Collapse
|
14
|
Samhan-Arias AK, Poejo J, Marques-da-Silva D, Martínez-Costa OH, Gutierrez-Merino C. Are There Lipid Membrane-Domain Subtypes in Neurons with Different Roles in Calcium Signaling? Molecules 2023; 28:7909. [PMID: 37894616 PMCID: PMC10708093 DOI: 10.3390/molecules28237909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/24/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Lipid membrane nanodomains or lipid rafts are 10-200 nm diameter size cholesterol- and sphingolipid-enriched domains of the plasma membrane, gathering many proteins with different roles. Isolation and characterization of plasma membrane proteins by differential centrifugation and proteomic studies have revealed a remarkable diversity of proteins in these domains. The limited size of the lipid membrane nanodomain challenges the simple possibility that all of them can coexist within the same lipid membrane domain. As caveolin-1, flotillin isoforms and gangliosides are currently used as neuronal lipid membrane nanodomain markers, we first analyzed the structural features of these components forming nanodomains at the plasma membrane since they are relevant for building supramolecular complexes constituted by these molecular signatures. Among the proteins associated with neuronal lipid membrane nanodomains, there are a large number of proteins that play major roles in calcium signaling, such as ionotropic and metabotropic receptors for neurotransmitters, calcium channels, and calcium pumps. This review highlights a large variation between the calcium signaling proteins that have been reported to be associated with isolated caveolin-1 and flotillin-lipid membrane nanodomains. Since these calcium signaling proteins are scattered in different locations of the neuronal plasma membrane, i.e., in presynapses, postsynapses, axonal or dendritic trees, or in the neuronal soma, our analysis suggests that different lipid membrane-domain subtypes should exist in neurons. Furthermore, we conclude that classification of lipid membrane domains by their content in calcium signaling proteins sheds light on the roles of these domains for neuronal activities that are dependent upon the intracellular calcium concentration. Some examples described in this review include the synaptic and metabolic activity, secretion of neurotransmitters and neuromodulators, neuronal excitability (long-term potentiation and long-term depression), axonal and dendritic growth but also neuronal cell survival and death.
Collapse
Affiliation(s)
- Alejandro K. Samhan-Arias
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), C/Arturo Duperier 4, 28029 Madrid, Spain;
- Instituto de Investigaciones Biomédicas ‘Sols-Morreale’ (CSIC-UAM), C/Arturo Duperier 4, 28029 Madrid, Spain
| | - Joana Poejo
- Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain;
| | - Dorinda Marques-da-Silva
- LSRE—Laboratory of Separation and Reaction Engineering and LCM—Laboratory of Catalysis and Materials, School of Management and Technology, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal;
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| | - Oscar H. Martínez-Costa
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), C/Arturo Duperier 4, 28029 Madrid, Spain;
- Instituto de Investigaciones Biomédicas ‘Sols-Morreale’ (CSIC-UAM), C/Arturo Duperier 4, 28029 Madrid, Spain
| | - Carlos Gutierrez-Merino
- Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain;
| |
Collapse
|
15
|
Tan X, Zhang K, Shi W, Tang Z. Research progress on the regulation and mechanism of borneol on the blood-brain barrier in pathological states: a narrative review focused on ischemic stroke and cerebral glioma. Transl Cancer Res 2023; 12:3198-3209. [PMID: 38130309 PMCID: PMC10731338 DOI: 10.21037/tcr-23-1487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/12/2023] [Indexed: 12/23/2023]
Abstract
Background and Objective The blood-brain barrier (BBB) serves as a dynamic, selective shield, safeguarding the central nervous system (CNS) by separating the brain from circulating blood, preserving its microenvironment, and ensuring stability. However, in the presence of brain pathology, drug delivery across the BBB and blood-tumor barrier (BTB) becomes challenging, hindering effective treatments. Borneol exhibits promise in bidirectionally modulating the BBB under pathological conditions, suggesting at potential clinical applications for related diseases. Our primary goal in this review is to investigate borneol's potential clinical utility in bidirectionally regulating the BBB under pathological conditions. Methods The PubMed database, CNKI (China National Knowledge Infrastructure), Wanfang Data were searched to retrieve articles on animal experiments and cell-based research published from January 1, 2003, to May 1, 2023, using the following medical subject headings (MeSH) terms: borneol, blood-brain barrier, ischemic stroke, cerebral gliomas, anti-inflammatory. The search was limited to articles published in English and Chinese. In total, 86 articles were deemed eligible for inclusion in this study. Key Content and Findings The breakdown of the BBB is a key pathological process in ischemic stroke and cerebral glioma. When used alone, the lipophilic properties of borneol can reduce the permeability of the BBB and restore its normal function, thereby repairing brain damage and protecting brain tissue. Its specific protective effects may be related to inflammatory regulation mechanisms. The anti-inflammatory and protective effects of borneol can be used to improve and treat lesions caused by ischemic stroke and cerebral glioma. Furthermore, when combined with other drugs, borneol can accelerate the opening of the BBB, improve permeability through physiological processes, and enhance drug penetration and distribution in the brain without causing pathological damage to the brain. Conclusions This review summarizes the mechanisms by which borneol regulates the BBB and BTB in ischemic stroke and cerebral glioma, and discusses the potential clinical applications of borneol in the treatment of these diseases. It is believed that in the future, as research methods are refined, more effective and targeted therapies for cerebral glioma and ischemic stroke will be explored related to the protective mechanism of the BBB under pathological conditions with borneol alone or in combination with other drugs.
Collapse
Affiliation(s)
- Xinghua Tan
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing, China
| | - Ke Zhang
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing, China
| | - Wenyin Shi
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zhihua Tang
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
16
|
Cao D, Li B, Cao C, Zhang J, Li X, Li H, Yu Z, Shen H, Ye M. Caveolin-1 aggravates neurological deficits by activating neuroinflammation following experimental intracerebral hemorrhage in rats. Exp Neurol 2023; 368:114508. [PMID: 37598879 DOI: 10.1016/j.expneurol.2023.114508] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/30/2023] [Accepted: 08/16/2023] [Indexed: 08/22/2023]
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is one of the stroke subtypes with the highest mortality. Secondary brain injury is associated with neurological dysfunction and poor prognosis after ICH. Caveolin-1 (CAV1) is the key protein of Caveolae. Previous studies have shown that CAV1 plays an important role in central nervous system diseases, and pointed out that in a collagenase-induced ICH model in vivo, CAV1 is associated with neuroinflammatory activation and poor neurological prognosis. In this study, we explore the role and the molecular mechanism of CAV1 in brain injury via a rat autologous whole blood injection model and an in vitro model of ICH. METHODS Adult male Sprague-Dawley rats ICH model was induced through autologous whole blood injecting into the right basal ganglia. The changes in protein levels of CAV1 in brain tissues of ICH rats were detected by western blot analysis. The immunofluorescent staining was used to explore the changes of CAV1 in microglia/macrophages (Iba1+ cells). Lentivirus vectors were administered by intracerebroventricular injection to induce CAV1 overexpression and knockdown respectively. The western blot analysis, immunofluorescence staining, enzyme-linked immunosorbent assay, terminal deoxynucleotidyl transferase dUTP nick end labeling and Nissl staining were performed to explore the role of CAV1 in secondary brain injury after ICH. Meanwhile, the rotarod test, foot fault test, adhesive-removal test, and Modified Garcia Test, as well as Morris Water Maze test, were performed to evaluate the behavioral cognitive impairment of ICH rats after genetic intervention. Additionally, BV-2 cells treated with oxygen hemoglobin for 24 h, were used as an in vitro model of ICH in this study to explore the molecular mechanism of CAV1 in brain injury; we performed western blot analysis after precise regulation of CAV1 in BV2 cells to observe changes in protein levels and phosphorylated levels of C-Src, IKK-β, and NF-κB. RESULTS The expression of CAV1 in microglia/macrophages (Iba1+ cells) was elevated and reached the peak at 24 h after ICH. CAV1 knockdown ameliorated ICH-induced neurological deficits, while CAV1 overexpression significantly worsened neurological dysfunction of ICH rats. CAV1 knockdown attenuated cellular apoptosis and promoted neuronal survival in brain tissues of ICH rats, while the ICH rats with CAV1 overexpression presented more cellular apoptosis and neuronal loss. Meanwhile, CAV1 knockdown inhibited the microglia activation and neuroinflammatory response, while CAV1 overexpression abolished these effects and aggravated neuroinflammation in brain tissues of ICH rats. Additionally, by inducing to CAV1 knockdown in BV2 cells in an in vitro model of ICH, the levels of p-C-Src, CAV-1, p-CAV-1, and p-IKK-β in cytoplasm and the level of NF-κB p65 in nucleus of BV2 cells were significantly decreased, while they were increased by inducing to CAV1 overexpression. CONCLUSIONS Our research revealed CAV1 aggravated neurological dysfunction in a rat ICH model. CAV1 knockdown exerted neuroprotective effect by suppressing microglia activation and neuroinflammation after ICH might via the C-Src/CAV1/IKK-β/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Demao Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Department of neurosurgery, The Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Bing Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Department of Neurosurgery, Yancheng City No.1 People's Hospital, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng 224006, Jiangsu Province, China
| | - Cheng Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Department of Neurocritical Intensive Care Unit, Jiangyin Clinical College of Xuzhou Medical College, Jiangyin, Jiangsu Province, China
| | - Juyi Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Ming Ye
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| |
Collapse
|
17
|
Larochelle J, Tishko RJ, Yang C, Ge Y, Phan LT, Gunraj RE, Stansbury SM, Liu L, Mohamadzadeh M, Khoshbouei H, Candelario-Jalil E. Receptor-interacting protein kinase 2 (RIPK2) profoundly contributes to post-stroke neuroinflammation and behavioral deficits with microglia as unique perpetrators. J Neuroinflammation 2023; 20:221. [PMID: 37777791 PMCID: PMC10543871 DOI: 10.1186/s12974-023-02907-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023] Open
Abstract
BACKGROUND Receptor-interacting protein kinase 2 (RIPK2) is a serine/threonine kinase whose activity propagates inflammatory signaling through its association with pattern recognition receptors (PRRs) and subsequent TAK1, NF-κB, and MAPK pathway activation. After stroke, dead and dying cells release a host of damage-associated molecular patterns (DAMPs) that activate PRRs and initiate a robust inflammatory response. We hypothesize that RIPK2 plays a damaging role in the progression of stroke injury by enhancing the neuroinflammatory response to stroke and that global genetic deletion or microglia-specific conditional deletion of Ripk2 will be protective following ischemic stroke. METHODS Adult (3-6 months) male mice were subjected to 45 min of transient middle cerebral artery occlusion (tMCAO) followed by 24 h, 48 h, or 28 days of reperfusion. Aged male and female mice (18-24 months) were subjected to permanent ischemic stroke and sacrificed 48 h later. Infarct volumes were calculated using TTC staining (24-48 h) or Cresyl violet staining (28d). Sensorimotor tests (weight grip, vertical grid, and open field) were performed at indicated timepoints. Blood-brain barrier (BBB) damage, tight junction proteins, matrix metalloproteinase-9 (MMP-9), and neuroinflammatory markers were assessed via immunoblotting, ELISA, immunohistochemistry, and RT-qPCR. Differential gene expression profiles were generated through bulk RNA sequencing and nanoString®. RESULTS Global genetic deletion of Ripk2 resulted in decreased infarct sizes and reduced neuroinflammatory markers 24 h after stroke compared to wild-type controls. Ripk2 global deletion also improved both acute and long-term behavioral outcomes with powerful effects on reducing infarct volume and mortality at 28d post-stroke. Conditional deletion of microglial Ripk2 (mKO) partially recapitulated our results in global Ripk2 deficient mice, showing reductive effects on infarct volume and improved behavioral outcomes within 48 h of injury. Finally, bulk transcriptomic profiling and nanoString data demonstrated that Ripk2 deficiency in microglia decreases genes associated with MAPK and NF-κB signaling, dampening the neuroinflammatory response after stroke injury by reducing immune cell activation and peripheral immune cell invasion. CONCLUSIONS These results reveal a hitherto unknown role for RIPK2 in the pathogenesis of ischemic stroke injury, with microglia playing a distinct role. This study identifies RIPK2 as a potent propagator of neuroinflammatory signaling, highlighting its potential as a therapeutic target for post-stroke intervention.
