1
|
Al-Kuraishy HM, Al-Gareeb AI, Zekry SH, Alruwaili M, Alexiou A, Papadakis M, Batiha GES. The possible role of cerebrolysin in the management of vascular dementia: Leveraging concepts. Neuroscience 2025:S0306-4522(25)00050-8. [PMID: 39832667 DOI: 10.1016/j.neuroscience.2025.01.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 01/06/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
Cerebrolysin (CBL) is a combination of neurotrophic peptides and amino acids derived from pig brains. CBL can cross the blood-brain barrier (BBB) and its biological effect is similar to the effect of endogenous neurotrophic effects. The mechanism of action of CBL is related to the induction of neurogenesis, neuroplasticity, neuroprotection, and neurotrophicity. Therefore, CBL may be effective against the development and progression of neurodegenerative diseases such as Alzheimer disease (AD) and cerebrovascular disorders such as vascular dementia (VD). Moreover, many studies highlighted that CBL is effective in the improvement of cognitive impairment in patients with neurodegenerative diseases. However, the underlying neuroprotective effects of CBL against the VD neuropathology were not fully elucidated. Thus, this review aims to discuss the possible therapeutic efficacy of CBL in the management of VD. In conclusion, CBL could be effective therapeutic strategy in preventing and treating VD by targeting neuroinflammation, BBB injury, and chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine College of Medicine Mustansiriyah University Baghdad Iraq.
| | - Ali I Al-Gareeb
- FRCP Jabir ibn Hayyan Medical University Al-Ameer Qu./Najaf - Iraq Po. Box (13) Kufa Iraq.
| | - Salwa H Zekry
- Department of Pharmacognosy, Faculty of Pharmacy, Sinai University- Arish Branch, Arish 45511 Egypt.
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Mohali, India
| | - Marios Papadakis
- University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke 42283 Wuppertal, Germany.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511 AlBeheira, Egypt
| |
Collapse
|
2
|
Chen B, Yang T, Liao Z, Sun F, Mei Z, Zhang W. Pathophysiology and Management Strategies for Post-Stroke Spasticity: An Update Review. Int J Mol Sci 2025; 26:406. [PMID: 39796261 PMCID: PMC11721500 DOI: 10.3390/ijms26010406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/22/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Post-stroke spasticity (PSS), characterized by a velocity-dependent increase in muscle tone and exaggerated reflexes, affects a significant portion of stroke patients and presents a substantial obstacle to post-stroke rehabilitation. Effective management and treatment for PSS remains a significant clinical challenge in the interdisciplinary aspect depending on the understanding of its etiologies and pathophysiology. We systematically review the relevant literature and provide the main pathogenic hypotheses: alterations in the balance of excitatory and inhibitory inputs to the descending pathway or the spinal circuit, which are secondary to cortical and subcortical ischemic or hemorrhagic injury, lead to disinhibition of the stretch reflex and increased muscle tone. Prolongation of motoneuron responses to synaptic excitation by persistent inward currents and secondary changes in muscle contribute to hypertonia. The guidelines for PSS treatment advocate for a variety of therapeutic approaches, yet they are hindered by constraints such as dose-dependent adverse effects, high cost, and limited therapeutic efficacy. Taken together, we highlight key processes of PSS pathophysiology and summarize many interventions, including neuroprotective agents, gene therapy, targeted therapy, physiotherapy, NexTGen therapy and complementary and alternative medicine. We aim to confer additional clinical benefits to patients and lay the foundation for the development of new potential therapies against PSS.
Collapse
Affiliation(s)
- Bei Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (B.C.); (T.Y.); (Z.L.); (F.S.)
| | - Tong Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (B.C.); (T.Y.); (Z.L.); (F.S.)
| | - Zi Liao
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (B.C.); (T.Y.); (Z.L.); (F.S.)
| | - Feiyue Sun
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (B.C.); (T.Y.); (Z.L.); (F.S.)
