1
|
Justić H, Barić A, Ratko M, Šimunić I, Radmilović M, Pongrac M, Škokić S, Dobrivojević Radmilović M. The temporal dynamic of bradykinin type 2 receptor effects reveals its neuroprotective role in the chronic phase of cerebral and retinal ischemic injury. J Cereb Blood Flow Metab 2025; 45:153-170. [PMID: 39113417 PMCID: PMC11572167 DOI: 10.1177/0271678x241270241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/31/2024] [Accepted: 06/17/2024] [Indexed: 11/20/2024]
Abstract
The activation of the bradykinin type 2 receptor is intricately involved in acute post-ischemic inflammatory responses. However, its precise role in different stages of ischemic injury, especially in the chronic phase, remains unclear. Following simultaneous cerebral and retinal ischemia, bradykinin type 2 receptor knockout mice and their controls were longitudinally monitored for 35 days via magnetic resonance imaging, fundus photography, fluorescein angiography, behavioral assessments, vascular permeability measurements, and immunohistochemistry, as well as glycemic status assessments. Without impacting the lesion size, bradykinin type 2 receptor deficiency reduced acute cerebral vascular permeability preventing the loss of pericytes and tight junctions. In the chronic phase of ischemia, however, it resulted in increased astrogliosis and cortical neuronal loss, as well as higher functional deficits. The retinal findings demonstrated a similar pattern. Bradykinin type 2 receptor deficiency delayed, but exacerbated the development of retinal necrosis, increased subacute vascular permeability, and promoted retinal ganglion cell loss in the chronic phase of ischemia. This investigation sheds light on the temporal dynamic of bradykinin type 2 receptor effects in ischemia, pointing to a therapeutic potential in the subacute and chronic phases of ischemic injury.
Collapse
Affiliation(s)
- Helena Justić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Anja Barić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Martina Ratko
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Iva Šimunić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Marin Radmilović
- Sestre milosrdnice University Hospital Center, Department of Ophthalmology, Zagreb, Croatia
| | - Marta Pongrac
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Siniša Škokić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Marina Dobrivojević Radmilović
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
- Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
2
|
Wang M, Guo C, Yang J, Li J, Hu J, Peng Z, Guo M, Zhang L, Li F, Yang Q, Zi W, Wang P. The effectiveness and safety of human urinary kallidinogenase in acute ischemic stroke patients undergoing endovascular therapy. J Cereb Blood Flow Metab 2024; 44:1565-1576. [PMID: 38459953 PMCID: PMC11418712 DOI: 10.1177/0271678x241238033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 12/30/2023] [Accepted: 01/17/2024] [Indexed: 03/11/2024]
Abstract
The effectiveness and safety of human urinary kallidinogenase (HUK) in acute ischemic stroke (AIS) patients undergoing endovascular therapy (EVT) due to large vessel occlusion (LVO) was unclear. A pooled analysis was performed using individual data from the DEVT and RESCUE BT trials. Patients were divided into two groups based on HUK treatment. The primary outcome was the 90-day modified Rankin Scale (mRS) score. Safety outcomes included 90-day mortality and symptomatic intracranial hemorrhage (sICH) within 48 hours. A total of 1174 patients were included in the study. Of these, 150 (12.8%) patients received HUK. The adjusted common odds ratio (OR) of the mRS score was 1.458 (95% confidence interval [CI] = 1.072-1.983; p = 0.016) favoring HUK. The incidence of sICH (2.0% vs. 8.6%; adjusted OR: 0.198; 95% CI: 0.061-0.638; p = 0.007) and mortality (11.3% vs.18.5%; adjusted OR: 0.496; 95% CI: 0.286-0.862; p = 0.013) was lower in HUK group than non-HUK group. This association was consistent with propensity score-matching and the inverse probability of treatment weighting analysis. In conclusion, HUK was safe and associated with a preferable prognosis in AIS patients due to LVO in the anterior circulation.
Collapse
Affiliation(s)
- Mengmeng Wang
- Department of Neurology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Shandong Province, China
- Clinical College, Weifang Medical University, Weifang, China
| | - Changwei Guo
- Department of Neurology, Xinqiao Hospital and The Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jie Yang
- Department of Neurology, Xinqiao Hospital and The Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jing Li
- Department of Neurology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Shandong Province, China
- Clinical College, Weifang Medical University, Weifang, China
| | - Jinrong Hu
- Department of Neurology, Xinqiao Hospital and The Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhouzhou Peng
- Department of Neurology, Xinqiao Hospital and The Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Meng Guo
- The Thirteenth People’s Hospital of Chongqing, Chongqing, China
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lingyu Zhang
- Department of Neurology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Shandong Province, China
- Clinical College, Weifang Medical University, Weifang, China
| | - Fengli Li
- Department of Neurology, Xinqiao Hospital and The Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qingwu Yang
- Department of Neurology, Xinqiao Hospital and The Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wenjie Zi
- Department of Neurology, Xinqiao Hospital and The Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Pengfei Wang
- Department of Neurology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Shandong Province, China
| |
Collapse
|
3
|
Qiu X, Ma C, Luo Z, Zhang Y, Kang J, Zhu D, Wang Z, Li L, Wei Z, Wang Z, Kang X. Bradykinin protects nucleus pulposus cells from tert-butyl hydroperoxide-induced damage and delays intervertebral disc degeneration. Int Immunopharmacol 2024; 134:112161. [PMID: 38728878 DOI: 10.1016/j.intimp.2024.112161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/12/2024]
Abstract
Intervertebral disc degeneration (IVDD) is a leading cause of degenerative spinal disorders, involving complex biological processes. This study investigates the role of the kallikrein-kinin system (KKS) in IVDD, focusing on the protective effects of bradykinin (BK) on nucleus pulposus cells (NPCs) under oxidative stress. Clinical specimens were collected, and experiments were conducted using human and rat primary NPCs to elucidate BK's impact on tert-butyl hydroperoxide (TBHP)-induced oxidative stress and damage. The results demonstrate that BK significantly inhibits TBHP-induced NPC apoptosis and restores mitochondrial function. Further analysis reveals that this protective effect is mediated through the BK receptor 2 (B2R) and its downstream PI3K/AKT pathway. Additionally, BK/PLGA sustained-release microspheres were developed and validated in a rat model, highlighting their potential therapeutic efficacy for IVDD. Overall, this study sheds light on the crucial role of the KKS in IVDD pathogenesis and suggests targeting the B2R as a promising therapeutic strategy to delay IVDD progression and promote disc regeneration.
Collapse
Affiliation(s)
- Xiaoming Qiu
- Department of Orthopedics, Lanzhou University Second Hospital, The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Gansu provincial hospital of TCM (The First Affiliated Hospital of Gansu University of Chinese Medicine), Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu 730000, China
| | - Chongwen Ma
- Department of Orthopedics, Lanzhou University Second Hospital, The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu 730000, China
| | - Zhangbin Luo
- Department of Orthopedics, Lanzhou University Second Hospital, The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu 730000, China
| | - Yibao Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu 730000, China
| | - Jihe Kang
- Department of Orthopedics, Lanzhou University Second Hospital, The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu 730000, China
| | - Daxue Zhu
- Department of Orthopedics, Lanzhou University Second Hospital, The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu 730000, China
| | - Zhaoheng Wang
- Department of Orthopedics, Lanzhou University Second Hospital, The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu 730000, China
| | - Lei Li
- Department of Orthopedics, Lanzhou University Second Hospital, The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu 730000, China
| | - Ziyan Wei
- Department of Orthopedics, Lanzhou University Second Hospital, The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu 730000, China
| | - Zhuanping Wang
- Department of endocrinology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, Gansu 730000, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu 730000, China
| | - Xuewen Kang
- Department of Orthopedics, Lanzhou University Second Hospital, The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Orthopaedics Key Laboratory of Gansu Province, Lanzhou, Gansu 730000, China.
| |
Collapse
|
4
|
Xu G, Dong F, Su L, Tan ZX, Lei M, Li L, Wen D, Zhang F. The role and therapeutic potential of nuclear factor κB (NF-κB) in ischemic stroke. Biomed Pharmacother 2024; 171:116140. [PMID: 38211425 DOI: 10.1016/j.biopha.2024.116140] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/13/2024] Open
Abstract
Stroke is a prevalent cerebrovascular condition with a global impact, causing significant rates of illness and death. Despite extensive research, the available treatment options for stroke remain restricted. Hence, it is crucial to gain a deeper understanding of the molecular mechanisms associated with the onset and advancement of stroke in order to establish a theoretical foundation for novel preventive and therapeutic approaches. NF-κB, also known as nuclear factor κB, is a transcription factor responsible for controlling the expression of numerous genes and plays a crucial role in diverse physiological processes. NF-κB is triggered and regulates neuroinflammation and other processes after stroke, promoting the generation of cytokine storms and contributing to the advancement of ischemic stroke (IS). Therefore, NF-κB could potentially play a vital role in stroke by regulating diverse pathophysiological processes. This review provides an overview of the functions of NF-κB in stroke and its governing mechanisms. In addition, our attention is directed towards various potential therapies that aim to inhibit the NF-κB signaling pathway in order to offer valuable insights for the advancement of innovative treatment approaches for stroke.
Collapse
Affiliation(s)
- Guangyu Xu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Lei Su
- Department of Radiotherapy, Affiliated Hospital of Hebei University, Baoding 071000, PR China
| | - Zi-Xuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Mingcheng Lei
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Lina Li
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang 050017, PR China; Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang 050017, PR China; Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, PR China.
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang 050051, PR China.
| |
Collapse
|
5
|
Vespakinin-M, a natural peptide from Vespa magnifica, promotes functional recovery in stroke mice. Commun Biol 2022; 5:74. [PMID: 35058552 PMCID: PMC8776894 DOI: 10.1038/s42003-022-03024-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 12/28/2021] [Indexed: 02/06/2023] Open
Abstract
Acute ischemic stroke triggers complex systemic pathological responses for which the exploration of drug resources remains a challenge. Wasp venom extracted from Vespa magnifica (Smith, 1852) is most commonly used to treat rheumatoid arthritis as well as neurological disorders. Vespakinin-M (VK), a natural peptide from wasp venom, has remained largely unexplored for stroke. Herein, we first confirmed the structure, stability, toxicity and distribution of VK as well as its penetration into the blood–brain barrier. VK (150 and 300 µg/kg, i.p.) was administered to improve stroke constructed by middle cerebral artery occlusion in mice. Our results indicate that VK promote functional recovery in mice after ischemia stroke, including an improvement of neurological impairment, reduction of infarct volume, maintenance of blood-brain barrier integrity, and an obstruction of the inflammatory response and oxidative stress. In addition, VK treatment led to reduced neuroinflammation and apoptosis associated with the activation of PI3K–AKT and inhibition of IκBα–NF-κB signaling pathways. Simultaneously, we confirmed that VK can combine with bradykinin receptor 2 (B2R) as detected by molecular docking, the B2R antagonist HOE140 could counteract the neuro-protective effects of VK on stroke in mice. Overall, targeting the VK–B2R interaction can be considered as a practical strategy for stroke therapy. Zhao et al establish the structure, stability, toxicity and distribution of vespakinin-M (VK) as well as its penetration into the blood–brain barrier in mice. They go on to show that VK promotes functional recovery in mice after ischemia stroke and shed light on the potential underlying mechanisms, which together indicates the potential therapeutic value of targeting VK in stroke therapy.