Collapse
Affiliation(s)
- Jonathan Larochelle
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Ryland J Tishko
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Yong Ge
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health, San Antonio, TX, USA
| | - Leah T Phan
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Rachel E Gunraj
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Sofia M Stansbury
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Lei Liu
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Mansour Mohamadzadeh
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health, San Antonio, TX, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, 1149 SW Newell Drive, Gainesville, FL, 32610, USA.
| |
Collapse
|
18
|
Ji Z, Zheng J, Ma Y, Lei H, Lin W, Huang J, Yang H, Zhang G, Li B, Shu B, Du X, Zhang J, Lin H, Liao Y. Emergency Treatment and Photoacoustic Assessment of Spinal Cord Injury Using Reversible Dual-Signal Transform-Based Selenium Antioxidant. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207888. [PMID: 37127878 DOI: 10.1002/smll.202207888] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 04/08/2023] [Indexed: 05/03/2023]
Abstract
Spinal cord injury (SCI), following explosive oxidative stress, causes an abrupt and irreversible pathological deterioration of the central nervous system. Thus, preventing secondary injuries caused by reactive oxygen species (ROS), as well as monitoring and assessing the recovery from SCI are critical for the emergency treatment of SCI. Herein, an emergency treatment strategy is developed for SCI based on the selenium (Se) matrix antioxidant system to effectively inhibit oxidative stress-induced damage and simultaneously real-time evaluate the severity of SCI using a reversible dual-photoacoustic signal (680 and 750 nm). Within the emergency treatment and photoacoustic severity assessment (ETPSA) strategy, the designed Se loaded boron dipyrromethene dye with a double hydroxyl group (Se@BDP-DOH) is simultaneously used as a sensitive reporter group and an excellent antioxidant for effectively eliminating explosive oxidative stress. Se@BDP-DOH is found to promote the recovery of both spinal cord tissue and locomotor function in mice with SCI. Furthermore, ETPSA strategy synergistically enhanced ROS consumption via the caveolin 1 (Cav 1)-related pathways, as confirmed upon treatment with Cav 1 siRNA. Therefore, the ETPSA strategy is a potential tool for improving emergency treatment and photoacoustic assessment of SCI.
Collapse
Affiliation(s)
- Zhisheng Ji
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, P. R. China
| | - Judun Zheng
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University, Guangzhou, 510091, P. R. China
| | - Yanming Ma
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, P. R. China
| | - Hongyi Lei
- Department of Anesthesiology, Longgang District Central Hospital of Shenzhen, Shenzhen, 518100, P. R. China
| | - Weiqiang Lin
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University, Guangzhou, 510091, P. R. China
| | - Jialin Huang
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University, Guangzhou, 510091, P. R. China
| | - Hua Yang
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, P. R. China
| | - Guowei Zhang
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, P. R. China
| | - Bin Li
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University, Guangzhou, 510091, P. R. China
| | - Bowen Shu
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University, Guangzhou, 510091, P. R. China
| | - Xianjin Du
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, P. R. China
| | - Jian Zhang
- Department of Biomedical Engineering, School of Basic Medical Science, Guang-zhou Medical University, Guangzhou, 511436, P. R. China
| | - Hongsheng Lin
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, P. R. China
| | - Yuhui Liao
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital, Southern Medical University, Guangzhou, 510091, P. R. China
- Department of Anesthesiology, Longgang District Central Hospital of Shenzhen, Shenzhen, 518100, P. R. China
- NHC Key Laboratory of Metabolic Cardiovascular Diseases Research, Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, 750004, P. R. China
| |
Collapse
|
19
|
Jezierski A, Huang J, Haqqani AS, Haukenfrers J, Liu Z, Baumann E, Sodja C, Charlebois C, Delaney CE, Star AT, Liu Q, Stanimirovic DB. Mouse embryonic stem cell-derived blood-brain barrier model: applicability to studying antibody triggered receptor mediated transcytosis. Fluids Barriers CNS 2023; 20:36. [PMID: 37237379 DOI: 10.1186/s12987-023-00437-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
Blood brain barrier (BBB) models in vitro are an important tool to aid in the pre-clinical evaluation and selection of BBB-crossing therapeutics. Stem cell derived BBB models have recently demonstrated a substantial advantage over primary and immortalized brain endothelial cells (BECs) for BBB modeling. Coupled with recent discoveries highlighting significant species differences in the expression and function of key BBB transporters, the field is in need of robust, species-specific BBB models for improved translational predictability. We have developed a mouse BBB model, composed of mouse embryonic stem cell (mESC-D3)-derived brain endothelial-like cells (mBECs), employing a directed monolayer differentiation strategy. Although the mBECs showed a mixed endothelial-epithelial phenotype, they exhibited high transendothelial electrical resistance, inducible by retinoic acid treatment up to 400 Ω cm2. This tight cell barrier resulted in restricted sodium fluorescein permeability (1.7 × 10-5 cm/min), significantly lower than that of bEnd.3 cells (1.02 × 10-3 cm/min) and comparable to human induced pluripotent stem cell (iPSC)-derived BECs (2.0 × 10-5 cm/min). The mBECs expressed tight junction proteins, polarized and functional P-gp efflux transporter and receptor mediated transcytosis (RMT) receptors; collectively important criteria for studying barrier regulation and drug delivery applications in the CNS. In this study, we compared transport of a panel of antibodies binding species selective or cross-reactive epitopes on BBB RMT receptors in both the mBEC and human iPSC-derived BEC model, to demonstrate discrimination of species-specific BBB transport mechanisms.
Collapse
Affiliation(s)
- Anna Jezierski
- Human Health Therapeutics Research Centre, National Research Council of Canada, ON, Ottawa, Canada.
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| | - Jez Huang
- Human Health Therapeutics Research Centre, National Research Council of Canada, ON, Ottawa, Canada
| | - Arsalan S Haqqani
- Human Health Therapeutics Research Centre, National Research Council of Canada, ON, Ottawa, Canada
| | - Julie Haukenfrers
- Human Health Therapeutics Research Centre, National Research Council of Canada, ON, Ottawa, Canada
| | - Ziying Liu
- Human Health Therapeutics Research Centre, National Research Council of Canada, ON, Ottawa, Canada
| | - Ewa Baumann
- Human Health Therapeutics Research Centre, National Research Council of Canada, ON, Ottawa, Canada
| | - Caroline Sodja
- Human Health Therapeutics Research Centre, National Research Council of Canada, ON, Ottawa, Canada
| | - Claudie Charlebois
- Human Health Therapeutics Research Centre, National Research Council of Canada, ON, Ottawa, Canada
| | - Christie E Delaney
- Human Health Therapeutics Research Centre, National Research Council of Canada, ON, Ottawa, Canada
| | - Alexandra T Star
- Human Health Therapeutics Research Centre, National Research Council of Canada, ON, Ottawa, Canada
| | - Qing Liu
- Human Health Therapeutics Research Centre, National Research Council of Canada, ON, Ottawa, Canada
| | - Danica B Stanimirovic
- Human Health Therapeutics Research Centre, National Research Council of Canada, ON, Ottawa, Canada
| |
Collapse
|
20
|
Xu Y, Chen B, Yi J, Tian F, Liu Y, Ouyang Y, Yuan C, Liu B. Buyang Huanwu Decoction alleviates cerebral ischemic injury through modulating caveolin-1-mediated mitochondrial quality control. Front Pharmacol 2023; 14:1137609. [PMID: 37234709 PMCID: PMC10206009 DOI: 10.3389/fphar.2023.1137609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/10/2023] [Indexed: 05/28/2023] Open
Abstract
Introduction: Mitochondrial quality control (MQC) is an important mechanism of neural repair after cerebral ischemia (CI). Recent studies have shown that caveolin-1 (Cav-1) is an important signaling molecule in the process of CI injury, but its mechanism of regulating MQC after CI is still unclear. Buyang Huanwu Decoction (BHD) is a classic traditional Chinese medicine formula that is often used to treat CI. Unfortunately, its mechanism of action is still obscure. Methods: In this study, we tested the hypothesis that BHD can regulate MQC through Cav-1 and exert an anti-cerebral ischemia injury effect. We used Cav-1 knockout mice and their homologous wild-type mice, replicated middle cerebral artery occlusion (MCAO) model and BHD intervention. Neurobehavioral scores and pathological detection were used to evaluate neurological function and neuron damage, transmission electron microscopy and enzymology detection of mitochondrial damage. Finally, western blot and RT-qPCR expression of MQC-related molecules were tested. Results: After CI, mice showed neurologic impairment, neuronal damage, and significant destruction of mitochondrial morphology and function, and MQC was imbalanced. Cav-1 deletion aggravated the damage to neurological function, neurons, mitochondrial morphology and mitochondrial function after CI, aggravated the imbalance of mitochondrial dynamics, and inhibited mitophagy and biosynthesis. BHD can maintain MQC homeostasis after CI through Cav-1 and improve CI injury. Discussion: Cav-1 can affect CI injury by regulating MQC, and this mechanism may be another target of BHD for anti-cerebral ischemia injury.