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (B.C.); (T.Y.); (Z.L.); (F.S.)
| | - Wenli Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
3
|
Kojder K, Jarosz K, Andrzejewska A, Solek-Pastuszka J, Skonieczna-Żydecka K, Kaczmarczyk M, Jurczak A. Cerebrolysin in Patients Diagnosed with Subarachnoid Hemorrhage-The Results of an Observational Cohort Study. Biomedicines 2024; 12:2492. [PMID: 39595058 PMCID: PMC11592139 DOI: 10.3390/biomedicines12112492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Subarachnoid hemorrhage (SAH) is associated with high mortality and a high level of disability. Progress in surgical and endovascular techniques has lowered the mortality rate in patients with SAH. However, many patients are left with neurological impairment. Objectives: In our study we wanted to examine the impact of Cerebrolysin on treatment results in patients with SAH diagnosis. Methods: The data of 47 patients, divided into Cerebrolysin (26) and non-Cerebrolysin (21) group were included. We examined the correlation between Cerebrolysin administration and additional Amantadine treatment or neuromonitoring, craniectomy, and endovascular treatment and its impact on the Glasgow Outcome Scale (GOS) score, length of stay (LOS), and mortality. Results: Our study shows that Cerebrolysin improves the mortality rate in combination with neuromonitoring in a group of patients with severe SAH. It does not affect the raw values of GOS or LOS in patients with SAH. Conclusions: Further studies with larger patient groups are needed to investigate the role of Cerebrolysin as an additional treatment in SAH.
Collapse
Affiliation(s)
- Klaudyna Kojder
- Anesthesiology and Intensive Care Department, Pomeranian Medical University, Rybacka 1 str., 70-204 Szczecin, Poland; (K.J.); (A.A.); (J.S.-P.)
| | - Konrad Jarosz
- Anesthesiology and Intensive Care Department, Pomeranian Medical University, Rybacka 1 str., 70-204 Szczecin, Poland; (K.J.); (A.A.); (J.S.-P.)
| | - Agata Andrzejewska
- Anesthesiology and Intensive Care Department, Pomeranian Medical University, Rybacka 1 str., 70-204 Szczecin, Poland; (K.J.); (A.A.); (J.S.-P.)
| | - Joanna Solek-Pastuszka
- Anesthesiology and Intensive Care Department, Pomeranian Medical University, Rybacka 1 str., 70-204 Szczecin, Poland; (K.J.); (A.A.); (J.S.-P.)
| | - Karolina Skonieczna-Żydecka
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland; (K.S.-Ż.); (M.K.)
| | - Mariusz Kaczmarczyk
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, Broniewskiego 24, 71-460 Szczecin, Poland; (K.S.-Ż.); (M.K.)
| | - Anna Jurczak
- Department of Specialist Nursery, Pomeranian Medical University, Zolnierska Str. 48, 71-210 Szczecin, Poland;
| |
Collapse
|
4
|
Liao YS, Zhang TC, Tang YQ, Yu P, Liu YN, Yuan J, Zhao L. Electroacupuncture reduces inflammatory damage following cerebral ischemia-reperfusion by enhancing ABCA1-mediated efferocytosis in M2 microglia. Mol Brain 2024; 17:61. [PMID: 39223647 PMCID: PMC11367741 DOI: 10.1186/s13041-024-01135-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Ischemic stroke (IS) is a severe cerebrovascular disease with high disability and mortality rates, where the inflammatory response is crucial to its progression and prognosis. Efferocytosis, the prompt removal of dead cells, can reduce excessive inflammation after IS injury. While electroacupuncture (EA) has been shown to decrease inflammation post-ischemia/reperfusion (I/R), its link to efferocytosis is unclear. Our research identified ATP-binding cassette transporter A1 (Abca1) as a key regulator of the engulfment process of efferocytosis after IS by analyzing public datasets and validating findings in a mouse model, revealing its close ties to IS progression. We demonstrated that EA can reduce neuronal cell death and excessive inflammation caused by I/R. Furthermore, EA treatment increased Abca1 expression, prevented microglia activation, promoted M2 microglia polarization, and enhanced their ability to phagocytose injured neurons in I/R mice. This suggests that EA's modulation of efferocytosis could be a potential mechanism for reducing cerebral I/R injury, making regulators of efferocytosis steps a promising therapeutic target for EA benefits.