Collapse
|
6
|
Peng Y, Liu D, Diao Z, Wang Z, Ding H, Cai B, Hu Y, Zhao G, Zheng M. Down-regulation of B2R contributes to preeclampsia by inhibiting human trophoblast cell invasion and angiogenesis. Pregnancy Hypertens 2020; 21:14-22. [DOI: 10.1016/j.preghy.2020.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 04/06/2020] [Accepted: 04/17/2020] [Indexed: 12/18/2022]
|
7
|
Zhang H, Pan Q, Xie Z, Chen Y, Wang J, Bihl J, Zhong W, Chen Y, Zhao B, Ma X. Implication of MicroRNA503 in Brain Endothelial Cell Function and Ischemic Stroke. Transl Stroke Res 2020; 11:1148-1164. [PMID: 32285355 DOI: 10.1007/s12975-020-00794-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/12/2022]
Abstract
The role of miR-503 in brain endothelium and ischemic stroke (IS) remains unclear. We aimed to study the relationship between plasma miR-503 and the onset time, severity, subtypes, and von Willebrand Factor (vWF) level in IS patients and to investigate the roles and underlying mechanisms of miR-503 in middle cerebral artery occlusion (MCAO) mice and cultured cerebral vascular endothelial cells (ECs). In MCAO mice, the effects of plasma from acute severe IS patients (ASS) with or without miR-503 antagomir on brain and ECs damage were determined. In cultured human ECs, the effects of miR-503 overexpression or knockdown on the monolayer permeability, apoptosis, ROS, and NO generation were investigated. For mechanism study, the PI3K/Akt/eNOS pathway, cleaved caspase-3, and bcl-2 were analyzed. Results showed that plasma miR-503 was significantly increased in IS patients, especially in acute period and severe cases and subtypes of LAA and TACI, and was positively correlated with vWF. Logistic analysis indicated that miR-503 was an independent risk factor for IS, with the area under curve of 0.796 in ROC analysis. In MCAO mice, ASS pretreatment aggravated neurological injury, BBB damage, brain edema, CBF reduction, and decreased NO production while increased apoptosis and ROS generation in brain ECs, which were partly abolished by miR-503 antagomir. In cultured ECs, miR-503 overexpression and knockdown confirmed its effects on regulating monolayer permeability, cell apoptosis, NO, and ROS generation via PI3K/Akt/eNOS pathway or bcl-2 and cleaved caspase-3 proteins. These together indicate that miR-503 is a promising biomarker and novel therapeutic target for IS.
Collapse
Affiliation(s)
- Huiting Zhang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China
| | - Qunwen Pan
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China
| | - Zi Xie
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China
| | - Yanyu Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China
| | - Jinju Wang
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45430, USA
| | - Ji Bihl
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45430, USA
| | - Wangtao Zhong
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China
| | - Yanfang Chen
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45430, USA
| | - Bin Zhao
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China.
| | - Xiaotang Ma
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Institute of Neurology, Affiliated Hospital of Guangdong Medical University, 57 South Renmin Road, Zhanjiang, 524001, China.
| |
Collapse
|
8
|
High Level of Serum Tissue Kallikrein Is Associated with Favorable Outcome in Acute Ischemic Stroke Patients. DISEASE MARKERS 2019; 2019:5289715. [PMID: 31275448 PMCID: PMC6589205 DOI: 10.1155/2019/5289715] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/11/2019] [Indexed: 12/13/2022]
Abstract
Background/Objectives We sought to assess the association between a serum tissue kallikrein (TK) level and a 90-day outcome in acute ischemic stroke (AIS) patients who received acute reperfusion therapy. Methods Consecutive AIS patients within 6 hours after stroke onset between December 2015 and August 2017 were prospectively recruited. Blood samples were collected before acute reperfusion therapy for serum TK measurement. Outcome was modified Rankin scale (mRS) score at 90 days after stroke onset. Binary logistic regression was performed to analyze the association between the baseline TK level and the clinical outcome. Results Between December 2015 and August 2017, 75 patients (age range from 33 to 91 years, 72.0% male) were recruited in this study. Higher baseline TK was independently associated with a favorable functional outcome (mRS 0-2) (odds ratio 1.01, 95% confidence interval (CI) 1.00-1.02, p = 0.047) and low mortality rate (odds ratio 0.98, 95% CI 0.96-1.00, p = 0.049) at 90 days. Increased TK level was associated with 90 d mRS (0-2) with area under the curve of 0.719 (95% CI 0.596-0.842; p = 0.002). Conclusions Serum TK can be a promising predictor of clinical outcome in AIS patients who received acute reperfusion therapy.
Collapse
|
9
|
Wang J, Ji E, Lin C, Wang L, Dai L, Gao W. Effects of bradykinin on the survival of multiterritory perforator flaps in rats. World J Surg Oncol 2019; 17:44. [PMID: 30813916 PMCID: PMC6394035 DOI: 10.1186/s12957-019-1570-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/27/2019] [Indexed: 02/08/2023] Open
Abstract
Background Bradykinin, a vasoactive peptide, has many biological functions. For example, it accelerates angiogenesis. Thus, we studied the effects of bradykinin on the survival of perforator flaps. Methods Averagely, 50 male Sprague–Dawley rats were divided into control and bradykinin groups and underwent procedures to the multiterritory perforator flap. Areas of flap survival were tested 7 days later. Flap perfusion was evaluated by laser Doppler imaging. We assessed the extent of autophagy by determining LC3-II/I, Beclin 1, and p62. Flap angiogenesis was assessed by immunohistochemistry and H&E staining. We measured the level of vascular endothelial growth factor (VEGF) protein using western blot. We assessed oxidative stress by measuring the activity of superoxide dismutase (SOD) and malondialdehyde (MDA) levels. The apoptotic index was also evaluated by western blot, and we determined nitric oxide (NO) production using an NO assay kit. Results The bradykinin group exhibited significantly larger areas of flap survival, higher blood supply, and more neovascularization. The bradykinin group also had higher SOD activity, higher VEGF expression and NO content, and reduced MDA compared to the control group. Rats treated with bradykinin also had lower levels of apoptosis and autophagy relative to the control group. Conclusion Our results suggest that bradykinin promotes the survival of multiterritory perforator flaps by increasing angiogenesis, promoting the release of NO, suppressing apoptosis, reducing oxidative stress, and inhibiting autophagy.
Collapse
Affiliation(s)
- Jieke Wang
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, The Second Clinical Medical College of Wenzhou Medical University, Wenzhou Medical University, No. 109, Xue Yuan Road (West), Lucheng District, Wenzhou, 325000, China
| | - Encheng Ji
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, The Second Clinical Medical College of Wenzhou Medical University, Wenzhou Medical University, No. 109, Xue Yuan Road (West), Lucheng District, Wenzhou, 325000, China
| | - Chen Lin
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, The Second Clinical Medical College of Wenzhou Medical University, Wenzhou Medical University, No. 109, Xue Yuan Road (West), Lucheng District, Wenzhou, 325000, China
| | - Long Wang
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, The Second Clinical Medical College of Wenzhou Medical University, Wenzhou Medical University, No. 109, Xue Yuan Road (West), Lucheng District, Wenzhou, 325000, China
| | - Li Dai
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, The Second Clinical Medical College of Wenzhou Medical University, Wenzhou Medical University, No. 109, Xue Yuan Road (West), Lucheng District, Wenzhou, 325000, China
| | - Weiyang Gao
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, The Second School of Medicine, The Second Clinical Medical College of Wenzhou Medical University, Wenzhou Medical University, No. 109, Xue Yuan Road (West), Lucheng District, Wenzhou, 325000, China.
| |
Collapse
|
10
|
Alexander-Curtis M, Pauls R, Chao J, Volpi JJ, Bath PM, Verdoorn TA. Human tissue kallikrein in the treatment of acute ischemic stroke. Ther Adv Neurol Disord 2019; 12:1756286418821918. [PMID: 30719079 PMCID: PMC6348491 DOI: 10.1177/1756286418821918] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 10/24/2018] [Indexed: 01/08/2023] Open
Abstract
Acute ischemic stroke (AIS) remains a major cause of death and disability throughout the world. The most severe form of stroke results from large vessel occlusion of the major branches of the Circle of Willis. The treatment strategies currently available in western countries for large vessel occlusion involve rapid restoration of blood flow through removal of the offending blood clot using mechanical or pharmacological means (e.g. tissue plasma activator; tPA). This review assesses prospects for a novel pharmacological approach to enhance the availability of the natural enzyme tissue kallikrein (KLK1), an important regulator of local blood flow. KLK1 is responsible for the generation of kinins (bradykinin and kallidin), which promote local vasodilation and long-term vascularization. Moreover, KLK1 has been used clinically as a direct treatment for multiple diseases associated with impaired local blood flow including AIS. A form of human KLK1 isolated from human urine is approved in the People's Republic of China for subacute treatment of AIS. Here we review the rationale for using KLK1 as an additional pharmacological treatment for AIS by providing the biochemical mechanism as well as the human clinical data that support this approach.
Collapse
Affiliation(s)
| | - Rick Pauls
- DiaMedica Therapeutics, Minneapolis, MN, USA
| | - Julie Chao
- Medical University of South Carolina, Department of Biochemistry and Molecular Biology, Charleston, SC, USA
| | - John J Volpi
- Houston Methodist, Stanley H. Appel Department of Neurology, Houston, TX, USA
| | - Philip M Bath
- Stroke Trials Unit, University of Nottingham, City Hospital Campus, Nottingham, UK
| | | |
Collapse
|
11
|
Zhang JY, Bai QK, Zhang YD. Pretreatment with simvastatin upregulates expression of BK-2R and CD11b in the ischemic penumbra of rats. J Biomed Res 2018; 32:354-360. [PMID: 29784898 PMCID: PMC6163114 DOI: 10.7555/jbr.32.20160152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductases, collectively known as statins, have been shown to minimize cerebral ischemic events in patients. We assessed the mechanisms of simvastatin pretreatment in preventing cerebral ischemia/reperfusion injury in rats using a model of middle cerebral artery occlusion (MCAO). Rats were pretreated with simvastatin 14 days prior to MCAO induction. At 3, 24, and 48 hours after reperfusion, bradykinin levels in the ischemic penumbra were assayed by ELISA, mRNA levels of bradykinin B2 receptors (BK-2Rs) and CD11b were measured by fluorescent quantitative real-time PCR (RT-PCR), and co-expression of microglia and BK-2Rs was determined by immunofluorescence. Simvastatin had no effect on bradykinin expression in the ischemic penumbra at any time point. However, the levels of BK-2R and CD11b mRNA in the ischemic penumbra, which were significantly decreased 3 hours after ischemia-reperfusion, were increased in simvastatin-pretreated rats. Moreover, the co-expression of BK-2Rs and microglia was confirmed by immunofluorescence analysis. These results suggest that the beneficial effects of simvastatin pretreatment before cerebral ischemia/reperfusion injury in rats may be partially due to increased expression of BK-2R and CD11b in the ischemic penumbra.