Collapse
Affiliation(s)
- Yaqian Xu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
- MOE Key Laboratory of Research and Translation on Prevention and Treatment of Major Diseases in Internal Medicine of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Bowei Chen
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
- MOE Key Laboratory of Research and Translation on Prevention and Treatment of Major Diseases in Internal Medicine of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Jian Yi
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
- MOE Key Laboratory of Research and Translation on Prevention and Treatment of Major Diseases in Internal Medicine of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Fengming Tian
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
- MOE Key Laboratory of Research and Translation on Prevention and Treatment of Major Diseases in Internal Medicine of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yingfei Liu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
- MOE Key Laboratory of Research and Translation on Prevention and Treatment of Major Diseases in Internal Medicine of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yin Ouyang
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
- MOE Key Laboratory of Research and Translation on Prevention and Treatment of Major Diseases in Internal Medicine of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Chunyun Yuan
- Hunan Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, China
- Affiliated Hospital of Hunan Academy of Chinese Medicine, Changsha, China
| | - Baiyan Liu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
- MOE Key Laboratory of Research and Translation on Prevention and Treatment of Major Diseases in Internal Medicine of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Academy of Chinese Medicine, Changsha, China
| |
Collapse
|
21
|
Li Y, Liu B, Zhao T, Quan X, Han Y, Cheng Y, Chen Y, Shen X, Zheng Y, Zhao Y. Comparative study of extracellular vesicles derived from mesenchymal stem cells and brain endothelial cells attenuating blood-brain barrier permeability via regulating Caveolin-1-dependent ZO-1 and Claudin-5 endocytosis in acute ischemic stroke. J Nanobiotechnology 2023; 21:70. [PMID: 36855156 PMCID: PMC9976550 DOI: 10.1186/s12951-023-01828-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) disruption is a major adverse event after ischemic stroke (IS). Caveolin-1 (Cav-1), a scaffolding protein, played multiple roles in BBB permeability after IS, while the pros and cons of Cav-1 on BBB permeability remain controversial. Numerous studies revealed that extracellular vesicles (EVs), especially stem cells derived EVs, exerted therapeutic efficacy on IS; however, the mechanisms of BBB permeability needed to be clearly illustrated. Herein, we compared the protective efficacy on BBB integrity between bone marrow mesenchymal stem cells derived extracellular vesicles (BMSC-EVs) and EVs from brain endothelial cells (BEC-EVs) after acute IS and investigated whether the mechanism was associated with EVs antagonizing Cav-1-dependent tight junction proteins endocytosis. METHODS BMSC-EVs and BEC-EVs were isolated and characterized by nanoparticle tracking analysis, western blotting, and transmission electron microscope. Oxygen and glucose deprivation (OGD) treated b. End3 cells were utilized to evaluate brain endothelial cell leakage. CCK-8 and TRITC-dextran leakage assays were used to measure cell viability and transwell monolayer permeability. Permanent middle cerebral artery occlusion (pMCAo) model was established, and EVs were intravenously administered in rats. Animal neurological function tests were applied, and microvessels were isolated from the ischemic cortex. BBB leakage and tight junction proteins were analyzed by Evans Blue (EB) staining and western blotting, respectively. Co-IP assay and Cav-1 siRNA/pcDNA 3.1 vector transfection were employed to verify the endocytosis efficacy of Cav-1 on tight junction proteins. RESULTS Both kinds of EVs exerted similar efficacies in reducing the cerebral infarction volume and BBB leakage and enhancing the expressions of ZO-1 and Claudin-5 after 24 h pMCAo in rats. At the same time, BMSC-EVs were outstanding in ameliorating neurological function. Simultaneously, both EVs treatments suppressed the highly expressed Cav-1 in OGD-exposed b. End3 cells and ischemic cerebral microvessels, and this efficacy was more prominent after BMSC-EVs administration. Cav-1 knockdown reduced OGD-treated b. End3 cells monolayer permeability and recovered ZO-1 and Claudin-5 expressions, whereas Cav-1 overexpression aggravated permeability and enhanced the colocalization of Cav-1 with ZO-1 and Claudin-5. Furthermore, Cav-1 overexpression partly reversed the lower cell leakage by BMSC-EVs and BEC-EVs administrations in OGD-treated b. End3 cells. CONCLUSIONS Our results demonstrated that Cav-1 aggravated BBB permeability in acute ischemic stroke, and BMSC-EVs exerted similar antagonistic efficacy to BEC-EVs on Cav-1-dependent ZO-1 and Claudin-5 endocytosis. BMSC-EVs treatment was superior in Cav-1 suppression and neurological function amelioration.
Collapse
Affiliation(s)
- Yiyang Li
- grid.437123.00000 0004 1794 8068Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR China
| | - Bowen Liu
- grid.268505.c0000 0000 8744 8924Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Tingting Zhao
- grid.259384.10000 0000 8945 4455Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR China
| | - Xingping Quan
- grid.437123.00000 0004 1794 8068Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR China
| | - Yan Han
- grid.437123.00000 0004 1794 8068Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR China
| | - Yaxin Cheng
- grid.437123.00000 0004 1794 8068Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR China
| | - Yanling Chen
- grid.417409.f0000 0001 0240 6969Department of Pathophysiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong China
| | - Xu Shen
- grid.410745.30000 0004 1765 1045Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica and State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Zheng
- grid.437123.00000 0004 1794 8068Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR China ,grid.437123.00000 0004 1794 8068Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR China
| | - Yonghua Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, China. .,Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, China.
| |
Collapse
|
22
|
Yang M, Tang L, Hu Z, Tang X. Application of Neuroimaging for the Prediction of Hemorrhagic Transformation after Intravenous Thrombolysis in Acute Ischemic Stroke. Cerebrovasc Dis 2023; 52:1-10. [PMID: 35661647 DOI: 10.1159/000524749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/12/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Ischemic stroke is a common cerebrovascular disease with high morbidity, disability, and mortality worldwide. Currently, recombinant tissue plasminogen activator is the main intravenous thrombolysis agent for the treatment of acute ischemic stroke within 4.5 h after onset. Hemorrhagic transformation (HT) is the most serious complication of intravenous thrombolysis, which can significantly aggravate clinical poor prognosis. Therefore, it is important to early predict the risk of post-thrombolysis HT in patients with acute ischemic stroke. SUMMARY Recently, several studies have reported that neuroimaging techniques have potential value in predicting HT after intravenous thrombolysis in patients with acute ischemic stroke. The corresponding neuroimaging parameters may be effective predictors of HT after intravenous thrombolysis. In this review, we summarized and discussed the application of neuroimaging techniques and related parameters in predicting HT after intravenous thrombolysis. KEY MESSAGES Recognizing and understanding the predictive performance of neuroimaging parameters for HT may help assess the risk of HT after intravenous thrombolysis in patients with acute ischemic stroke and make an appropriate treatment decision.
Collapse
Affiliation(s)
- Miaomiao Yang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China,
| | - Lisha Tang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiangqi Tang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
23
|
Zhang L, Wang C, Zhao M, Li X, Qu H, Xu J, Li D. Prognostic Values Serum Cav-1 and NGB Levels in Early Neurological Deterioration After Intravenous Thrombolysis in Patients with Acute Ischemic Stroke. Clin Appl Thromb Hemost 2023; 29:10760296231219707. [PMID: 38092682 PMCID: PMC10722930 DOI: 10.1177/10760296231219707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/15/2023] [Accepted: 11/24/2023] [Indexed: 12/17/2023] Open
Abstract
Early neurological deterioration after intravenous thrombolysis (IAT) leads to increased mortality and morbidity in patients with acute ischemic stroke (AIS). This study investigated the correlation between serum Cav-1 and NGB levels and END after IAT and explored their predictive values for poor prognosis of AIS. Totally 210 patients with AIS who underwent IAT within 4.5 h of onset were included and assigned into END group (n = 90) and Non-END group (n = 120). ELISA was used to detect serum Cav-1 and NGB levels before IAT in AIS patients. The prognosis of END patients after 3 months of treatment was evaluated using the modified Rankin Scale. Logistic multifactorial regression was used to analyze independent risk factors for END and poor prognosis after IAT. ROC curve was used to analyze the predictive effect of Cav-1 and NGB on END and poor prognosis after IAT. The area under the ROC curve was analyzed by MedCalc comparison. Compared with the Non-END group, serum Cav-1 was lower and NGB was higher in the END group. Cav-1 and NGB were independent risk factors for END after IAT. Cav-1 + NGB better predicted END after IAT than Cav-1 or NGB alone. Cav-1 and NGB were independent risk factors for END poor prognosis after IAT. Cav-1 combined with NGB better predicted poor prognosis of END after IAT than Cav-1 or NGB alone. Serum Cav-1 combined with NGB may assist in predicting the risk of END occurrence and poor prognosis after IAT in patients with AIS.