Collapse
Affiliation(s)
- Yu-Sha Liao
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, Sichuan, China
| | - Tie-Chun Zhang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, Sichuan, China
| | - Yu-Qi Tang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, Sichuan, China
| | - Pei Yu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, Sichuan, China
| | - Ya-Ning Liu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, Sichuan, China
| | - Jing Yuan
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, Sichuan, China.
- Key Laboratory of Acupuncture for Senile Disease (Chengdu University of TCM), Ministry of Education, Chengdu, 611137, Sichuan, China.
| | - Ling Zhao
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, No. 1166 Liutai Avenue, Chengdu, 611137, Sichuan, China.
- Key Laboratory of Acupuncture for Senile Disease (Chengdu University of TCM), Ministry of Education, Chengdu, 611137, Sichuan, China.
- Clinical Research Center for Acupuncture and Moxibustion in Sichuan Province, Chengdu, 610075, China.
| |
Collapse
|
5
|
Zhang Z, Jia Z. Pre-IVM with C-type natriuretic peptide promotes mitochondrial biogenesis of bovine oocytes via activation of CREB. Sci Rep 2024; 14:16260. [PMID: 39009622 PMCID: PMC11250819 DOI: 10.1038/s41598-024-67094-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
The aim of this study was to evaluate the effects of C-type natriuretic peptide (CNP) treatment prior to in vitro maturation (IVM) on mitochondria biogenesis in bovine oocyte matured in vitro and explore the related causes. The results showed that treatment with CNP before IVM significantly improved mitochondrial content, elevated the expression of genes related to mitochondria biogenesis, and increased the protein levels of phosphorylation of cAMP-response element binding protein (p-CREB) in bovine oocytes following IVM. However, further studies revealed that treatment with CNP before IVM could not increased the protein levels of p-CREB in bovine oocytes when natriuretic peptide receptor 2 activities was inhibited using the relative specific inhibitor Gö6976. In addition, treatment with CNP before IVM could not improved mitochondrial content or elevated the expression of genes related to mitochondria biogenesis in bovine oocytes when CREB activities was abolished using the specific inhibitor 666-15. In summary, these results provide evidence that treatment of bovine oocytes with CNP before IVM promotes mitochondrial biogenesis in vitro, possibly by activating CREB.
Collapse
Affiliation(s)
- Zehua Zhang
- College of Animal Science and Technology, Inner Mongolia Minzu University, 536 West Huolinhe Street, Tongliao, 028000, Inner Mongolia, People's Republic of China
| | - Zhenwei Jia
- College of Animal Science and Technology, Inner Mongolia Minzu University, 536 West Huolinhe Street, Tongliao, 028000, Inner Mongolia, People's Republic of China.
| |
Collapse
|
6
|
Guan X, Wu J, Geng J, Ji D, Wei D, Ling Y, Zhang Y, Jiang G, Pang T, Huang Z. A Novel Hybrid of Telmisartan and Borneol Ameliorates Neuroinflammation and White Matter Injury in Ischemic Stroke Through ATF3/CH25H Axis. Transl Stroke Res 2024; 15:195-218. [PMID: 36577854 DOI: 10.1007/s12975-022-01121-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022]
Abstract
Cerebral ischemic stroke causes substantial white matter injury, which is further aggravated by neuroinflammation mediated by microglia/astrocytes. Given the anti-neuroinflammatory action of telmisartan and the enhancing blood-brain barrier (BBB) permeability potential of resuscitation-inducing aromatic herbs, 13 hybrids (3a-m) of telmisartan (or its simplified analogues) with resuscitation-inducing aromatic agents were designed, synthesized, and biologically evaluated. Among them, the optimal compound 3a (the ester hybrid of telmisartan and (+)-borneol) potently inhibited neuroinflammation mediated by microglia/astrocytes and ameliorated ischemic stroke. Particularly, 3a significantly conferred protection for white matter integrity after cerebral ischemic stroke via decreasing abnormally dephosphorylated neurofilament protein, upregulating myelin basic protein, and attenuating oligodendrocyte damage. Further RNA-sequencing data revealed that 3a upregulated expression of transcriptional regulator ATF3 to reduce the expression of CH25H, prevented proinflammatory state of lipid-droplet-accumulating microglia/astrocytes to limit excessive inflammation, and eventually protected neighboring oligodendrocytes to prevent white matter injury. Taken with the desirable pharmacokinetics behavior and improved brain distribution, 3a may be a feasible therapeutic agent for ischemic stroke and other neurological disorders with white matter injury.