Collapse
Affiliation(s)
- Jian-Ying Zhang
- Department of Neurology, Pudong People's Hospital, Shanghai 201299, China
| | - Qing-Ke Bai
- Department of Neurology, Pudong People's Hospital, Shanghai 201299, China
| | - Ying-Dong Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| |
Collapse
|
12
|
Miao Z, Guo M, Zhou S, Sun X, Wang F, Lu H, Cui Z. Smoking and drinking influence the advancing of ischemic stroke disease by targeting PTGS2 and TNFAIP3. Exp Ther Med 2018; 16:61-66. [PMID: 29977356 PMCID: PMC6030864 DOI: 10.3892/etm.2018.6138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 01/08/2018] [Indexed: 01/26/2023] Open
Abstract
In the present study, we explored the influence of cigarette smoking and alcohol drinking on gene expression level and related functions and pathways on the development of ischemic stroke (IS) disease. The gene expression profile of E-GEOD-22255 was obtained from 20 IS samples (7 patients without smoking or drinking history and 13 patients with smoking or drinking history) and 20 controls (9 normal controls without smoking or drinking history and 11 controls with smoking or drinking history). The correlation degree between gene expression and grouping were measured by significance analysis of microarray (SAM). Smoking or drinking-related DEGs were screened. GO functional and KEGG pathway enrichment analyses were processed. Based on the KEGG database, a pathway relationship network was constructed. DEGs in significant functions and pathways were inserted and regarded as key DEGs. Gene co-expression network was constructed based on the expression value of key genes. In total, 319 IS-related DEGs, which were induced by smoking and drinking, were screened and enriched in various functions and pathways, including inflammatory response, nuclear factor-κB (NF-κB) signaling pathway and influenza A. Pathway relationship network was constructed with 44 nodes and the hub node was the MAPK signaling pathway. After merging, 87 key DEGs were obtained. The gene co-expression network with 43 node edges was constructed and the hub node was prostaglandin-endoperoxide synthase 2. In IS patients, smoking and drinking may induce different expression of many genes, including PTGS2, TNFAIP3, ZFP36 and NFKBIZ. In addition, these genes participated in various pathways, such as inflammatory response.
Collapse
Affiliation(s)
- Zhimin Miao
- Department of Neurology Two, The People's Hospital of Shouguang, Weifang, Shandong 262700, P.R. China
| | - Meifang Guo
- Department of Oncology Two, The People's Hospital of Shouguang, Weifang, Shandong 262700, P.R. China
| | - Suqin Zhou
- Department of Blood Rheumatic Immunology, The People's Hospital of Shouguang, Weifang, Shandong 262700, P.R. China
| | - Xuemei Sun
- Department of Respiratory Medicine One, The People's Hospital of Shouguang, Weifang, Shandong 262700, P.R. China
| | - Fang Wang
- Department of General Surgery Three, The People's Hospital of Shouguang, Weifang, Shandong 262700, P.R. China
| | - Haiying Lu
- Department of Yang Kou Medical Ward, The People's Hospital of Shouguang, Weifang, Shandong 262700, P.R. China
| | - Zhenhong Cui
- Department of Gastroenterology, The Fourth People's Hospital of Jinan, Jinan, Shandong 250031, P.R. China
| |
Collapse
|
13
|
Nokkari A, Abou-El-Hassan H, Mechref Y, Mondello S, Kindy MS, Jaffa AA, Kobeissy F. Implication of the Kallikrein-Kinin system in neurological disorders: Quest for potential biomarkers and mechanisms. Prog Neurobiol 2018; 165-167:26-50. [PMID: 29355711 PMCID: PMC6026079 DOI: 10.1016/j.pneurobio.2018.01.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/15/2018] [Indexed: 01/06/2023]
Abstract
Neurological disorders represent major health concerns in terms of comorbidity and mortality worldwide. Despite a tremendous increase in our understanding of the pathophysiological processes involved in disease progression and prevention, the accumulated knowledge so far resulted in relatively moderate translational benefits in terms of therapeutic interventions and enhanced clinical outcomes. Aiming at specific neural molecular pathways, different strategies have been geared to target the development and progression of such disorders. The kallikrein-kinin system (KKS) is among the most delineated candidate systems due to its ubiquitous roles mediating several of the pathophysiological features of these neurological disorders as well as being implicated in regulating various brain functions. Several experimental KKS models revealed that the inhibition or stimulation of the two receptors of the KKS system (B1R and B2R) can exhibit neuroprotective and/or adverse pathological outcomes. This updated review provides background details of the KKS components and their functions in different neurological disorders including temporal lobe epilepsy, traumatic brain injury, stroke, spinal cord injury, Alzheimer's disease, multiple sclerosis and glioma. Finally, this work will highlight the putative roles of the KKS components as potential neurotherapeutic targets and provide future perspectives on the possibility of translating these findings into potential clinical biomarkers in neurological disease.
Collapse
Affiliation(s)
- Amaly Nokkari
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Hadi Abou-El-Hassan
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Mark S Kindy
- Department of Pharmaceutical Science, College of Pharmacy, University of South Florida, Tampa, FL, USA; James A. Haley VA Medical Center, Tampa, FL, USA
| | - Ayad A Jaffa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon; Department of Medicine, Medical University of South, Charleston, SC, USA.
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon; Center for Neuroproteomics & Biomarkers Research, Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
14
|
Liang D, He X, Wang Z, Li C, Gao B, Wu J, Bai Y. Remote limb ischemic postconditioning promotes motor function recovery in a rat model of ischemic stroke via the up-regulation of endogenous tissue kallikrein. CNS Neurosci Ther 2018; 24:519-527. [PMID: 29399973 PMCID: PMC6489769 DOI: 10.1111/cns.12813] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 12/27/2017] [Accepted: 12/29/2017] [Indexed: 01/16/2023] Open
Abstract
AIMS Remote ischemic conditionings, such as pre- and per-conditioning, are known to provide cardioprotection in animal models of ischemia. However, little is known about the neuroprotection effect of postconditioning after cerebral ischemia. In this study, we aim to evaluate the motor function rescuing effect of remote limb ischemic postconditioning (RIPostC) in a rat model of acute cerebral stroke. METHODS Left middle cerebral artery occlusion (MCAO) was performed to generate the rat model of ischemic stroke, followed by daily RIPostC treatment for maximum 21 days. The motor function after RIPostC was assessed with foot fault test and balance beam test. Local infarct volume was measured through MRI scanning. Neuronal status was evaluated with Nissl's, HE, and MAP2 immunostaining. Lectin immunostaining was performed to evaluate the microvessel density and area. RESULTS Daily RIPostC for more than 21 days promoted motor function recovery and provided long-lasting neuroprotection after MCAO. Reduced infarct volume, rescued neuronal loss, and enhanced microvessel density and size in the injured areas were observed. In addition, the RIPostC effect was associated with the up-regulation of endogenous tissue kallikrein (TK) level in circulating blood and local ischemic brain regions. A TK receptor antagonist HOE-140 partially reversed RIPostC-induced improvements, indicating the specificity of endogenous TK mediating the neuroprotection effect of RIPostC. CONCLUSION Our study demonstrates RIPostC treatment as an effective rehabilitation therapy to provide motor function recovery and alleviate brain impairment in a rat model of acute cerebral ischemia. We also for the first time provide evidence showing that the up-regulation of endogenous TK from remote conditioning regions underlies the observed effects of RIPostC.
Collapse
Affiliation(s)
- Dan Liang
- Department of Rehabilitation MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Xi‐Biao He
- Shanghai University of Medicine & Health SciencesShanghaiChina
| | - Zheng Wang
- Department of NeurologyHuashan HospitalState Key Laboratory of Medical NeurobiologyFudan UniversityShanghaiChina
| | - Ce Li
- Department of Rehabilitation MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Bei‐Yao Gao
- Department of Rehabilitation MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Jun‐Fa Wu
- Department of Rehabilitation MedicineHuashan HospitalFudan UniversityShanghaiChina
| | - Yu‐Long Bai
- Department of Rehabilitation MedicineHuashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
15
|
The Long-Term Outcome Comparison of Different Time-Delayed Kallikrein Treatments in a Mouse Cerebral Ischemic Model. Stem Cells Int 2018; 2018:1706982. [PMID: 29760720 PMCID: PMC5907522 DOI: 10.1155/2018/1706982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 09/23/2017] [Accepted: 10/10/2017] [Indexed: 11/18/2022] Open
Abstract
Delayed administration of kallikrein after cerebral infarction can improve neurological function. However, the appropriate kallkrein treatment time after ischemic stroke has not been illuminated. In this study, we compared the long-term outcome among three kallikrein therapeutic regimens starting at different time points following mouse cerebral ischemia. Furthermore, the protective mechanisms involving neurogenesis, angiogenesis, and AKT-GSK3β-VEGF signaling pathway were analyzed. Human tissue kallikrein was injected through the tail vein daily starting at 8 h, 24 h, or 36 h after right middle cerebral artery occlusion (MCAO) until the 28th day. Three therapeutic regimens all protected against neurological dysfunction, but kallikrein treatment starting at 8 h after MCAO had the best efficacy. Additionally, kallikrein treatment at 8 h after MCAO significantly enhanced cell proliferation including neural stem cell and induced differentiation of neural stem cell into mature neuron. Kallikrein treatment starting at 8 h also promoted more angiogenesis than other two treatment regimens, which was associated with AKT-GSK3β-VEGF signaling pathway. Thus, we confirm that three delayed kallikrein treatments provide protection against cerebral infarction and furthermore suggest that kallikrein treatment starting at 8 h had a better effect than that at 24 h and 36 h. These findings provide the experimental data contributing to better clinical application of exogenous kallikrein.
Collapse
|
16
|
Li MH, Ruan LY, Chen C, Xing YX, Hong W, Du RH, Wang JS. Protective effects of Polygonum multiflorum on ischemic stroke rat model analysed by 1H NMR metabolic profiling. J Pharm Biomed Anal 2018; 155:91-103. [PMID: 29625260 DOI: 10.1016/j.jpba.2018.03.049] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 03/22/2018] [Accepted: 03/25/2018] [Indexed: 01/27/2023]
Abstract
Stroke is the third most common cause of death in most industrialized countries. Polygonum multiflorum (He-Shou-Wu, HSW) is one of the traditional Chinese medicines with multiple pharmacological activities which is widely used in Chinese recipe. This study aims to explore the protective effect of HSW on ischemic stroke rat model and to elucidate the underlying mechanisms. The mortality rate, neurological deficit, cerebral infarct size, histopathology, immunohistochemistry, biochemical parameters, quantitative real-time polymerase chain reaction and western blotting were used to access the treatment effects of HSW on ischemic stroke. Proton nuclear magnetic resonance (1H NMR) based metabolomics analysis disclosed that HSW could relieve stroke rats suffering from the ischemia/reperfusion injury by ameliorating the disturbed energy and amino acids metabolisms, alleviating the oxidative stress from reactive oxygen species and reducing the inflammation. HSW treatment increased levels of cellular antioxidants that scavenged reactive oxygen species during ischemia-reperfusion via the nuclear erythroid 2-related factor 2 signaling pathway, and exert anti-inflammatory effect by decreasing the levels of inflammatory factors such as cyclooxygenase-2, interleukin-1β, interleukin-6 and tumor necrosis factor-α. The integrated metabolomics approach showed its potential in understanding mechanisms of HSW in relieving ischemic stroke. Further study to develop HSW as an effective therapeutic agent to treat ischemic stroke is warranted.