Collapse
Affiliation(s)
- Lihong Zhang
- Department of Neurointervention and Neurological Intensive Care, Dalian Central Hospital Affiliated to Dalian University of Technology, Dalian City, Liaoning Province, China
| | - Cui Wang
- Department of Neurology, Dalian Central Hospital Affiliated to Dalian University of Technology, Dalian City, Liaoning Province, China
| | - Manhong Zhao
- Department of Neurointervention and Neurological Intensive Care, Dalian Central Hospital Affiliated to Dalian University of Technology, Dalian City, Liaoning Province, China
| | - Xuesong Li
- Department of Radiology, Dalian Central Hospital Affiliated to Dalian University of Technology, Dalian City, Liaoning Province, China
| | - Hong Qu
- Bidding and Procurement Office, The Second Affiliated Hospital of Dalian Medical University, City, Liaoning Province, China
| | - Jianping Xu
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, China
| | - Di Li
- Department of Neurointervention and Neurological Intensive Care, Dalian Central Hospital Affiliated to Dalian University of Technology, Dalian City, Liaoning Province, China
| |
Collapse
|
24
|
Jiang X, Li Y, Fu D, You T, Wu S, Xin J, Wen J, Huang Y, Hu C. Caveolin-1 ameliorates acetaminophen-aggravated inflammatory damage and lipid deposition in non-alcoholic fatty liver disease via the ROS/TXNIP/NLRP3 pathway. Int Immunopharmacol 2023; 114:109558. [PMID: 36700765 DOI: 10.1016/j.intimp.2022.109558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/24/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
The overuse of acetaminophen (APAP) may cause more severe hepatotoxicity in patients with non-alcoholic fatty liver disease (NAFLD). Caveolin-1 (CAV1), is an essential regulator of metabolic function, which can alleviate liver damage by scavenging reactive oxygen species (ROS). Evidence suggests that the NOD-like receptor family pyrin domain-containing 3 (NLRP3) -mediated pyroptosis is involved in the development of NAFLD. Moreover, thioredoxin-interactive protein (TXNIP) activation is a key event linking ROS to NLRP3 inflammasome. However, whether CAV1 alleviates APAP-aggravated hepatotoxicity in NAFLD via the ROS/TXNIP/NLRP3 pathway remains unclear. An in vivo fatty liver model was established by feeding mice a high-fat diet for 56 days. Additionally, using in vitro approach, AML-12 cells were incubated with free fatty acids for 48 h and APAP was added during the last 24 h. We found that the overuse of APAP in NAFLD not only induced oxidative stress, but also increased TXNIP expression, NLRP3-mediated pyroptosis, and lipid deposition. In addition to inhibiting ROS generation and lipid deposition, overexpression of CAV1 reduced the elevated levels of TXNIP expression and NLRP3-mediated pyroptosis. However, the effect of CAV1 on TXNIP expression, NLRP3-mediated pyroptosis, and lipid deposition was reversed by CAV1 small interfering RNA (siRNA) intervention. Finally, N-acetyl cysteine (NAC) treatment reduced CAV1 siRNA-mediated changes in TXNIP expression and NLRP3-mediated pyroptosis levels. These results demonstrate that the inhibitory effect of CAV1 on NLRP3-mediated pyroptosis may be mediated through the ROS/TXNIP axis. Moreover, the current study provides novel mechanistic insights into the protective effects of CAV1 on APAP-aggravated hepatotoxicity in NAFLD.
Collapse
Affiliation(s)
- Xiangfu Jiang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Yu Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Dongdong Fu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Tingyu You
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Shuai Wu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Jiao Xin
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Jiagen Wen
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Yan Huang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Chengmu Hu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei 230032, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
25
|
Yu L, Jin Z, Li M, Liu H, Tao J, Xu C, Wang L, Zhang Q. Protective potential of hydroxysafflor yellow A in cerebral ischemia and reperfusion injury: An overview of evidence from experimental studies. Front Pharmacol 2022; 13:1063035. [PMID: 36588739 PMCID: PMC9797593 DOI: 10.3389/fphar.2022.1063035] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Ischemic stroke, mostly caused by thromboembolic or thrombotic arterial occlusions, is a primary leading cause of death worldwide with high morbidity and disability. Unfortunately, no specific medicine is available for the treatment of cerebral I/R injury due to its limitation of therapeutic window. Hydroxysafflor yellow A, a natural product extracted from Carthamus tinctorius, has been extensively investigated on its pharmacological properties in cerebrovascular diseases. However, review focusing on the beneficial role of HSYA against cerebral I/R injury is still lacking. In this paper, we reviewed the neuroprotective effect of HSYA in preclinical studies and the underlying mechanisms involved, as well as clinical data that support the pharmacological activities. Additionally, the sources, physicochemical properties, biosynthesis, safety and limitations of HSYA were also reviewed. As a result, HSYA possesses a wide range of beneficial effects against cerebral I/R injury, and its action mechanisms include anti-excitotoxicity, anti-oxidant stress, anti-apoptosis, anti-inflammation, attenuating BBB leakage and regulating autophagy. Collectively, HSYA might be applied as one of the promising alternatives in ischemic stroke treatment.
Collapse
Affiliation(s)
- Lu Yu
- Comprehensive Department of Traditional Chinese Medicine, First Department of Integration, Department of Neurology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Lu Yu, ; Qiujuan Zhang, ; Liwei Wang,
| | - Zhe Jin
- Department of Neurology, Renji Hospital Baoshan Branch, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mincheng Li
- Comprehensive Department of Traditional Chinese Medicine, First Department of Integration, Department of Neurology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huifang Liu
- Department of Neurology, Shanghai Jinshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Jie Tao
- Comprehensive Department of Traditional Chinese Medicine, First Department of Integration, Department of Neurology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chuan Xu
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liwei Wang
- Comprehensive Department of Traditional Chinese Medicine, First Department of Integration, Department of Neurology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Lu Yu, ; Qiujuan Zhang, ; Liwei Wang,
| | - Qiujuan Zhang
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Lu Yu, ; Qiujuan Zhang, ; Liwei Wang,
| |
Collapse
|
26
|
Chen H, She Q, Liu Y, Chen J, Qin Y, Lu C. The peripheral Epac1/p-Cav-1 pathway underlies the disruption of the vascular endothelial barrier following skin/muscle incision and retraction-induced chronic postsurgical pain. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1377. [PMID: 36660643 PMCID: PMC9843368 DOI: 10.21037/atm-22-6069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022]
Abstract
Background Vascular endothelial barrier disruption is pivotal in the development of acute and chronic pain. Here, we demonstrate a previously unidentified molecular mechanism in which activation of the peripheral Epac1/p-Cav-1 pathway accelerated the disruption of the vascular endothelial barrier, thereby promoting chronic postsurgical pain (CPSP). Methods We established a rat model of CPSP induced by skin/muscle incision and retraction (SMIR). Pain behaviors were assessed by the mechanical withdrawal threshold (MWT) at different times. Local muscle tissues around the incision were isolated to detect the vascular permeability and the expression of Epac1 and Cav-1. They were assessed by western blot and immunofluorescence staining. Results SMIR increased vascular endothelial permeability and the number of macrophages and endothelial cells in the muscle tissues around the incision. The peripheral upregulation of Epac1 was macrophage-derived, whereas that of p-Cav-1 was both macrophage and endothelial cell-derived in the SMIR model. Moreover, the Epac1 agonist 8-pCPT could induce mechanical sensitivity, increase the expression of p-Cav-1, and disrupt vascular endothelial barrier in normal rats. The Epac1 inhibitor CE3F4 attenuated established SMIR-induced mechanical hyperalgesia, the upregulation of p-Cav-1 and vascular endothelial barrier. Finally, we showed that intrathecal injection of Cav-1siRNA relieved SMIR-induced mechanical allodynia, but had no effects of the expression of Epac1. Conclusions Collectively, these results revealed a molecular mechanism for modulating CPSP through the peripheral Epac1/Cav-1 pathway. Importantly, targeting Epac1/Cav-1 signaling might be a potential treatment for CPSP.
Collapse
Affiliation(s)
- Hongsheng Chen
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Qing She
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yanfang Liu
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Junjie Chen
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yibin Qin
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Cui'e Lu
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
27
|
Choudhary N, Tewari D, Nabavi SF, Kashani HRK, Lorigooini Z, Filosa R, Khan FB, Masoudian N, Nabavi SM. Plant based food bioactives: A boon or bane for neurological disorders. Crit Rev Food Sci Nutr 2022; 64:3279-3325. [PMID: 36369694 DOI: 10.1080/10408398.2022.2131729] [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] [Indexed: 11/14/2022]
Abstract
Neurological disorders are the foremost occurring diseases across the globe resulting in progressive dysfunction, loss of neuronal structure ultimately cell death. Therefore, attention has been drawn toward the natural resources for the search of neuroprotective agents. Plant-based food bioactives have emerged as potential neuroprotective agents for the treatment of neurodegenerative disorders. This comprehensive review primarily focuses on various plant food bioactive, mechanisms, therapeutic targets, in vitro and in vivo studies in the treatment of neurological disorders to explore whether they are boon or bane for neurological disorders. In addition, the clinical perspective of plant food bioactives in neurological disorders are also highlighted. Scientific evidences point toward the enormous therapeutic efficacy of plant food bioactives in the prevention or treatment of neurological disorders. Nevertheless, identification of food bioactive components accountable for the neuroprotective effects, mechanism, clinical trials, and consolidation of information flow are warranted. Plant food bioactives primarily act by mediating through various pathways including oxidative stress, neuroinflammation, apoptosis, excitotoxicity, specific proteins, mitochondrial dysfunction, and reversing neurodegeneration and can be used for the prevention and therapy of neurodegenerative disorders. In conclusion, the plant based food bioactives are boon for neurological disorders.