Collapse
Affiliation(s)
- Ming-Hui Li
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Ling-Yu Ruan
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Cheng Chen
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Yue-Xiao Xing
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Wei Hong
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Rong-Hui Du
- State Key Laboratory of Pharmaceutical Biotechnology, School of Medicine, Nanjing University, Nanjing 210093, China
| | - Jun-Song Wang
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China.
| |
Collapse
|
17
|
Zhang Q, Wang J, Zhang C, Liao S, Li P, Xu D, Lv Y, Yang M, Kong L. The components of Huang-Lian-Jie-Du-Decoction act synergistically to exert protective effects in a rat ischemic stroke model. Oncotarget 2018; 7:80872-80887. [PMID: 27779107 PMCID: PMC5348361 DOI: 10.18632/oncotarget.12645] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/28/2016] [Indexed: 11/25/2022] Open
Abstract
Huang-Lian-Jie-Du-Decoction (HLJDD, Oren-gedoku-to in Japanese) is commonly used in traditional Chinese medicine (TCM) to treat ischemic stroke. This study investigated the efficacy of various combinations of the major components of HLJDD, berberine (A), baicalin (B), and jasminoidin (C), on the treatment of ischemic stroke modeled by middle cerebral artery occlusion (MCAO) in rats. The effects of A, B and C individually and their combinations were investigated using proton nuclear magnetic resonance (1H NMR)-based metabolomics complemented with neurologic deficit scoring, infarct volume measurement, biochemistry, histopathology and immunohistochemistry, as well as quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting. Ischemic stroke produces severe oxidative stress, which induces further damage. Our results show that the ABC combination treatment increased levels of cellular antioxidants that scavenged reactive oxygen species during ischemia-reperfusion via the nuclear erythroid 2-related factor 2 (Nrf2) signaling cascade. These protective effects were not observed with the other treatments. These results suggest that a combination of component herbs in HLJDD exhibit stronger effects than the individual herbs alone. Our integrated metabolomics approach also provides a tractable, powerful tool for understanding the science behind TCM formulations.
Collapse
Affiliation(s)
- Qian Zhang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P.R. China
| | - Junsong Wang
- Center for Molecular Metabolism, Nanjing University of Science and Technology, Nanjing, 210094, P.R. China
| | - Chao Zhang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P.R. China
| | - Shanting Liao
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P.R. China
| | - Pei Li
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P.R. China
| | - Dingqiao Xu
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P.R. China
| | - Yan Lv
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P.R. China
| | - Minghua Yang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P.R. China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P.R. China
| |
Collapse
|
18
|
Devetzi M, Goulielmaki M, Khoury N, Spandidos DA, Sotiropoulou G, Christodoulou I, Zoumpourlis V. Genetically‑modified stem cells in treatment of human diseases: Tissue kallikrein (KLK1)‑based targeted therapy (Review). Int J Mol Med 2018; 41:1177-1186. [PMID: 29328364 PMCID: PMC5819898 DOI: 10.3892/ijmm.2018.3361] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/02/2018] [Indexed: 12/12/2022] Open
Abstract
The tissue kallikrein-kinin system (KKS) is an endogenous multiprotein metabolic cascade which is implicated in the homeostasis of the cardiovascular, renal and central nervous system. Human tissue kallikrein (KLK1) is a serine protease, component of the KKS that has been demonstrated to exert pleiotropic beneficial effects in protection from tissue injury through its anti-inflammatory, anti-apoptotic, anti-fibrotic and anti-oxidative actions. Mesenchymal stem cells (MSCs) or endothelial progenitor cells (EPCs) constitute populations of well-characterized, readily obtainable multipotent cells with special immunomodulatory, migratory and paracrine properties rendering them appealing potential therapeutics in experimental animal models of various diseases. Genetic modification enhances their inherent properties. MSCs or EPCs are competent cellular vehicles for drug and/or gene delivery in the targeted treatment of diseases. KLK1 gene delivery using adenoviral vectors or KLK1 protein infusion into injured tissues of animal models has provided particularly encouraging results in attenuating or reversing myocardial, renal and cerebrovascular ischemic phenotype and tissue damage, thus paving the way for the administration of genetically modified MSCs or EPCs with the human tissue KLK1 gene. Engraftment of KLK1-modified MSCs and/or KLK1-modified EPCs resulted in advanced beneficial outcome regarding heart and kidney protection and recovery from ischemic insults. Collectively, findings from pre-clinical studies raise the possibility that tissue KLK1 may be a novel future therapeutic target in the treatment of a wide range of cardiovascular, cerebrovascular and renal disorders.
Collapse
Affiliation(s)
- Marina Devetzi
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Maria Goulielmaki
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Nicolas Khoury
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | | | - Ioannis Christodoulou
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| |
Collapse
|
19
|
Neuroprotection of bradykinin/bradykinin B2 receptor system in cerebral ischemia. Biomed Pharmacother 2017; 94:1057-1063. [DOI: 10.1016/j.biopha.2017.08.042] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 12/17/2022] Open
|
20
|
Zhai K, Kong X, Liu B, Lou J. Bioinformatics analysis of gene expression profiling for identification of potential key genes among ischemic stroke. Medicine (Baltimore) 2017; 96:e7564. [PMID: 28834871 PMCID: PMC5571993 DOI: 10.1097/md.0000000000007564] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
This study aimed to identify the key differentially expressed genes (DEGs) following ischemic stroke (IS).The GSE22255 microarray dataset, which contains samples from peripheral blood mononuclear cells of 20 IS patients and 20 sex- and age-matched controls, was downloaded from the Gene Expression Omnibus. After data pre-processing, DEGs were identified using the Linear Models for Microarray Data package in R. The Search Tool for the Retrieval of Interacting Genes database was used to predict the interactions among the products of DEGs, and then Cytoscape software was used to visualize the protein-protein interaction (PPI) network. DEGs in the PPI network were then analyzed using the Database for Annotation, Visualization, and Integrated Discovery online software to predict their underlying functions through functional and pathway enrichment analyses.A total of 144 DEGs were identified in IS samples compared with control samples, including 75 upregulated and 69 downregulated genes. Genes with higher degrees in the PPI network included FOS (degree = 26), TP53 (degree = 22), JUN (degree = 20), EGR1 (degree = 18), JUNB (degree = 16), and ATF3 (degree = 15), and these genes may function in IS by interacting with each other (e.g., EGR1-JUN, EGR1-TP53, ATF3-FOS, and JUNB-FOS). Functional enrichment analysis indicated that the downregulated TP53 gene was enriched in immune response and protein targeting categories.ATF3 and EGR1 may have an important protective effect on IS, whereas FOS, JUN, and JUNB may be associated with the development of IS. In addition, TP53 may function as an indicator of poor prognosis for IS through its association with the immune response and protein targeting.
Collapse
Affiliation(s)
- Kaihua Zhai
- Department of Internal Neurology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan
| | - Xiangli Kong
- Neurology Department, The First Affiliated Hospital of Xi’an Medical University, Xi’an, Shanxi Province, China
| | - Boyu Liu
- Department of Endocrinology, Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, People's Republic of China
| | - Jiyu Lou
- Department of Internal Neurology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan
| |
Collapse
|
21
|
Luan Y, Ruan Y, Wang T, Zhuan L, Wen Z, Chen R, Zhang Y, Cui K, Yang J, Wang S, Liu J, Ye Z, Wang D. Preserved Erectile Function in the Aged Transgenic Rat Harboring Human Tissue Kallikrein 1. J Sex Med 2017; 13:1311-1322. [PMID: 27555503 DOI: 10.1016/j.jsxm.2016.07.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/07/2016] [Accepted: 07/09/2016] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Human tissue kallikrein 1 (hKLK1) has enormous potential for the protection of vasodilation and endothelial function in the cardiovascular system. Our previous study proved the decreased expression of kallikrein 1 in the corpus cavernosum (CC) of aged rats, but the role of kallikrein 1 in age-related erectile dysfunction remains unknown. AIM To explore the effect and underlying mechanisms of hKLK1 on age-related erectile dysfunction. METHODS Male wild-type Sprague-Dawley rats (WTR) and transgenic rats harboring the hKLK1 gene (TGR) were fed to 4 and 27 months of age, respectively, and divided into four groups: young WTR (yWTR) as the control, young TGR (yTGR), aged WTR (aWTR), and aged TGR (aTGR). Rats' erectile function was evaluated by the cavernous nerve electrostimulation method. Then, CCs were collected for verification of hKLK1 followed by measurement of nitric oxide (NO)-cyclic guanosine monophosphate (cGMP) and RhoA-Rho-kinase (ROCK) signaling activities. Masson trichrome staining and terminal deoxynucleotidyl transferase 2'-deoxyuridine 5'-triphosphate nick end labeling assay were conducted to evaluate penile fibrosis and apoptosis. MAIN OUTCOME MEASURES Erectile response, NO-cGMP and RhoA-ROCK pathway-related indices, ratio of smooth muscle to collagen, and apoptosis index. RESULTS The hKLK1 alleviated the decrease of erectile function in the aWTR group. Endothelial NO synthase (eNOS) and phospho-eNOS(Ser1177) expressions, NO synthase activity, and NO and cGMP levels were decreased, whereas phospho-eNOS(Thr495), L-type Ca(2+) channel, RhoA, ROCK1, ROCK2, and transforming growth factor β1 proteins were increased in the CCs of the aWTR group compared with the control yWTR group. These changes were obviously mitigated in the aTGR group. Moreover, hKLK1 prevented the sharp decrease of the ratio of smooth muscle to collagen and the increase of the apoptosis index in the CCs of the aWTR group. CONCLUSION These results suggest that hKLK1 could play a preventive role in age-related erectile dysfunction by activation of the NO-cGMP pathway and inhibition of the RhoA-ROCK pathway and by antitissue fibrotic and apoptotic effects.
Collapse
Affiliation(s)
- Yang Luan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yajun Ruan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Li Zhuan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zheng Wen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ruibao Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kai Cui
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shaogang Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jihong Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Daowen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
22
|
Yang J, Su J, Wan F, Yang N, Jiang H, Fang M, Xiao H, Wang J, Tang J. Tissue kallikrein protects against ischemic stroke by suppressing TLR4/NF-κB and activating Nrf2 signaling pathway in rats. Exp Ther Med 2017; 14:1163-1170. [PMID: 28810574 DOI: 10.3892/etm.2017.4614] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 03/24/2017] [Indexed: 01/04/2023] Open
Abstract
Brain damage following cerebral ischemia-reperfusion (I/R) is a complicated pathophysiological course, in which inflammation and oxidative stress have been suggested to serve an important role. Toll-like receptor 4 (TLR4) has been suggested to be involved in secondary inflammatory process in cerebral ischemia. Nuclear factor erythroid 2-related factor 2 (Nrf2), an important regulator of the antioxidant host defense, maintains the cellular redox homeostasis. Tissue kallikrein (TK) has been proven to elicit a variety of biological effects in ischemic stroke through its anti-inflammatory and anti-oxidant properties. However, the mechanisms underlying its beneficial effects remain poorly defined. The present study examined the hypothesis that TK attenuates ischemic cerebral injury via the TLR4/nuclear factor-κB (NF-κB) and Nrf2 signaling pathways. Using a transient rat middle cerebral artery occlusion (MCAO) model, the effects of immediate and delayed TK treatment subsequent to reperfusion were investigated. The neurological deficits, infarct size, and the expression of TLR4/NF-κB and Nrf2 pathway in ischemic brain tissues were measured at 24 following MCAO. The results indicated that TK immediate treatment significantly improved neurological deficits and reduced the infarct size, accompanied by the inhibition of TLR4 and NF-κB levels, and the activation of Nrf2 pathway. Furthermore, TK delayed treatment also exerted neuroprotection against I/R injury. However, the neuroprotective effect of TK immediate treatment was better compared with that of TK delayed treatment. In conclusion, the results indicated that TK protected the brain against ischemic injury in rats after MCAO through its anti-oxidative and anti-inflammatory effects. Suppression of TLR4/NF-κB and activation of the Nrf2 pathway contributed to the neuroprotective effects induced by TK in cerebral ischemia. Therefore, TK may provide an effective intervention with a wider therapeutic window for ischemic stroke.
Collapse
Affiliation(s)
- Jiawei Yang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China.,Department of Neurology, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jianhua Su
- Department of Neurology, The Affiliated Jintan Hospital of Medical College of Jiangsu University, Jintan, Jiangsu 213200, P.R. China
| | - Fen Wan
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Nan Yang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Haibo Jiang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Mingming Fang
- Department of Neurology, Jiangsu Hospital of Chinese Traditional and Western Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Hang Xiao
- Department of Neurotoxicology, Nanjing Medical University, Nanjing, Jiangsu 211199, P.R. China
| | - Jun Wang
- Department of Neurotoxicology, Nanjing Medical University, Nanjing, Jiangsu 211199, P.R. China
| | - Jinrong Tang
- Department of Neurology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
23
|
Zhang Q, Wang J, Liao S, Li P, Xu D, Lv Y, Yang M, Kong L. Optimization of Huang-Lian-Jie-Du-Decoction for Ischemic Stroke Treatment and Mechanistic Study by Metabolomic Profiling and Network Analysis. Front Pharmacol 2017; 8:165. [PMID: 28400733 PMCID: PMC5368223 DOI: 10.3389/fphar.2017.00165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 03/13/2017] [Indexed: 11/29/2022] Open
Abstract
Optimal drug proportions and mechanism deciphering of multicomponent drugs are critical for developing novel therapies to cope with complex diseases, such as stroke. In the present study, orthogonal experimental design was applied to explore the optimal proportion of the four component herbs in Huang-Lian-Jie-Du-Decoction (HLJDD) on the treatment of ischemic stroke. The treatment efficacies and mechanisms were assessed using global and amino acids (AAs) targeted metabolomics, as well as correlation network analysis. The global NMR metabolomics results revealed that AAs metabolism was significantly perturbed in middle cerebral artery occlusion rats. The levels of 23 endogenous AAs were then subjected to HPLC-QTOF-MS/MS analysis. These results complemented with neurobehavioral evaluations, cerebral infarct assessments, biochemical evaluations, histological inspections and immunohistochemistry observations strongly demonstrated that HLJDD with optimal proportion of 6 (Rhizoma coptidis): 4 (Radix scutellariae): 1 (Cortex phellodendr): 3 (Fructus Gardeniae) had the best efficacy on ischemic stroke, which could be ascribed to its modulation on AA metabolism. This integrated metabolomics approach showed the potential and applicable in deciphering the complex mechanisms of traditional Chinese medicine formulae on the treatment of complicated diseases, which provided new means to assess the treatment effects of herb combinations and to further development of drugs or therapies based on these formulae.
Collapse
Affiliation(s)
- Qian Zhang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University Nanjing, China
| | - Junsong Wang
- Center for Molecular Metabolism, Nanjing University of Science and Technology Nanjing, China
| | - Shanting Liao
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University Nanjing, China
| | - Pei Li
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University Nanjing, China
| | - Dingqiao Xu
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University Nanjing, China
| | - Yan Lv
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University Nanjing, China
| | - Minghua Yang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University Nanjing, China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University Nanjing, China
| |
Collapse
|
24
|
Dong TF, Lv HX, Niu XL, Gui YK, Zhang P, Yan HQ, Li T. Effect of Urinary Kallidinogenase on Transforming Growth Factor-β1 and High-Sensitivity C-Reactive Protein Expression in Rat Focal Cerebral Ischemic Injury. Med Sci Monit 2016; 22:2852-8. [PMID: 27521289 PMCID: PMC4988362 DOI: 10.12659/msm.892724] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background In this study we investigated the effect of urinary kallidinogenase (UK) on transforming growth factor beta 1 (TGF-β1) expression in brain tissue. We also explored the neuroprotective mechanism of UK against ischemic injury by measuring serum high-sensitivity C-reactive protein (hs-CRP) level changes after rat cerebral ischemic injury. Material/Methods The rat middle cerebral artery ischemia/reperfusion model was established using the suture method. Sprague-Dawley rats were randomly divided into 3 groups: treatment, Gegen control, and blank control. Each group was subsequently divided into 5 subgroups according to time (6, 12, 24, 48, and 72 h). Rats in the treatment group were administered UK as treatment. TGF-β1 expression was observed at each time point using SABC and immunohistochemical staining methods to estimate cerebral infarct volume percentage. Serum hs-CRP levels were also measured. Results TGF-β1 protein expression in ischemic brain tissues of the treatment group significantly increased at each time point (P<0.01) compared with both control groups. Treatment group serum hs-CRP levels significantly decreased at each time point (P<0.05) compared with both control groups. Conclusions UK exerts a neuroprotective effect by upregulating TGF-β1 expression and inhibiting excessive inflammatory responses.
Collapse
Affiliation(s)
- Ting-Fang Dong
- Department of Pharmacy, The First Afiliated Hospital of Xinxiang Medical College, Weihui, Henan, China (mainland)
| | - Hai-Xia Lv
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China (mainland)
| | - Xiao-Lu Niu
- Department of Neurology, The First Afiliated Hospital of Xinxiang Medical College, Weihui, Henan, China (mainland)
| | - Yong-Kun Gui
- Department of Neurology, The First Afiliated Hospital of Xinxiang Medical College, Weihui, Henan, China (mainland)
| | - Ping Zhang
- Department of Neurology, The First Afiliated Hospital of Xinxiang Medical College, Weihui, Henan, China (mainland)
| | - Hai-Qing Yan
- Department of Neurology, The First Afiliated Hospital of Xinxiang Medical College, Weihui, Henan, China (mainland)
| | - Tong Li
- Department of Neurology, The First Afiliated Hospital of Xinxiang Medical College, Weihui, Henan, China (mainland)
| |
Collapse
|
25
|
Yunchang M, Qinxue D, Binbin J, Xin H, Lili Y, Linbi C, Wujun G, Pengbo Z, Junlu W. Human tissue kallikrein ameliorates cerebral vasospasm in a rabbit model of subarachnoid hemorrhage. Neurol Res 2016; 37:1082-9. [PMID: 26923578 DOI: 10.1080/01616412.2015.1110305] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVES Cerebral vasospasm (CVS) and early brain injury are major causes of morbidity and mortality following subarachnoid hemorrhage (SAH). We investigated the efficiency of human tissue kallikrein (HTK) to prevent CVS in a rabbit model of SAH. METHODS Forty-eight Japanese white rabbits were randomly divided into four groups (n = 12 each): control (sham-operated), SAH, SAH + phosphate-buffered saline (PBS, vehicle), and SAH + HTK. Basilar artery (BA) diameters were measured by three-dimensional computed tomography angiography at three time points. Endothelin-1 (ET-1) and nitric oxide (NO) levels in the cerebrospinal fluid (CSF) were assayed 24 h before and 5 and 7 days after SAH. After the last measurement, the animals were killed, and endothelial cell apoptosis was assessed. Bax and Bcl-2 levels in the BA were measured by western blotting. RESULTS HTK was found to significantly reduce CVS following SAH in rabbits. Inverse changes were observed in ET-1 and NO levels in the CSF collected from the SAH group. HTK increased levels of NO, which has a vasodilatory effect, but did not affect levels of ET-1, which has a vasoconstrictive effect. CTA revealed that HTK treatment significantly increased BA diameter. Moreover, HTK treatment reduced the number of apoptotic cells following SAH, presumably by increasing and decreasing Bcl-2 and Bax expression, respectively. CONCLUSION HTK ameliorated CVS and inhibited apoptosis in the BA in a rabbit model of SAH.
Collapse
Affiliation(s)
- Mo Yunchang
- 1 Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University , Xi'an, Shanxi 710004, China
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Sang H, Liu L, Wang L, Qiu Z, Li M, Yu L, Zhang H, Shi R, Yu S, Guo R, Ye R, Liu X, Zhang R. Opposite roles of bradykinin B1 and B2 receptors during cerebral ischaemia-reperfusion injury in experimental diabetic rats. Eur J Neurosci 2016; 43:53-65. [PMID: 26565562 DOI: 10.1111/ejn.13133] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 10/29/2015] [Accepted: 11/06/2015] [Indexed: 02/05/2023]
Affiliation(s)
- Hongfei Sang
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Ling Liu
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Liumin Wang
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Zhongming Qiu
- Department of Neurology; The 117th Hospital of PLA; Xihu District Hangzhou Zhejiang Province China
| | - Min Li
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Linjie Yu
- Nanjing University School of Medicine; Nanjing China
| | - Hao Zhang
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Ruifeng Shi
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Shuhong Yu
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Ruibing Guo
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Ruidong Ye
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Xinfeng Liu
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| | - Renliang Zhang
- Department of Neurology; Jinling Hospital; Medical School of Nanjing University; 305 East Zhongshan Road Nanjing 210002 Jiangsu Province China
| |
Collapse
|
27
|
Tissue Kallikrein Activity, Detected by a Novel Method, May Be a Predictor of Recurrent Stroke: A Case-Control Study. DISEASE MARKERS 2015; 2015:159750. [PMID: 26451066 PMCID: PMC4584216 DOI: 10.1155/2015/159750] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 08/24/2015] [Accepted: 08/30/2015] [Indexed: 11/21/2022]
Abstract
Aim. Tissue kallikrein (TK) protein content in plasma has been shown to be negatively associated with both incident and recurrent strokes. The aims of this study were to develop a novel method for detecting TK activity and to investigate its association with event-free survival over 5 years in Chinese first-ever stroke patients. Methods. We designed a case-control study with 321 stroke patients (174: ischemic stroke, 147: hemorrhagic stroke) and 323 healthy local controls. TK activity was measured by a novel assay utilizing the immunological characteristics of TK and the catalysis of benzoyl arginine ethyl ester hydrochloride (BAEE). Results. TK protein levels above 0.200 mg/L in plasma were not associated with urinary TK activity or the risk of stroke recurrence. TK activity was significantly lower in stroke patients compared with controls (1.583 ± 0.673 Eu/mL versus 1.934 ± 0.284 Eu/mL, P < 0.001). After adjusting for traditional risk factors, TK activity was negatively associated, in a dose-response manner, with the risk of overall stroke recurrence and positively associated with event-free survival during a 5-year follow-up (relative risk (RR), 0.69; 95% CI, 0.57–0.84; P < 0.001). Conclusions. Our findings suggest that urinary TK activity may be a stronger predictor of stroke recurrence than plasma TK levels.
Collapse
|
28
|
Han L, Li J, Chen Y, Zhang M, Qian L, Chen Y, Wu Z, Xu Y, Li J. Human Urinary Kallidinogenase Promotes Angiogenesis and Cerebral Perfusion in Experimental Stroke. PLoS One 2015. [PMID: 26222055 PMCID: PMC4519127 DOI: 10.1371/journal.pone.0134543] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Angiogenesisis a key restorative mechanism in response to ischemia, and pro-angiogenic therapy could be beneficial in stroke. Accumulating experimental and clinical evidence suggest that human urinary kallidinogenase (HUK) improves stroke outcome, but the underlying mechanisms are not clear. The aim of current study was to verify roles of HUK in post-ischemic angiogenesis and identify relevant mediators. In rat middle cerebral artery occlusion (MCAO) model, we confirmed that HUK treatment could improve stroke outcome, indicated by reduced infarct size and improved neurological function. Notably, the 18F-FDG micro-PET scan indicated that HUK enhanced cerebral perfusion in rats after MCAO treatment. In addition, HUK promotespost-ischemic angiogenesis, with increased vessel density as well as up-regulated VEGF andapelin/APJ expression in HUK-treated MCAO mice. In endothelial cell cultures, induction of VEGF and apelin/APJ expression, and ERK1/2 phosphorylation by HUK was further confirmed. These changes were abrogated by U0126, a selective ERK1/2 inhibitor. Moreover, F13A, a competitive antagonist of APJ receptor, significantly suppressed HUK-induced VEGF expression. Furthermore, angiogenic functions of HUK were inhibited in the presence of selective bradykinin B1 or B2 receptor antagonist both in vitro and in vivo. Our findings indicate that HUK treatment promotes post-ischemic angiogenesis and cerebral perfusion via activation of bradykinin B1 and B2 receptors, which is potentially due to enhancement expression of VEGF and apelin/APJ in ERK1/2 dependent way.