Collapse
Affiliation(s)
- Neeraj Choudhary
- Department of Pharmacognosy, Adesh Institute of Pharmacy and Biomedical Sciences, Adesh University, Bathinda, Punjab, India
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Seyed Fazel Nabavi
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
- Nutringredientes Research Center, Federal Institute of Education, Science and Technology (IFCE), Baturite, Ceara, Brazil
| | - Hamid Reza Khayat Kashani
- Department of Neurosurgery, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Lorigooini
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Rosanna Filosa
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
- Department of Science and Technology, University of Sannio, 82100, Benevento, Italy
| | - Farheen Badrealam Khan
- Department of Biology, College of Science, The United Arab Emirates University, Al Ain, 15551 United Arab Emirates
| | - Nooshin Masoudian
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
| | - Seyed Mohammad Nabavi
- Advanced Medical Pharma (AMP-Biotec), Biopharmaceutical Innovation Centre Via Cortenocera, 82030, San Salvatore Telesino, (BN), Italy
- Nutringredientes Research Center, Federal Institute of Education, Science and Technology (IFCE), Baturite, Ceara, Brazil
| |
Collapse
|
28
|
Luo J, Chen D, Qin B, Kong D. Molecular mechanisms for the prevention and promoting the recovery from ischemic stroke by nutraceutical laminarin: A comparative transcriptomic approach. Front Nutr 2022; 9:999426. [PMID: 36118760 PMCID: PMC9479852 DOI: 10.3389/fnut.2022.999426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/08/2022] [Indexed: 11/23/2022] Open
Abstract
Stroke is the second leading cause of death and a major cause of disability worldwide. Ischemic stroke caused by atherosclerosis accounts for approximately 87% of all stroke cases. Ischemic stroke is a preventable disease; therefore, a better understanding of the molecular mechanisms underlying its pathogenesis and recovery processes could provide therapeutic targets for drug development and reduce the associated mortality rate. Laminarin, a polysaccharide, is a nutraceutical that can be found in brown algae. Accumulating evidence suggests that laminarin could reduce the detrimental effects of neuroinflammation on brain damage after stroke. However, the molecular mechanism underlying its beneficial effects remains largely unknown. In the present study, we used a middle cerebral artery occlusion (MCAO) rat model and applied comparative transcriptomics to investigate the molecular targets and pathways involved in the beneficial effects of laminarin on ischemic stroke. Our results show the involvement of laminarin targets in biological processes related to blood circulation, oxygen supply, and anti-inflammatory responses in the normal brain. More importantly, laminarin treatment attenuated brain damage and neurodeficits caused by ischemic stroke. These beneficial effects are controlled by biological processes related to blood vessel development and brain cell death through the regulation of canonical pathways. Our study, for the first time, delineated the molecular mechanisms underlying the beneficial effects of laminarin on ischemic stroke prevention and recovery and provides novel therapeutic targets for drug development against ischemic stroke.
Collapse
|
29
|
New Therapeutic Approaches to and Mechanisms of Ginsenoside Rg1 against Neurological Diseases. Cells 2022; 11:cells11162529. [PMID: 36010610 PMCID: PMC9406801 DOI: 10.3390/cells11162529] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
Neurological diseases, including Parkinson’s disease (PD), Alzheimer’s disease (AD), Huntington’s disease (HD), stroke, cerebral infarction, ischemia-reperfusion injury, depression and, stress, have high incidence and morbidity and often lead to disability. However, there is no particularly effective medication against them. Therefore, finding drugs with a suitable efficacy, low toxicity and manageable effects to improve the quality of life of patients is an urgent problem. Ginsenoside Rg1 (Rg1) is the main active component of ginseng and has a variety of pharmacological effects. In this review, we focused on the therapeutic potential of Rg1 for improving neurological diseases. We introduce the mechanisms of Ginsenoside Rg1 in neurological diseases, including apoptosis, neuroinflammation, the microRNA (miRNA) family, the mitogen-activated protein kinase (MAPK) family, oxidative stress, nuclear factor-κB (NF-κB), and learning and memory of Rg1 in neurological diseases. In addition, Rg1 can also improve neurological diseases through the interaction of different signal pathways. The purpose of this review is to explore more in-depth ideas for the clinical treatment of neurological diseases (including PD, AD, HD, stroke, cerebral infarction, ischemia–reperfusion injury, depression, and stress). Therefore, Rg1 is expected to become a new therapeutic method for the clinical treatment of neurological diseases.
Collapse
|
30
|
Popov LD. Deciphering the relationship between caveolae-mediated intracellular transport and signalling events. Cell Signal 2022; 97:110399. [PMID: 35820545 DOI: 10.1016/j.cellsig.2022.110399] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 11/30/2022]
Abstract
The caveolae-mediated transport across polarized epithelial cell barriers has been largely deciphered in the last decades and is considered the second essential intracellular transfer mechanism, after the clathrin-dependent endocytosis. The basic cell biology knowledge was supplemented recently, with the molecular mechanisms beyond caveolae generation implying the key contribution of the lipid-binding proteins (the structural protein Caveolin and the adapter protein Cavin), along with the bulb coat stabilizing molecules PACSIN-2 and Eps15 homology domain protein-2. The current attention is focused also on caveolae architecture (such as the bulb coat, the neck, the membrane funnel inside the bulb, and the associated receptors), and their specific tasks during the intracellular transport of various cargoes. Here, we resume the present understanding of the assembly, detachment, and internalization of caveolae from the plasma membrane lipid raft domains, and give an updated view on transcytosis and endocytosis, the two itineraries of cargoes transport via caveolae. The review adds novel data on the signalling molecules regulating caveolae intracellular routes and on the transport dysregulation in diseases. The therapeutic possibilities offered by exploitation of Caveolin-1 expression and caveolae trafficking, and the urgent issues to be uncovered conclude the review.
Collapse
Affiliation(s)
- Lucia-Doina Popov
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania.
| |
Collapse
|
31
|
Zhou M, Li D, Shen Q, Gao L, Zhuang P, Zhang Y, Guo H. Storax Inhibits Caveolae-Mediated Transcytosis at Blood-Brain Barrier After Ischemic Stroke in Rats. Front Pharmacol 2022; 13:876235. [PMID: 35873558 PMCID: PMC9304983 DOI: 10.3389/fphar.2022.876235] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Background and Purpose: Blood-brain barrier (BBB) disruption following ischemic stroke (IS) contributes to hemorrhagic transformation, brain edema, increased neural dysfunction, secondary injury, and mortality. The prevailing view attributes the destruction of tight junction proteins (TJs) to the resulting BBB damage following IS. However, recent studies define a stepwise impairment of the transcellular barrier followed by the paracellular barrier which accounts for the BBB leakage in IS. The increased endothelial transcytosis that has been proven to be caveolae-mediated, preceding and independent of TJs disintegration. Emerging experimental investigations suggested Storax attenuates BBB damage after stroke. This study aimed to test our hypothesis that Storax inhibits caveolae-mediated transcytosis at BBB after ischemic stroke in rats. Methods: Male Wistar rats (250-300 g) were subjected to transient middle cerebral artery occlusion (t-MCAO). Brain water content and the cerebral infarction size were assessed by brain tissue drying-wet method and 2,3,5-triphenyltetrazolium chloride (TTC) staining. BBB permeability was detected by the leakage of Evans blue and Albumin-Alexa594. The ultrastructure of BBB was examined by transmission electron microscopy (TEM). Cav-1 and Mfsd2a were quantified by western blotting and immunofluorescence staining, AQP4, PDGFR-β, ZO-1 and Occludin were quantified by western blotting. Results: Storax treatment of 0.1 g/kg had no significant effects on brain lesions. Storax treatment of 0.2, 0.4, and 0.8 g/kg led to a significant decrease in infarction size, and the Storax 0.4, 0.8 g/kg groups displayed a significant reduction in brain water content. Storax treatment of 0.8 g/kg showed mild toxic reactions. Thus, 0.4 g/kg Storax was selected as the optimal dose for subsequent studies. Storax significantly inhibited the fluorescent albumin intensity in the brain parenchyma and the number of caveolae in ECs, alongside attenuating the ultrastructural disruption of BBB at 6 h after stroke. Meanwhile, Storax significantly increased the expression of Mfsd2a and PDGFR-β, and decrease the expression of Cav-1 and AQP4, corresponding to the significantly decreased Cav-1 positive cells and increased Mfsd2a positive cells. However, Storax has no significant effects on Evan blue leakage or the expression ZO-1, Occludin. Conclusion: Our experimental findings demonstrate Storax treatment inhibits caveolae-mediated transcytosis at BBB in the focal stroke model of rats. We also speculate that regulation of Cav-1, Mfsd2a, AQP4, and PDGFR-β expressions might be associated with its beneficial pharmacological effect, but remain to define and elucidate in future investigation.
Collapse
Affiliation(s)
- Min Zhou
- Department of Traditional Chinese Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Dongna Li
- Chinese Materia Medica College, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qian Shen
- Chinese Materia Medica College, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lei Gao
- The Microscopy Core Facility, Westlake University, Hangzhou, China
| | - Pengwei Zhuang
- Chinese Materia Medica College, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanjun Zhang
- Chinese Materia Medica College, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hong Guo
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
32
|
Wang K, Chen Y, Zhu X, Zou W, Zhou F. Ginkgo biloba Extract Attenuates Light-Induced Photoreceptor Degeneration by Modulating CAV-1—Redoxosome Signaling. Antioxidants (Basel) 2022; 11:antiox11071268. [PMID: 35883759 PMCID: PMC9311990 DOI: 10.3390/antiox11071268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/17/2022] [Accepted: 06/24/2022] [Indexed: 02/05/2023] Open
Abstract
The clinical potential of Ginkgo biloba extract (GBE) in the prevention and/or treatment of retinal degenerative diseases has been widely explored; however, the underlying molecular mechanism is poorly understood. Photoreceptor degeneration is the hallmark of retinal degenerative diseases and leads to vision impairment or loss. In this study, the effect of GBE against white light (WL) illumination-induced photoreceptor degeneration was investigated, as well as its underlying mechanism. To evaluate the in vitro activity of GBE, analysis of cell viability, cell apoptosis, oxidative stress, NOX (NADH oxidase) activity and mitochondrial membrane potential (MMP), as well as Western blotting and transcriptome sequencing and analysis, were conducted. To evaluate the in vivo activity of GBE, HE staining, electroretinography (ERG), Terminal-deoxynucleoitidyl transferase (TdT)-mediated nick end labeling (TUNEL) assay and immunofluorescence analysis were conducted. Our study showed that GBE treatment significantly attenuated WL illumination-induced oxidative damage in photoreceptor 661W cells—a finding that was also verified in C57BL/6J mice. Further molecular study revealed that WL illumination downregulated caveolin-1 (CAV-1) expression, interrupted CAV-1-NOX2 interaction, re-located NOX2 from the cell membrane to the cytoplasm and induced the formation of redoxosomes, which led to cell death. However, these cytotoxic events were significantly alleviated by GBE treatment. Interestingly, CAV-1 overexpression showed a consistent protective effect with GBE, while CAV-1 silencing impacted the protective effect of GBE against WL illumination-induced oxidative damage in in vitro and in vivo models. Thus, GBE was identified to prevent photoreceptor cell death due to CAV-1-dependent redoxosome activation, oxidative stress and mitochondrial dysfunction resulting from WL illumination. Overall, our study reveals the protective effect of GBE on photoreceptors against WL illumination-induced oxidative damage in in vitro and in vivo models, which effect is mediated through the modulation of CAV-1-redoxosome signaling. Our findings contribute to better understanding the therapeutic effect of GBE in preventing photoreceptor degeneration in retinal degenerative diseases, and GBE may become a novel therapeutic agent that is effective in reducing the morbidity of these diseases.