Collapse
Affiliation(s)
- Lijuan Han
- Departments of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Jie Li
- Departments of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Department of Neurology, Affiliated Yixing People's Hospital of Jiangsu University, Yixing, China
| | - Yanting Chen
- Departments of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Meijuan Zhang
- Departments of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Lai Qian
- Departments of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Yan Chen
- Departments of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Zhengzheng Wu
- Departments of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Yun Xu
- Departments of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
- * E-mail: (YX); (JL)
| | - Jingwei Li
- Departments of Neurology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
- * E-mail: (YX); (JL)
| |
Collapse
|
29
|
Bradykinin-induced inhibition of proliferation rate during neurosphere differentiation: Consequence or cause of neuronal enrichment? Cytometry A 2015; 87:929-35. [DOI: 10.1002/cyto.a.22705] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 04/10/2015] [Accepted: 05/21/2015] [Indexed: 12/26/2022]
|
30
|
Negraes PD, Trujillo CA, Pillat MM, Teng YD, Ulrich H. Roles of kinins in the nervous system. Cell Transplant 2015; 24:613-23. [PMID: 25839228 DOI: 10.3727/096368915x687778] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The kallikrein-kinin system (KKS) is an endogenous pathway involved in many biological processes. Although primarily related to blood pressure control and inflammation, its activation goes beyond these effects. Neurogenesis and neuroprotection might be stimulated by bradykinin being of great interest for clinical applications following brain injury. This peptide is also an important player in spinal cord injury pathophysiology and recovery, in which bradykinin receptor blockers represent substantial therapeutic potential. Here, we highlight the participation of kinin receptors and especially bradykinin in mediating ischemia pathophysiology in the central and peripheral nervous systems. Moreover, we explore the recent advances on mechanistic and therapeutic targets for biological, pathological, and neural repair processes involving kinins.
Collapse
Affiliation(s)
- Priscilla D Negraes
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
31
|
Chao J, Bledsoe G, Chao L. Kallikrein-kinin in stem cell therapy. World J Stem Cells 2014; 6:448-457. [PMID: 25258666 PMCID: PMC4172673 DOI: 10.4252/wjsc.v6.i4.448] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 08/27/2014] [Accepted: 09/01/2014] [Indexed: 02/06/2023] Open
Abstract
The tissue kallikrein-kinin system exerts a wide spectrum of biological activities in the cardiovascular, renal and central nervous systems. Tissue kallikrein-kinin modulates the proliferation, viability, mobility and functional activity of certain stem cell populations, namely mesenchymal stem cells (MSCs), endothelial progenitor cells (EPCs), mononuclear cell subsets and neural stem cells. Stimulation of these stem cells by tissue kallikrein-kinin may lead to protection against renal, cardiovascular and neural damage by inhibiting apoptosis, inflammation, fibrosis and oxidative stress and promoting neovascularization. Moreover, MSCs and EPCs genetically modified with tissue kallikrein are resistant to hypoxia- and oxidative stress-induced apoptosis, and offer enhanced protective actions in animal models of heart and kidney injury and hindlimb ischemia. In addition, activation of the plasma kallikrein-kinin system promotes EPC recruitment to the inflamed synovium of arthritic rats. Conversely, cleaved high molecular weight kininogen, a product of plasma kallikrein, reduces the viability and vasculogenic activity of EPCs. Therefore, kallikrein-kinin provides a new approach in enhancing the efficacy of stem cell therapy for human diseases.
Collapse
|
32
|
Abstract
Objective:To promote understandings about the pathogenesis of ischemic stroke (IS) through mining key genes, functions and pathways with microarray technology.Methods:Differentially expressed genes (DEGs) in blood between patients with IS and healthy people were screened out through comparing microarray data obtained from Gene Expression Omnibus. Overrepresented functions in DEGs were revealed by Gene Ontology (GO) enrichment analysis. Interaction network was constructed for the top 24 DEGs with information from Human Protein Reference Database (HPRD). Relevant microRNAs (miRNAs) were retrieved from three databases: TargetScan, miRBase and miRanda.Results:A total of 503 DEGs were obtained. Functional enrichment analysis showed that immune response, signaling pathways and apoptosis were significantly over-represented. Six key genes with big degree, betweenness and clustering coefficient were then revealed, which might play important roles in the development of IS. In addition, 57 differentially expressed miRNAs targeting the 6 genes were retrieved.Conclusions:Our study provides insights into the pathogenesis of IS and potential targets to treat the disease.
Collapse
|
33
|
Tissue kallikrein (kallidinogenase) protects against retinal ischemic damage in mice. Eur J Pharmacol 2014; 738:74-82. [DOI: 10.1016/j.ejphar.2014.05.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 05/13/2014] [Accepted: 05/14/2014] [Indexed: 12/21/2022]
|
34
|
Dobrivojević M, Špiranec K, Sinđić A. Involvement of bradykinin in brain edema development after ischemic stroke. Pflugers Arch 2014; 467:201-12. [DOI: 10.1007/s00424-014-1519-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 04/07/2014] [Accepted: 04/09/2014] [Indexed: 01/04/2023]
|
35
|
Pabon M, Tamboli C, Tamboli S, Acosta S, De La Pena I, Sanberg PR, Tajiri N, Kaneko Y, Borlongan CV. ESTROGEN REPLACEMENT THERAPY FOR STROKE. CELL MEDICINE 2014; 6:111-122. [PMID: 24999442 DOI: 10.3727/215517913x672263] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Stroke is the third most common cause of death and severe disability among Western populations. Overall, the incidence of stroke is uniformly higher in men than in women. Stroke is rare in women during the reproductive years, and rapidly increases after menopause, strongly suggesting that estrogen (E2) plays an important role in the prevention of stroke. Ongoing studies are currently evaluating both the benefits and risks associated with E2 replacement therapy and hormone replacement therapy in stroke. Equally important is the role of E2 receptor (ER), as studies indicate that ER populations in several tissue sites may significantly change during stress and aging. Such changes may affect the patient's susceptibility to neurological disorders including stroke, and greatly affect the response to selective E2 receptor modulators (SERMs). Replacement therapies may be inefficient with low ER levels. The goal of this review paper is to discuss an animal model that will allow investigations of the potential therapeutic effects of E2 and its derivatives in stroke. We hypothesize that E2 neuroprotection is, in part, receptor mediated. This hypothesis is a proof of principle approach to demonstrate a role for specific ER subtypes in E2 neuroprotection. To accomplish this, we use a retroviral mediated gene transfer strategy that express subtypes of the ER gene in regions of the rat brain most susceptible to neuronal damage, namely the striatum and cortex. The animal model is exposed to experimental stroke conditions involving middle cerebral artery occlusion (MCAo) method, and eventually the extent of neuronal damage will be evaluated. A reduction in neuronal damage is expected when E2 is administered with specific ER subtypes. From this animal model, an optimal E2 dose and treatment regimen can be determined. The animal model can help identify potential E2-like therapeutics in stroke, and screen for beneficial or toxic additives present in commercial E2 preparations that are currently available. Such studies will be informative in designing drug therapies for stroke.
Collapse
Affiliation(s)
- Mibel Pabon
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Cyrus Tamboli
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Sarosh Tamboli
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Sandra Acosta
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Ike De La Pena
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Paul R Sanberg
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Naoki Tajiri
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Yuji Kaneko
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL USA
| |
Collapse
|
36
|
Chao J, Bledsoe G, Chao L. Tissue kallikrein-kinin therapy in hypertension and organ damage. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2014; 69:37-57. [PMID: 25130039 DOI: 10.1007/978-3-319-06683-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Tissue kallikrein is a serine proteinase that cleaves low molecular weight kininogen to produce kinin peptides, which in turn activate kinin receptors to trigger multiple biological functions. In addition to its kinin-releasing activity, tissue kallikrein directly interacts with the kinin B2 receptor, protease-activated receptor-1, and gamma-epithelial Na channel. The tissue kallikrein-kinin system (KKS) elicits a wide spectrum of biological activities, including reducing hypertension, cardiac and renal damage, restenosis, ischemic stroke, and skin wound injury. Both loss-of-function and gain-of-function studies have shown that the KKS plays an important endogenous role in the protection against health pathologies. Tissue kallikrein/kinin treatment attenuates cardiovascular, renal, and brain injury by inhibiting oxidative stress, apoptosis, inflammation, hypertrophy, and fibrosis and promoting angiogenesis and neurogenesis. Approaches that augment tissue kallikrein-kinin activity might provide an effective strategy for the treatment of hypertension and associated organ damage.
Collapse
|
37
|
Lan W, Yang F, Liu L, Yin Q, Li M, Li Z, Sang H, Xu G, Ma M, Zhang Z, Liu Z, Liu X, Zhang R. Tissue kallikrein preventing the restenosis after stenting of symptomatic MCA atherosclerotic stenosis (KPRASS). Int J Stroke 2013; 9:533-5. [PMID: 24354519 DOI: 10.1111/ijs.12229] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 10/10/2013] [Indexed: 11/28/2022]
Abstract
RATIONALE Many recent studies suggest that the kallikrein-kinin system play a protective role in the impairment of vascular smooth muscle cells and vascular endothelial cell. AIMS The study aims to determine whether tissue kallikrein is efficacy for preventing the long-term in-stent restenosis after stenting of symptomatic atherosclerotic stenosis of the middle cerebral artery M1 segment. DESIGN This is a Phase II, randomized, single-blinded, controlled trial. In line with SAMMPRIS stenting indications, patients (n = 90) with the symptomatic the middle cerebral artery M1 segment stenosis ≥ 70% and successfully treated with stent will be enrolled. Eligible patients will be randomized using computer generated numbers, and allocated to receive tissue kallikrein treatment or not. Patients in tissue kallikrein treatment group will be prescribed with intravenous infusion of tissue kallikrein (0.15 PNAU/d, dissolved in 100 ml saline) for 7 days after stenting and then oral administration of pancreatic kallikrein enteric-coated tablet (240 U, 3/d) to the end of study. As the foundation treatment, all the enrolled patients will receive aspirin (100 mg/d), clopidogrel (75 mg/d), and atorvastatin (20 mg/d) for the first 6 months and continue with the combination of aspirin and atorvastatin at the previous dosage. STUDY OUTCOMES Patients will be evaluated at 1, 6 and 12 months after stenting. The primary outcomes are the in-stent restenosis rate, new stroke or aggravation of the previous ischemic stroke ipsilateral to the severe stenotic artery. Secondary outcomes include stroke of other artery territories, myocardial infarction and vascular death. Modification of stroke knowledge, exercise and diet habit, smoking cessation and available laboratory data will also be recorded. CONCLUSION As our pilot study, tissue kallikrein would be expected to prevent the long-term in-stent restenosis after stenting of the symptomatic middle cerebral artery dramatically.
Collapse
Affiliation(s)
- Wenya Lan
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Li Y, Raman I, Du Y, Yan M, Min S, Yang J, Fang X, Li W, Lu J, Zhou XJ, Mohan C, Li QZ. Kallikrein transduced mesenchymal stem cells protect against anti-GBM disease and lupus nephritis by ameliorating inflammation and oxidative stress. PLoS One 2013; 8:e67790. [PMID: 23935844 PMCID: PMC3720854 DOI: 10.1371/journal.pone.0067790] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 05/26/2013] [Indexed: 12/20/2022] Open
Abstract
Previously we have shown that kallikreins (klks) play a renoprotective role in nephrotoxic serum induced nephritis. In this study, we have used mesenchymal stem cells (MSCs) as vehicles to deliver klks into the injured kidneys and have measured their therapeutic effect on experimental antibody induced nephritis and lupus nephritis. Human KLK-1 (hKLK1) gene was transduced into murine MSCs using a retroviral vector to generate a stable cell line, hKLK1-MSC, expressing high levels of hKLK1. 129/svj mice subjected to anti-GBM induced nephritis were transplanted with 10(6) hKLK1-MSCs and hKLK1 expression was confirmed in the kidneys. Compared with vector-MSCs injected mice, the hKLK1-MSCs treated mice showed significantly reduced proteinuria, blood urea nitrogen (BUN) and ameliorated renal pathology. Using the same strategy, we treated lupus-prone B6.Sle1.Sle3 bicongenic mice with hKLK1-MSCs and demonstrated that hKLK1-MSCs delivery also attenuated lupus nephritis. Mechanistically, hKLK1-MSCs reduced macrophage and T-lymphocyte infiltration into the kidney by suppressing the expression of inflammation cytokines. Moreover, hKLK1 transduced MSCs were more resistant to oxidative stress-induced apoptosis. These findings advance genetically modified MSCs as potential gene delivery tools for targeting therapeutic agents to the kidneys in order to modulate inflammation and oxidative stress in lupus nephritis.
Collapse
Affiliation(s)
- Yajuan Li
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Indu Raman
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Yong Du
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Mei Yan
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Soyoun Min
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Jichen Yang
- Quantitative Biomedical Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Xiangdong Fang
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Wei Li
- Key Laboratory of Medical Genetics, Wenzhou Medical College School of Laboratory Medicine & Life Science, Wenzhou, China
| | - Jianxin Lu
- Key Laboratory of Medical Genetics, Wenzhou Medical College School of Laboratory Medicine & Life Science, Wenzhou, China
| | - Xin J. Zhou
- Renal Path Diagnostics, Pathologist BioMedical Laboratories, Lewisville, Texas, United States of America
| | - Chandra Mohan
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- BME Departments, University of Houston, Houston, Texas, United States of America
| | - Quan-Zhen Li
- Department of Immunology and Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Key Laboratory of Medical Genetics, Wenzhou Medical College School of Laboratory Medicine & Life Science, Wenzhou, China
| |
Collapse
|
39
|
Torres-Rivera W, Pérez D, Park KY, Carrasco M, Platt MO, Eterović VA, Ferchmin PA, Ulrich H, Martins AH. Kinin-B2 receptor exerted neuroprotection after diisopropylfluorophosphate-induced neuronal damage. Neuroscience 2013; 247:273-9. [PMID: 23735753 DOI: 10.1016/j.neuroscience.2013.05.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 05/23/2013] [Accepted: 05/24/2013] [Indexed: 02/06/2023]
Abstract
The kinin-B2 receptor (B2BKR) activated by its endogenous ligand bradykinin participates in various metabolic processes including the control of arterial pressure and inflammation. Recently, functions for this receptor in brain development and protection against glutamate-provoked excitotoxicity have been proposed. Here, we report neuroprotective properties for bradykinin against organophosphate poisoning using acute hippocampal slices as an in vitro model. Following slice perfusion for 10min with diisopropylfluorophosphate (DFP) to initiate the noxious stimulus, responses of pyramidal neurons upon an electric impulse were reduced to less than 30% of control amplitudes. Effects on synaptic-elicited population spikes were reverted when preparations had been exposed to bradykinin 30min after challenging with DFP. Accordingly, bradykinin-induced population spike recovery was abolished by HOE-140, a B2BKR antagonist. However, the kinin-B1 receptor (B1BKR) agonist Lys-des-Arg(9)-bradykinin, inducing the phosphorylation of mitogen-activated protein kinase (MEK/MAPK) and cell death, abolished bradykinin-mediated neuroprotection, an effect, which was reverted by the ERK inhibitor PD98059. In agreement with pivotal B1BKR functions in this process, antagonism of endogenous B1BKR activity alone was enough for restoring population spike activity. On the other hand pralidoxime, an oxime, reactivating acetylcholinesterase (AChE) after organophosphate poisoning, induced population spike recovery after DFP exposure in the presence of bradykinin and Lys-des-Arg(9)-bradykinin. Lys-des-Arg(9)-bradykinin did not revert protection exerted by pralidoxime, however when instead bradykinin and Ly-des-Arg(9)-bradykinin were superfused together, recovery of population spikes diminished. These findings again confirm the neuroprotective feature of bradykinin, which is, diminished by its endogenous metabolites, stimulating the B1BKR, providing a novel understanding of the physiological roles of these receptors.
Collapse
Affiliation(s)
- W Torres-Rivera
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR 00956, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Shao X, Yang R, Yan M, Li Y, Du Y, Raman I, Zhang B, Wakeland EK, Igarashi P, Mohan C, Li QZ. Inducible expression of kallikrein in renal tubular cells protects mice against spontaneous lupus nephritis. ARTHRITIS AND RHEUMATISM 2013; 65:780-791. [PMID: 23280471 DOI: 10.1002/art.37798] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 11/08/2012] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To ascertain whether engineered expression of kallikreins within the kidneys, using an inducible Cre/loxP system, can ameliorate murine lupus nephritis. METHODS In mice with a lupus-prone genetic background, we engineered the expression of tamoxifen-inducible Cre recombinase under the control of a kidney-specific promoter whose activation initiates murine kallikrein-1 expression within the kidneys. These transgenic mice were injected with either tamoxifen or vehicle at age 2 months and then were monitored for 8 months for kallikrein expression and disease. RESULTS Elevated expression of kallikrein was detected in the kidney and urine of tamoxifen-injected mice but not in controls. At age 10 months, all vehicle-injected mice developed severe lupus nephritis, as evidenced by increased proteinuria (mean ± SD 13.43 ± 5.65 mg/24 hours), increased blood urea nitrogen (BUN) and serum creatinine levels (39.86 ± 13.45 mg/dl and 15.23 ± 6.89 mg/dl, respectively), and severe renal pathology. In contrast, the tamoxifen-injected mice showed significantly reduced proteinuria (6.6 ± 4.12 mg/24 hours), decreased BUN and serum creatinine levels (15.71 ± 8.17 mg/dl and 6.64 ± 3.39 mg/dl, respectively), and milder renal pathology. Tamoxifen-induced up-regulation of renal kallikrein expression increased nitric oxide production and dampened renal superoxide production and inflammatory cell infiltration, alluding to some of the pathways through which kallikreins may be operating within the kidneys. CONCLUSION Local expression of kallikreins within the kidney has the capacity to dampen lupus nephritis, possibly by modulating inflammation and oxidative stress.
Collapse
Affiliation(s)
- Xinli Shao
- University of Texas-Southwestern Medical Center, Dallas, TX 75235, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Rhim T, Lee DY, Lee M. Drug delivery systems for the treatment of ischemic stroke. Pharm Res 2013; 30:2429-44. [PMID: 23307348 DOI: 10.1007/s11095-012-0959-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 12/07/2012] [Indexed: 12/13/2022]
Abstract
Stroke is the third leading cause of death in the United States. Reduced cerebral blood flow causes acute damage to the brain due to excitotoxicity, reactive oxygen species (ROS), and ischemia. Currently, the main treatment for stroke is to revive the blood flow by using thrombolytic agents. Reviving blood flow also causes ischemia-reperfusion (I/R) damage. I/R damage results from inflammation and apoptosis and can persist for days to weeks, increasing the infarct size. Drugs can be applied to stroke to intervene in the sub-acute and chronic phases. Chemical, peptide, and genetic therapies have been evaluated to reduce delayed damage to the brain. These drugs have different characteristics, requiring that delivery carriers be developed based on these characteristics. The delivery route is another important factor affecting the efficiency of drug delivery. Various delivery routes have been developed, such as intravenous injection, intranasal administration, and local direct injection to overcome the blood-brain-barrier (BBB). In this review, the delivery carriers and delivery routes for peptide and gene therapies are discussed and examples are provided. Combined with new drugs, drug delivery systems will eventually provide useful treatments for ischemic stroke.
Collapse
Affiliation(s)
- Taiyoun Rhim
- Department of Bioengineering, College of Engineering, Hanyang University, 17 Haengdang-dong, Seongdong-gu, Seoul, 133-791, Republic of Korea
| | | | | |
Collapse
|
42
|
Albert-Weißenberger C, Sirén AL, Kleinschnitz C. Ischemic stroke and traumatic brain injury: the role of the kallikrein-kinin system. Prog Neurobiol 2012; 101-102:65-82. [PMID: 23274649 DOI: 10.1016/j.pneurobio.2012.11.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 11/15/2012] [Accepted: 11/20/2012] [Indexed: 12/16/2022]
Abstract
Acute ischemic stroke and traumatic brain injury are a major cause of mortality and morbidity. Due to the paucity of therapies, there is a pressing clinical demand for new treatment options. Successful therapeutic strategies for these conditions must target multiple pathophysiological mechanisms occurring at different stages of brain injury. In this respect, the kallikrein-kinin system is an ideal target linking key pathological hallmarks of ischemic and traumatic brain damage such as edema formation, inflammation, and thrombosis. In particular, the kinin receptors, plasma kallikrein, and coagulation factor XIIa are highly attractive candidates for pharmacological development, as kinin receptor antagonists or inhibitors of plasma kallikrein and coagulation factor XIIa are neuroprotective in animal models of stroke and traumatic brain injury. Nevertheless, conflicting preclinical evaluation as well as limited and inconclusive data from clinical trials suggest caution when transferring observations made in animals into the human situation. This review summarizes current evidence on the pathological significance of the kallikrein-kinin system during ischemic and traumatic brain damage, with a particular focus on experimental data derived from animal models. Experimental findings are also compared with human data if available, and potential therapeutic implications are discussed.
Collapse
|
43
|
Trujillo CA, Negraes PD, Schwindt TT, Lameu C, Carromeu C, Muotri AR, Pesquero JB, Cerqueira DM, Pillat MM, de Souza HDN, Turaça LT, Abreu JG, Ulrich H. Kinin-B2 receptor activity determines the differentiation fate of neural stem cells. J Biol Chem 2012; 287:44046-61. [PMID: 23132855 DOI: 10.1074/jbc.m112.407197] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Bradykinin is not only important for inflammation and blood pressure regulation, but also involved in neuromodulation and neuroprotection. Here we describe novel functions for bradykinin and the kinin-B2 receptor (B2BkR) in differentiation of neural stem cells. In the presence of the B2BkR antagonist HOE-140 during rat neurosphere differentiation, neuron-specific β3-tubulin and enolase expression was reduced together with an increase in glial protein expression, indicating that bradykinin-induced receptor activity contributes to neurogenesis. In agreement, HOE-140 affected in the same way expression levels of neural markers during neural differentiation of murine P19 and human iPS cells. Kinin-B1 receptor agonists and antagonists did not affect expression levels of neural markers, suggesting that bradykinin-mediated effects are exclusively mediated via B2BkR. Neurogenesis was augmented by bradykinin in the middle and late stages of the differentiation process. Chronic treatment with HOE-140 diminished eNOS and nNOS as well as M1-M4 muscarinic receptor expression and also affected purinergic receptor expression and activity. Neurogenesis, gliogenesis, and neural migration were altered during differentiation of neurospheres isolated from B2BkR knock-out mice. Whole mount in situ hybridization revealed the presence of B2BkR mRNA throughout the nervous system in mouse embryos, and less β3-tubulin and more glial proteins were expressed in developing and adult B2BkR knock-out mice brains. As a underlying transcriptional mechanism for neural fate determination, HOE-140 induced up-regulation of Notch1 and Stat3 gene expression. Because pharmacological treatments did not affect cell viability and proliferation, we conclude that bradykinin-induced signaling provides a switch for neural fate determination and specification of neurotransmitter receptor expression.