Collapse
Affiliation(s)
- Ke Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China; (Y.C.); (X.Z.)
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Correspondence: (K.W.); (W.Z.); Tel.: +86-510-8551-4482 (K.W.)
| | - Yuan Chen
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China; (Y.C.); (X.Z.)
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xue Zhu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China; (Y.C.); (X.Z.)
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Wenjun Zou
- Department of Ophthalmology, The Affiliated Wuxi No. 2 People’s Hospital of Nanjing Medical University, Wuxi 214002, China
- Correspondence: (K.W.); (W.Z.); Tel.: +86-510-8551-4482 (K.W.)
| | - Fanfan Zhou
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia;
| |
Collapse
|
33
|
Forouzandeh M, Mostafavi H, Ghasemloo E, Mohammadi P, Hosseini M, Eskandari M. Increased Expression of Tight Junction Proteins and Blood-Brain Barrier Integrity in MCAO Rats Following Injection of miR-149-5p. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2022; 11:223-235. [PMID: 37605737 PMCID: PMC10440002 DOI: 10.22088/ijmcm.bums.11.3.223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 06/18/2023] [Accepted: 07/09/2023] [Indexed: 08/23/2023]
Abstract
Cerebral ischemia is a common neurodegenerative disease in which damage to the blood-brain barrier (BBB) is the main consequence. In cerebral ischemia, the level of miR-149-5p and tight junction proteins are decreased, while the level of Calpine is increased, finally leading to increased BBB permeability. This study investigated the effect of miR-149-5p mimic on the expression of Calpain, Occludin, and ZO-1 and the consequences of cerebral ischemia. Cerebral ischemia model was performed via middle cerebral artery occlusion (MCAO) method on female Wistar rats. Four groups of Wistar rats were studied: Sham, cerebral ischemia without treatment, Scramble miR, and miR-149-5p mimic treatment. Then, neurological defects and BBB permeability (via Evans blue staining), cerebral edema (cerebrospinal fluid percentage), and ZO-1, Occludin, and Calapin expression (by quantitative real time- PCR) were investigated. qRT-PCR results showed miR-149-5p expression decreases after cerebral ischemia induction. In addition, Occludin and ZO-1 expression significantly increased in miR-149-5p group. In contrast, Calapin expression, BBB permeability, brain water content and neurological defects were significantly decreased. It seems that the increased level of miR-149-5p exerts its protective effect on cerebral ischemia due to increasing of tight junction proteins.
Collapse
Affiliation(s)
- Meysam Forouzandeh
- Faculty of Life Sciences and Biotechnology, Shahid-Beheshti Univ ersity, Tehran, Iran.
| | - Hossein Mostafavi
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Physiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Elham Ghasemloo
- Department of Physiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Parvin Mohammadi
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Masoume Hosseini
- Laboratory Expert, Depertment of Physiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Mehdi Eskandari
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
- Department of Physiology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
34
|
Chen S, Nazeri A, Baek H, Ye D, Yang Y, Yuan J, Rubin JB, Chen H. A review of bioeffects induced by focused ultrasound combined with microbubbles on the neurovascular unit. J Cereb Blood Flow Metab 2022; 42:3-26. [PMID: 34551608 PMCID: PMC8721781 DOI: 10.1177/0271678x211046129] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/16/2021] [Accepted: 08/22/2021] [Indexed: 01/29/2023]
Abstract
Focused ultrasound combined with circulating microbubbles (FUS+MB) can transiently enhance blood-brain barrier (BBB) permeability at targeted brain locations. Its great promise in improving drug delivery to the brain is reflected by a rapidly growing number of clinical trials using FUS+MB to treat various brain diseases. As the clinical applications of FUS+MB continue to expand, it is critical to have a better understanding of the molecular and cellular effects induced by FUS+MB to enhance the efficacy of current treatment and enable the discovery of new therapeutic strategies. Existing studies primarily focus on FUS+MB-induced effects on brain endothelial cells, the major cellular component of BBB. However, bioeffects induced by FUS+MB expand beyond the BBB to cells surrounding blood vessels, including astrocytes, microglia, and neurons. Together these cell types comprise the neurovascular unit (NVU). In this review, we examine cell-type-specific bioeffects of FUS+MB on different NVU components, including enhanced permeability in endothelial cells, activation of astrocytes and microglia, as well as increased intraneuron protein metabolism and neuronal activity. Finally, we discuss knowledge gaps that must be addressed to further advance clinical applications of FUS+MB.
Collapse
Affiliation(s)
- Si Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Arash Nazeri
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hongchae Baek
- Imaging Institute and Neurological Institute, Cleveland Clinic, Cleveland Clinic, Cleveland, OH, USA
| | - Dezhuang Ye
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Yaoheng Yang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Jinyun Yuan
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Joshua B Rubin
- Department of Pediatrics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
- Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
35
|
Wang LM, Wang YT, Yang WX. Engineered nanomaterials induce alterations in biological barriers: focus on paracellular permeability. Nanomedicine (Lond) 2021; 16:2725-2741. [PMID: 34870452 DOI: 10.2217/nnm-2021-0165] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Engineered nanoparticles (ENPs) are widely used in medical diagnosis and treatment, as food additives and as energy materials. ENPs may exert adverse or beneficial effects on the human body, which may be linked to interactions with biological barriers. In this review, the authors summarize the influences of four typical metal/metal oxide nanomaterials (Ag, TiO2, Au, ZnO nanoparticles) on the paracellular permeability of biological barriers. Disruptions on tight junctions, adhesion junctions, gap junctions and desmosomes via complex signaling pathways, such as the MAPK, PKC and ROCK signaling pathways, affect paracellular permeability. Reactive oxygen species and cytokines underlie the mechanism of ENP-triggered alterations in paracellular permeability. This review provides the information necessary for the cautious application of nanoparticles in medicine and life sciences in the future.
Collapse
Affiliation(s)
- Lan-Min Wang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Yu-Ting Wang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, PR China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, 310058, PR China
| |
Collapse
|
36
|
Microarray profiling predicts early neurological and immune phenotypic traits in advance of CNS disease during disease progression in Trypanosoma. b. brucei infected CD1 mouse brains. PLoS Negl Trop Dis 2021; 15:e0009892. [PMID: 34762691 PMCID: PMC8584711 DOI: 10.1371/journal.pntd.0009892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/10/2021] [Indexed: 01/09/2023] Open
Abstract
Human African trypanosomiasis (HAT), also known as sleeping sickness, is a major cause of mortality and morbidity in sub-Saharan Africa. We hypothesised that recent findings of neurological features and parasite brain infiltration occurring at much earlier stages in HAT than previously thought could be explained by early activation of host genetic programmes controlling CNS disease. Accordingly, a transcriptomal analysis was performed on brain tissue at 0, 7, 14, 21 and 28dpi from the HAT CD1/GVR35 mouse model. Up to 21dpi, most parasites are restricted to the blood and lymphatic system. Thereafter the trypanosomes enter the brain initiating the encephalitic stage. Analysis of ten different time point Comparison pairings, revealed a dynamic transcriptome comprising four message populations. All 7dpi Comparisons had by far more differentially expressed genes compared to all others. Prior to invasion of the parenchyma, by 7dpi, ~2,000 genes were up-regulated, denoted [7dpi↑] in contrast to a down regulated population [7dpi↓] also numbering ~2,000. However, by 14dpi both patterns had returned to around the pre-infected levels. The third, [28dpi↑] featured over three hundred transcripts which had increased modestly up to14dpi, thereafter were significantly up-regulated and peaked at 28dpi. The fourth, a minor population, [7dpi↑-28dpi↑], had similar elevated levels at 7dpi and 28dpi. KEGG and GO enrichment analysis predicted a diverse phenotype by 7dpi with changes to innate and adaptive immunity, a Type I interferon response, neurotransmission, synaptic plasticity, pleiotropic signalling, circadian activity and vascular permeability without disruption of the blood brain barrier. This key observation is consistent with recent rodent model neuroinvasion studies and clinical reports of Stage 1 HAT patients exhibiting CNS symptoms. Together, these findings challenge the strict Stage1/Stage2 phenotypic demarcation in HAT and show that that significant neurological, and immune changes can be detected prior to the onset of CNS disease.
Collapse
|
37
|
Huang SS, Cao S, Qin YB, Lu CE, Shen SR. Role of caveolin-1 in chronic postsurgical pain in rats. Exp Ther Med 2021; 22:1289. [PMID: 34630644 PMCID: PMC8461512 DOI: 10.3892/etm.2021.10724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 08/17/2021] [Indexed: 11/06/2022] Open
Abstract
Chronic postsurgical pain (CPSP) has a high incidence, but the underlying mechanisms remain elusive. Previous studies have indicated that caveolin-1 (Cav-1) plays a notable role in pain modulation. To study the role of Cav-1 in CPSP in the present study, a rat model of skin/muscle incision and retraction (SMIR) was established. Under anesthesia, skin and superficial muscle of the medial thigh were incised and a small pair of retractors inserted. It was revealed that SMIR increased the expression of Cav-1 in the dorsal root ganglion (DRG) and the injured tissue around the incision. Furthermore, the infiltration of endothelial cells and macrophages in the injured tissue around the incision increased constantly, and the vascular permeability increased due to the destruction of the vascular endothelial barrier function around the injured tissue. Cav-1 was mainly expressed by CD68-positive macrophages and CD34-positive endothelial cells in the injured tissues around the incision, while it was also primarily localized in the medium and large neurofilament 200-positive neurons and a small number of calcitonin gene-related peptide- and isolectin B4-positive small and medium-sized neurons in the DRG. The results demonstrated that the sustained high expression levels of Cav-1 in the injured tissue around the incision could lead to the dysfunction of the vascular endothelial barrier and, thus, could induce the inflammatory response through the lipoprotein transport of endothelial cells, thereby resulting in peripheral sensitization. In addition, the sustained high expression levels of Cav-1 in the DRG could sensitize large-sized neurons and change the transmission mode of noxious stimuli. The findings of the present study indicated that a Cav-1-mediated process could participate in neuronal transmission pathways associated with pain modulation.
Collapse
Affiliation(s)
- Sai-Sai Huang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Su Cao
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yi-Bin Qin
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Cui E Lu
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Shi-Ren Shen
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
38
|
Li L, Cheng SQ, Guo W, Cai ZY, Sun YQ, Huang XX, Yang J, Ji J, Chen YY, Dong YF, Cheng H, Sun XL. Oridonin prevents oxidative stress-induced endothelial injury via promoting Nrf-2 pathway in ischaemic stroke. J Cell Mol Med 2021; 25:9753-9766. [PMID: 34514714 PMCID: PMC8505855 DOI: 10.1111/jcmm.16923] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 12/19/2022] Open
Abstract
Oridonin, a natural diterpenoid compound extracted from a Chinese herb, has been proved to exert anti‐oxidative stress effects in various disease models. The aim of the present study was to investigate the protective effects of oridonin on oxidative stress‐induced endothelial injury in ischaemic stroke. We found oridonin repaired blood‐brain barrier (BBB) integrity presented with upregulation of tight junction proteins (TJ proteins) expression, inhibited the infiltration of periphery inflammatory cells and neuroinflammation and thereby reduced infarct volume in ischaemic stroke mice. Furthermore, our results showed that oridonin could protect against oxidative stress‐induced endothelial injury via promoting nuclear translocation of nuclear factor‐erythroid 2 related factor 2 (Nrf‐2). The specific mechanism could be the activation of AKT(Ser473)/GSK3β(Ser9)/Fyn signalling pathway. Our findings revealed the therapeutic effect and mechanism of oridonin in ischaemic stroke, which provided fundamental evidence for developing the extracted compound of Chinese herbal medicine into an innovative drug for ischaemic stroke treatment.
Collapse
Affiliation(s)
- Lei Li
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Shu-Qi Cheng
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Wei Guo
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Zhen-Yu Cai
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Yu-Qin Sun
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Xin-Xin Huang
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jin Yang
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Juan Ji
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Ya-Yun Chen
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Yin-Feng Dong
- Nanjing University of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hong Cheng
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiu-Lan Sun
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China.,Nanjing University of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
39
|
Pang L, Yang S, Dai W, Wu S, Kong J. Role of caveolin-1 in human organ function and disease: friend or foe? Carcinogenesis 2021; 43:2-11. [PMID: 34436568 DOI: 10.1093/carcin/bgab080] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/14/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022] Open
Abstract
Caveolin-1 (Cav-1) is a structural protein component of caveolae, which are invaginations of the plasma membrane involved in various cellular processes, including endocytosis, extracellular matrix organization, cholesterol distribution, cell migration, and signaling. Mounting evidence over the last 10-15 years has demonstrated a central role of Cav-1 in many diseases, such as cancer, diabetes, and fibrosis. Cav-1 plays positive and negative roles in various diseases through its different regulation pathways. Here, we review the current knowledge on Cav-1 in different diseases and discuss the role of this protein in human organs and diseases.
Collapse
Affiliation(s)
- Liwei Pang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shaojie Yang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wanlin Dai
- Innovation Institute of China Medical University, Shenyang, Liaoning, China
| | - Shuodong Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jing Kong
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
40
|
Caveolin-1, a novel player in cognitive decline. Neurosci Biobehav Rev 2021; 129:95-106. [PMID: 34237390 DOI: 10.1016/j.neubiorev.2021.06.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/29/2021] [Indexed: 12/12/2022]
Abstract
Cognitive decline (CD), which related to vascular dementia, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis and diabetes mellitus, is a growing health concern that has a great impact on the patients' quality of life. Although extensive efforts, the mechanisms of CD are still far from being clarified, not to mention the effective treatment and prevention strategies. Caveolin-1 (Cav-1), a trans-membrane protein, is a major component of the caveolae structure and scaffolding proteins. Recently, ample evidence depicts a strong correlation between Cav-1 and CD, however, the specific role of Cav-1 in CD has not been clearly examined and how they might be connected have yet to be identified. This review seeks to provide a comprehensive overview about how Cav-1 modulates pathogeneses of CD-associated diseases. In summary, Cav-1 can promote structural and functional plasticity of neurons, improve neurogenesis, relieve mitochondrial dysfunction, inhibit inflammation and suppress oxidative stress, which have shed light on the idea that Cav-1 may be an efficacious therapeutic target to treat CD.
Collapse
|
41
|
Jiao-Yan Y, Qing-Qing L, Xi L, Mei Z, Ting S, Na H, Wei J, Rui-Tao Z, Peng Y, Qi Y. Oxymatrine improves blood-brain barrier integrity after cerebral ischemia-reperfusion injury by downregulating CAV1 and MMP9 expression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 84:153505. [PMID: 33626426 DOI: 10.1016/j.phymed.2021.153505] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/20/2021] [Accepted: 02/06/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Ischemic stroke (IS) is a major neurological condition associated with extremely high morbidity and mortality worldwide. Oxymatrine (OMT), a quinolizidine alkaloid extracted from the root of Sophora flavescens, has neuroprotective properties and protects against IS. However, whether its protective effect involves alterations in the integrity of the blood-brain barrier (BBB) is unknown. PURPOSE Here, we used in vivo and in vitro models of IS to evaluate the protective effects of OMT and to establish whether its effects are mediated via the modulation of the BBB function. METHODS We assessed the effects of OMT by using neurological function scores, triphenyltetrazolium chloride staining, Nissl staining, and terminal deoxynucleotidyl transferase dUTP nick end labeling. RESULTS OMT significantly prevented cellular damage, improved neurological function, and reduced BBB permeability in a mouse model of cerebral ischemia-reperfusion. Additionally, OMT protected the function of the tight junctions of bEend.3 cells against the consequences of oxygen-glucose deprivation. Furthermore, intracranial lentivirus injection of short hairpin RNA targeting Cav1 decreased caveolin-1 expression and inhibited the neuroprotective effects of OMT. CONCLUSIONS OMT attenuated ischemia-reperfusion injury-induced damage to the BBB, and this neuroprotective action was at least partially dependent on the expression levels of CAV1 and MMP9 proteins. Therefore, OMT may offer effective protection against BBB injury induced by ischemia-reperfusion episodes.
Collapse
Affiliation(s)
- Yu Jiao-Yan
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Liu Qing-Qing
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Li Xi
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Zhao Mei
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Sun Ting
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Hu Na
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Jiang Wei
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Zhang Rui-Tao
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Yang Peng
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Yang Qi
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China.
| |
Collapse
|
42
|
Abstract
Acute brain injuries such as traumatic brain injury and stroke affect 85 million people a year worldwide, and many survivors suffer from long-term physical, cognitive, or psychosocial impairments. There are few FDA-approved therapies that are effective at preventing, halting, or ameliorating the state of disease in the brain after acute brain injury. To address this unmet need, one potential strategy is to leverage the unique physical and biological properties of nanomaterials. Decades of cancer nanomedicine research can serve as a blueprint for innovation in brain injury nanomedicines, both to emulate the successes and also to avoid potential pitfalls. In this review, we discuss how shared disease physiology between cancer and acute brain injuries can inform the design of novel nanomedicines for acute brain injuries. These disease hallmarks include dysregulated vasculature, an altered microenvironment, and changes in the immune system. We discuss several nanomaterial strategies that can be engineered to exploit these disease hallmarks, for example, passive accumulation, active targeting of disease-associated signals, bioresponsive designs that are "smart", and immune interactions.
Collapse
|
43
|
Filippenkov IB, Dergunova LV, Limborska SA, Myasoedov NF. Neuroprotective Effects of Peptides in the Brain: Transcriptome Approach. BIOCHEMISTRY (MOSCOW) 2021; 85:279-287. [PMID: 32564732 DOI: 10.1134/s0006297920030037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The importance of studying the action mechanisms of drugs based on natural regulatory peptides is commonly recognized. Particular attention is paid to the peptide drugs that contribute to the restoration of brain functions after acute cerebrovascular accidents (stroke), which for many years continues to be one of the main problems and threats to human health. However, molecular genetic changes in the brain in response to ischemia, as well as the mechanisms of protective effects of peptides, have not been sufficiently studied. This limits the use of neuroprotective peptides and makes it difficult to develop new, more efficient drugs with targeted action on brain functions. Transcriptome analysis is a promising approach for studying the mechanisms of the damaging effects of cerebral ischemia and neuroprotective action of peptide drugs. Beside investigating the role of mRNAs in protein synthesis, the development of new neuroprotection strategies requires studying the involvement of regulatory RNAs in ischemia. Of greatest interest are microRNAs (miRNAs) and circular RNAs (circRNAs), which are expressed predominantly in the brain. CircRNAs can interact with miRNAs and diminish their activity, thereby inhibiting miRNA-mediated repression of mRNAs. It has become apparent that analysis of the circRNA/miRNA/mRNA system is essential for deciphering the mechanisms of brain damage and repair. Here, we present the results of studies on the ischemia-induced changes in the activity of genes and peptide-mediated alterations in the transcriptome profiles in experimental ischemia and formulate the basic principles of peptide regulation in the ischemia-induced damage.