Collapse
Affiliation(s)
- Cleber A Trujillo
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil 05508-000
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Lu RY, Luo DF, Xiao SH, Yang LH, Zhao J, Ji EN, Tao EX, Xing YG, Zhu FY, Luan P, Liu J. Kallikrein gene transfer induces angiogenesis and further improves regional cerebral blood flow in the early period after cerebral ischemia/reperfusion in rats. CNS Neurosci Ther 2012; 18:395-9. [PMID: 22533724 DOI: 10.1111/j.1755-5949.2012.00305.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
AIMS The aims of this study were to find out whether kallikrein could induce angiogenesis and affect the cerebral blood flow (rCBF) in the early period after cerebral ischemia/reperfusion (CI/R). METHODS The adenovirus carried human tissue kallikrein (HTK) gene was administrated into the periinfarction region after CI/R. At 12, 24, and 72 h after treatments, neurological deficits were evaluated; expression of HTK and vascular endothelial growth factor (VEGF) were detected by immunohistochemistry staining; the infarction volume was measured; and rCBF was examined by( 14) C-iodoantipyrine microtracing technique. RESULTS The expression of VEGF was enhanced significantly in pAdCMV-HTK group than controls over all time points (P < 0.05). Furthermore, the rCBF in pAdCMV-HTK group increased markedly than controls at 24 and 72 h after treatment (P < 0.05), and the improved neurological deficit was accompanied by reduced infarction volume in pAdCMV-HTK group 24 and 72 h posttreatment. CONCLUSION In the early period after CI/R, kallikrein could induce the angiogenesis and improve rCBF in periinfarction region, and further reduce the infarction volume and improve the neurological deficits.
Collapse
Affiliation(s)
- Rui-Yan Lu
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Tissue kallikrein protects neurons from hypoxia/reoxygenation-induced cell injury through Homer1b/c. Cell Signal 2012; 24:2205-15. [PMID: 22575735 DOI: 10.1016/j.cellsig.2012.04.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 04/27/2012] [Indexed: 11/24/2022]
Abstract
Previous studies have demonstrated that human tissue kallikrein (TK) gene delivery protects against mouse cerebral ischemia/reperfusion (I/R) injury through bradykinin B2 receptor (B2R) activation. We have also reported that exogenous TK administration can suppress glutamate- or acidosis-induced neurotoxicity through the extracellular signal-regulated kinase1/2 (ERK1/2) pathway. To further explore the neuroprotection mechanisms of TK, in the present study we performed immunoprecipitation analysis and identified a scaffolding protein Homer1b/c using MALDI-TOF MS analysis. Here, we tested the hypothesis that TK reduces cell injury induced by oxygen and glucose deprivation/reoxygenation (OGD/R) through activating Homer1b/c. We found that TK increased the expression of Homer1b/c in a concentration- and time-dependent manner. Moreover, TK facilitated the translocation of Homer1b/c to the plasma membrane under OGD/R condition by confocal microscope assays. We also observed that overexpression of Homer1b/c showed the neuroprotection against OGD/R-induced cell injury by enhancing cell survival, reducing LDH release, caspase-3 activity and cell apoptosis. However, the knockdown of Homer1b/c by small interfering RNA showed the opposite effects, indicating that Homer1b/c had protective effects against OGD/R-induced neuronal injury. More interestingly, TK exerted its much more significantly neuroprotective effects after Homer1b/c overexpression, whereas it exerted its reduced effects after Homer1b/c knockdown. In addition, TK pretreatment increased the phosphorylation of the ERK1/2 and Akt-GSK3β through Homer1b/c activation. The beneficial effects of Homer1b/c were abolished by the ERK1/2 or PI3K antagonist. Therefore, we propose novel signaling mechanisms involved in the anti-hypoxic function of TK through activation of Homer1b/c-ERK1/2 and Homer1b/c-PI3K-Akt signaling pathways.
Collapse
|
46
|
Song X, Han L, Liu Y. Remodeling of motor cortex function in acute cerebral infarction patients following human urinary kallidinogenase: A functional magnetic resonance imaging evaluation after 6 months. Neural Regen Res 2012; 7:867-73. [PMID: 25737716 PMCID: PMC4342716 DOI: 10.3969/j.issn.1673-5374.2012.11.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 02/24/2012] [Indexed: 12/01/2022] Open
Abstract
A total of 29 patients were treated within 48 hours after acute subcortical cerebral infarction with Xuesaitong or Xuesaitong plus human urinary kallidinogenase for 14 days. Neurological deficits, activity of daily living, and evaluations of distal upper limb motor functions at the 6-month follow-up showed that patients treated with Xuesaitong plus human urinary kallidinogenase recovered better than with Xuesaitong alone. In addition, functional MRI revealed that activation sites were primarily at the ipsilesional side of injury in all patients. Human urinary kallidinogenase induced hyperactivation of the ipsilesional primary sensorimotor cortex, premotor cortex, supplementary motor area, and contralesional posterior parietal cortex. Results showed that human urinary kallidinogenase improved symptoms of neurological deficiency by enhancing remodeling of long-term cortical motor function in patients with acute cerebral infarction.
Collapse
Affiliation(s)
- Xuezhu Song
- Department of Neurology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, Guangdong Province, China
| | - Lixin Han
- Magnetic Resonance Center of Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, Guangdong Province, China
| | - Yan Liu
- Department of Neurology, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou 510010, Guangdong Province, China
| |
Collapse
|
47
|
Kinin-B2 receptor mediated neuroprotection after NMDA excitotoxicity is reversed in the presence of kinin-B1 receptor agonists. PLoS One 2012; 7:e30755. [PMID: 22348022 PMCID: PMC3277507 DOI: 10.1371/journal.pone.0030755] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 12/21/2011] [Indexed: 01/21/2023] Open
Abstract
Background Kinins, with bradykinin and des-Arg9-bradykinin being the most important ones, are pro-inflammatory peptides released after tissue injury including stroke. Although the actions of bradykinin are in general well characterized; it remains controversial whether the effects of bradykinin are beneficial or not. Kinin-B2 receptor activation participates in various physiological processes including hypotension, neurotransmission and neuronal differentiation. The bradykinin metabolite des-Arg9-bradykinin as well as Lys-des-Arg9-bradykinin activates the kinin-B1 receptor known to be expressed under inflammatory conditions. We have investigated the effects of kinin-B1 and B2 receptor activation on N-methyl-D-aspartate (NMDA)-induced excitotoxicity measured as decreased capacity to produce synaptically evoked population spikes in the CA1 area of rat hippocampal slices. Principal Findings Bradykinin at 10 nM and 1 µM concentrations triggered a neuroprotective cascade via kinin-B2 receptor activation which conferred protection against NMDA-induced excitotoxicity. Recovery of population spikes induced by 10 nM bradykinin was completely abolished when the peptide was co-applied with the selective kinin-B2 receptor antagonist HOE-140. Kinin-B2 receptor activation promoted survival of hippocampal neurons via phosphatidylinositol 3-kinase, while MEK/MAPK signaling was not involved in protection against NMDA-evoked excitotoxic effects. However, 100 nM Lys-des-Arg9-bradykinin, a potent kinin-B1 receptor agonist, reversed bradykinin-induced population spike recovery. The inhibition of population spikes recovery was reversed by PD98059, showing that MEK/MAPK was involved in the induction of apoptosis mediated by the B1 receptor. Conclusions Bradykinin exerted protection against NMDA-induced excitotoxicity which is reversed in the presence of a kinin-B1 receptor agonist. As bradykinin is converted to the kinin-B1 receptor metabolite des-Arg9-bradykinin by carboxypeptidases, present in different areas including in brain, our results provide a mechanism for the neuroprotective effect in vitro despite of the deleterious effect observed in vivo.
Collapse
|
48
|
Rust NM, Papa MP, Scovino AM, da Silva MMC, Calzavara-Silva CE, Marques ETDA, Peçanha LMT, Scharfstein J, Arruda LB. Bradykinin enhances Sindbis virus infection in human brain microvascular endothelial cells. Virology 2012; 422:81-91. [DOI: 10.1016/j.virol.2011.10.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 08/03/2011] [Accepted: 10/04/2011] [Indexed: 12/16/2022]
|
49
|
Simonyi A, Serfozo P, Lehmidi TM, Cui J, Gu Z, Lubahn DB, Sun AY, Sun GY. The neuroprotective effects of apocynin. Front Biosci (Elite Ed) 2012; 4:2183-93. [PMID: 22202030 DOI: 10.2741/535] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The recognition of health benefits of phytomedicines and herbal supplements lead to an increased interest to understand the cellular and molecular basis of their biological activities. Apocynin (4-hydroxy-3-methoxy-acetophenone) is a constituent of the Himalayan medicinal herb Picrorhiza kurroa which is regarded as an inhibitor of nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase, a superoxide-producing enzyme. NADPH oxidase appears to be especially important in the modulation of redox-sensitive signaling pathways and also has been implicated in neuronal dysfunction and degeneration, and neuroinflammmation in diseases ranging from stroke, Alzheimer's and Parkinson's diseases to psychiatric disorders. In this review, we aim to give an overview of current literature on the neuroprotective effects of apocynin in the prevention and treatment of neurodegenerative disorders. Particular attention is given to in vivo studies.
Collapse
Affiliation(s)
- Agnes Simonyi
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Zhang Q, Ding H, Yan J, Wang W, Ma A, Zhu Z, Cianflone K, Hu FB, Hui R, Wang DW. Plasma tissue kallikrein level is negatively associated with incident and recurrent stroke: a multicenter case-control study in China. Ann Neurol 2011; 70:265-73. [PMID: 21823154 DOI: 10.1002/ana.22404] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Tissue kallikrein (TK) cleaves kininogen to produce the potent bioactive compounds kinin and bradykinin, which lower blood pressure and protect the heart, kidneys, and blood vessels. Reduction in TK levels is associated with cardiovascular disease and diabetes in animal models. In this study, we investigated the association of TK levels with event-free survival over 5 years in Chinese first-ever stroke patients. METHODS We conducted a case-control study with 1,268 stroke patients (941 cerebral infarction, 327 cerebral hemorrhage) and 1,210 controls. Plasma TK levels were measured with an enzyme-linked immunosorbent assay. We used logistic regression and Cox proportional hazards models to assess the relationship between TK levels and risk of first-time or recurrent stroke. RESULTS Plasma TK levels were significantly lower in stroke patients (0.163 ± 0.064mg/l vs 0.252 ± 0.093mg/l, p < 0.001), especially those with ischemic stroke. After adjustment for traditional risk factors, plasma TK levels were negatively associated with the risk of first-ever stroke (odds ratio [OR], 0.344; 95% confidence interval [CI], 0.30-0.389; p < 0.001) and stroke recurrence and positively associated with event-free survival during 5 years of follow-up (relative risk, 0.82; 95% CI, 0.74-0.90; p < 0.001). Compared with the first quartile of plasma TK levels, the ORs for first-ever stroke patients were as follows: second quartile, 0.77 (95% CI, 0.56-1.07); third quartile, 0.23 (95% CI, 0.17-0.32); fourth quartile, 0.04 (95% CI, 0.03-0.06). INTERPRETATION Lower plasma TK levels are independently associated with first-ever stroke and are an independent predictor of recurrence after an initial stroke.
Collapse
Affiliation(s)
- Qin Zhang
- Institute of Hypertension and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|