Collapse
Affiliation(s)
- I B Filippenkov
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia.
| | - L V Dergunova
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| | - S A Limborska
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| | - N F Myasoedov
- Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, 123182, Russia
| |
Collapse
|
44
|
Ahmadpour D, Grange-Messent V. Involvement of Testosterone Signaling in the Integrity of the Neurovascular Unit in the Male: Review of Evidence, Contradictions, and Hypothesis. Neuroendocrinology 2021; 111:403-420. [PMID: 32512571 DOI: 10.1159/000509218] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 06/08/2020] [Indexed: 11/19/2022]
Abstract
Age-related central nervous system function decline and increased susceptibility of females compared to males with respect to prevalence of several neurodegenerative and neuropsychiatric diseases are both based on the principle that hormonal factors could be involved. These cerebral disorders are characterized by an alteration of blood-brain barrier (BBB) properties and chronic neuroinflammation, which lead to disease progression. Neuroinflammation, in turn, contributes to BBB dysfunction. The BBB and its environment, called the neurovascular unit (NVU), are crucial for cerebral homeostasis and neuronal function. Interestingly, sex steroids influence BBB properties and modulate neuroinflammatory responses. To date however, the majority of work reported has focused on the effects of estrogens on BBB function and neuroinflammation in female mammals. In contrast, the effects of testosterone signaling on the NVU in males are still poorly studied. The aim of this review was to summarize and discuss the literature, providing insights and contradictions to highlight hypothesis and the need for further investigations.
Collapse
Affiliation(s)
- Delnia Ahmadpour
- Sorbonne Université, INSERM U1130, CNRS UMR 8246, Neuroscience Paris-Seine, Institut de Biologie Paris-Seine, Paris, France
| | - Valérie Grange-Messent
- Sorbonne Université, INSERM U1130, CNRS UMR 8246, Neuroscience Paris-Seine, Institut de Biologie Paris-Seine, Paris, France,
| |
Collapse
|
45
|
Torices S, Roberts SA, Park M, Malhotra A, Toborek M. Occludin, caveolin-1, and Alix form a multi-protein complex and regulate HIV-1 infection of brain pericytes. FASEB J 2020; 34:16319-16332. [PMID: 33058236 PMCID: PMC7686148 DOI: 10.1096/fj.202001562r] [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: 06/19/2020] [Revised: 09/18/2020] [Accepted: 10/02/2020] [Indexed: 12/24/2022]
Abstract
HIV-1 enters the brain by altering properties of the blood-brain barrier (BBB). Recent evidence indicates that among cells of the BBB, pericytes are prone to HIV-1 infection. Occludin (ocln) and caveolin-1 (cav-1) are critical determinants of BBB integrity that can regulate barrier properties of the BBB in response to HIV-1 infection. Additionally, Alix is an early acting endosomal factor involved in HIV-1 budding from the cells. The aim of the present study was to evaluate the role of cav-1, ocln, and Alix in HIV-1 infection of brain pericytes. Our results indicated that cav-1, ocln, and Alix form a multi-protein complex in which they cross-regulate each other's expression. Importantly, the stability of this complex was affected by HIV-1 infection. Modifications of the complex resulted in diminished HIV-1 infection and alterations of the cytokine profile produced by brain pericytes. These results identify a novel mechanism involved in HIV-1 infection contributing to a better understanding of the HIV-1 pathology and the associated neuroinflammatory responses.
Collapse
Affiliation(s)
- Silvia Torices
- Department of Biochemistry and Molecular BiologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Samantha A. Roberts
- Department of Biochemistry and Molecular BiologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Minseon Park
- Department of Biochemistry and Molecular BiologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Arun Malhotra
- Department of Biochemistry and Molecular BiologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| | - Michal Toborek
- Department of Biochemistry and Molecular BiologyUniversity of Miami Miller School of MedicineMiamiFLUSA
| |
Collapse
|
46
|
AKAP12 Supports Blood-Brain Barrier Integrity against Ischemic Stroke. Int J Mol Sci 2020; 21:ijms21239078. [PMID: 33260683 PMCID: PMC7730430 DOI: 10.3390/ijms21239078] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/21/2020] [Accepted: 11/22/2020] [Indexed: 12/15/2022] Open
Abstract
A-kinase anchor protein 12 (AKAP12) is a scaffolding protein that associates with intracellular molecules to regulate multiple signal transductions. Although the roles of AKAP12 in the central nervous system are still relatively understudied, it was previously shown that AKAP12 regulates blood-retinal barrier formation. In this study, we asked whether AKAP12 also supports the function and integrity of the blood-brain barrier (BBB). In a mouse model of focal ischemia, the expression level of AKAP12 in cerebral endothelial cells was upregulated during the acute phase of stroke. Also, in cultured cerebral endothelial cells, oxygen-glucose deprivation induced the upregulation of AKAP12. When AKAP12 expression was suppressed by an siRNA approach in cultured endothelial cells, endothelial permeability was increased along with the dysregulation of ZO-1/Claudin 5 expression. In addition, the loss of AKAP12 expression caused an upregulation/activation of the Rho kinase pathway, and treatment of Rho kinase inhibitor Y-27632 mitigated the increase of endothelial permeability in AKAP12-deficient endothelial cell cultures. These in vitro findings were confirmed by our in vivo experiments using Akap12 knockout mice. Compared to wild-type mice, Akap12 knockout mice showed a larger extent of BBB damage after stroke. However, the inhibition of rho kinase by Y-27632 tightened the BBB in Akap12 knockout mice. These data may suggest that endogenous AKAP12 works to alleviate the damage and dysfunction of the BBB caused by ischemic stress. Therefore, the AKAP12-rho-kinase signaling pathway represents a novel therapeutic target for stroke.
Collapse
|
47
|
Caveolin-1 Derived from Brain Microvascular Endothelial Cells Inhibits Neuronal Differentiation of Neural Stem/Progenitor Cells In Vivo and In Vitro. Neuroscience 2020; 448:172-190. [DOI: 10.1016/j.neuroscience.2020.09.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 09/10/2020] [Accepted: 09/13/2020] [Indexed: 12/14/2022]
|
48
|
Tan Z, Jiang J, Tian F, Peng J, Yang Z, Li S, Long X. Serum Visinin-Like Protein 1 Is a Better Biomarker Than Neuron-Specific Enolase for Seizure-Induced Neuronal Injury: A Prospective and Observational Study. Front Neurol 2020; 11:567587. [PMID: 33071949 PMCID: PMC7544981 DOI: 10.3389/fneur.2020.567587] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 08/13/2020] [Indexed: 11/13/2022] Open
Abstract
Introduction: Visinin-like protein 1 (VILIP-1) is an established biomarker of neuronal injury. The levels of serum VILIP-1, neuron-specific enolase (NSE) and caveolin-1 (CAV-1) were measured to investigate potential of VILIP-1 as a biomarker for seizure-induced neuronal injury, and the correlation of VILIP-1 with severity of epilepsy and blood-brain barrier dysfunction were investigated. Materials and Methods: Patient with epilepsy from 14 to 70 years of age and age-, sex-matched healthy subjects were involved in this study. All blood sample of patients were collected within 3–72 h after the seizure. The severity of epilepsy and levels of serum VILIP-1, NSE and CAV-1 were measured. Accuracy of VILIP-1 and NSE was obtained from receiver operating curve analyses. Associations between VILIP-1 and severity of epilepsy, VILIP-1 and CAV-1 were investigated. Results: A total of 58 patients and 29 healthy control subjects were included in our study. The levels of serum VILIP-1, NSE, and CAV-1 in the patient group were significantly higher than those in the control group. VILIP-1 has higher and significant accuracy for assessing seizure-induced neuronal injury compared with NSE. VILIP-1 levels were positively associated with severity of epilepsy and CAV-1 in patients with epilepsy. Conclusions: VILIP-1 may be a better serum biomarker than NSE for assessing seizure-induced neuronal injury and even brain injury caused by various pathological condition. Further studies are required to explore the clinical contribution of VILIP-1 in diagnosis, treatment strategies and outcome assessments of epilepsy.
Collapse
Affiliation(s)
- Zheren Tan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jianlin Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Fafa Tian
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Jinxin Peng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhiquan Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Shuyu Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyan Long
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
49
|
Song Y, Xie F, Ma S, Deng G, Li Y, Nie Y, Wang F, Yu G, Gao Z, Chen K, Han L, Gao L. Caveolin-1 protects against DSS-induced colitis through inhibiting intestinal nitrosative stress and mucosal barrier damage in mice. Biochem Pharmacol 2020; 180:114153. [PMID: 32679126 DOI: 10.1016/j.bcp.2020.114153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 01/12/2023]
|
50
|
Ye JH, Shi JJ, Yin X, Wu HY, Xu XY, Yao YZ, Zhang WJ. Elevated Expression of CAV1 is Associated with Unfavorable Prognosis of Patients with Breast Cancer Who Undergo Surgery and Neoadjuvant Chemotherapy. Cancer Manag Res 2020; 12:8887-8892. [PMID: 33061584 PMCID: PMC7519866 DOI: 10.2147/cmar.s264673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/08/2020] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Neoadjuvant chemotherapy (NACT), which is standard treatment for locally advanced breast cancer, improves the resectability of patients with early breast cancer and reduces the extent of breast and axillary surgery. Caveolin-1 (CAV1) is implicated in human cancers, although its utility for cancer prognosis is unknown. Here, we investigated the expression of CAV1 in breast cancer tissues to evaluate its prognostic significance on patients with breast cancer administered NACT. METHODS CAV1 expression in 80 breast cancer tissue samples was evaluated using immunohistochemistry (IHC). The association between CAV1 levels and clinical factors was analyzed using the chi-square test and that between CAV1 and prognosis was evaluated using multivariate Cox regression and Kaplan-Meier analyses. RESULTS High levels of CAV1 were significantly associated with survival, and patients with overexpression of CAV1 had a poor prognosis. Adjusted multivariate Cox regression analyses revealed that a high level of CAV1 expression was an independent, significant prognostic factor for patients with breast cancer treated with NACT. DISCUSSION Overexpression of CAV1 in patients with breast cancer administered NACT was associated with shorter disease-free survival and overall survival. Therefore, high levels of CAV1 may serve as a prognostic biomarker for such patients.
Collapse
Affiliation(s)
- Jia-Hui Ye
- General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Jia-Jun Shi
- General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Xi Yin
- General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Hong-Yan Wu
- Pathology Department, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Xin-Yun Xu
- Pathology Department, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Yong-Zhong Yao
- General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| | - Wei-Jie Zhang
- General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People’s Republic of China
| |
Collapse
|