Collapse
Affiliation(s)
- Xin Guan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Institute of Pharmaceutical Sciences, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Jianbing Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Institute of Pharmaceutical Sciences, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Jiahui Geng
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Institute of Pharmaceutical Sciences, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Duorui Ji
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Institute of Pharmaceutical Sciences, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Dasha Wei
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Institute of Pharmaceutical Sciences, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Yong Ling
- School of Pharmacy, Nantong University, Nantong, 226001, People's Republic of China
| | - Yihua Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Institute of Pharmaceutical Sciences, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China
| | - Guojun Jiang
- Department of Pharmacy, Zhejiang Xiaoshan Hospital, Hangzhou, 311201, People's Republic of China
| | - Tao Pang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Institute of Pharmaceutical Sciences, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China.
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210023, People's Republic of China.
| | - Zhangjian Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Institute of Pharmaceutical Sciences, China Pharmaceutical University, #24 Tong Jia Xiang Street, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
7
|
Kalinin MN, Khasanova DR. Heterogeneous treatment effects of Cerebrolysin as an early add-on to reperfusion therapy: post hoc analysis of the CEREHETIS trial. Front Pharmacol 2024; 14:1288718. [PMID: 38249342 PMCID: PMC10796496 DOI: 10.3389/fphar.2023.1288718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/11/2023] [Indexed: 01/23/2024] Open
Abstract
Background: There has been intensive research into enhancing the effects of reperfusion therapy to mitigate hemorrhagic transformation (HT) in stroke patients. Using neuroprotective agents alongside intravenous thrombolysis (IVT) appears a promising approach. Cerebrolysin is one of the candidates since it consists of neuropeptides mimicking the action of neurotrophic factors on brain protection and repair. Objectives: We looked at treatment effects of Cerebrolysin as an early add-on to IVT in stroke patients with varying HT risk. Methods: It was post hoc analysis of the CEREHETIS trial (ISRCTN87656744). Patients with middle cerebral artery infarction (n = 238) were selected from the intention-to-treat population. To stratify participants according to their HT risk, the DRAGON, SEDAN and HTI scores were computed for each eligible subject using on-admission data. The study endpoints were any and symptomatic HT, and functional outcome measured with the modified Rankin Scale (mRS) on day 90. Favorable functional outcome (FFO) was defined as an mRS ≤2. The performance of each stratification tool was estimated with regression approaches. Heterogeneous treatment effect analysis was conducted using techniques of meta-analysis and the matching-smoothing method. Results: The HTI score outperformed other tools in terms of HT risk stratification. Heterogeneity of Cerebrolysin treatment effects was moderate (I2, 35.8%-56.7%; H2, 1.56-2.31) and mild (I2, 10.9%; H2, 1.12) for symptomatic and any HT, respectively. A significant positive impact of Cerebrolysin on HT and functional outcome was observed in the moderate (HTI = 1) and high (HTI ≥2) HT risk patients, but it was neutral in those with the low (HTI = 0) risk. In particular, there was a steady decline in the rate of symptomatic (HTI = 0 vs. HTI = 4: by 4.3%, p = 0.077 vs. 21.1%, p < 0.001) and any HT (HTI = 0 vs. HTI = 4: by 1.2%, p = 0.737 vs. 32.7%, p < 0.001). Likewise, an mRS score reduction (HTI = 0 vs. HTI = 4: by 1.8%, p = 0.903 vs. 126%, p < 0.001) with a reciprocal increase of the fraction of FFO patients (HTI = 0 vs. HTI = 4: by 1.2% p = 0.757 vs. 35.5%, p < 0.001) was found. Conclusion: Clinically meaningful heterogeneity of Cerebrolysin treatment effects on HT and functional outcome was established in stroke patients. The beneficial effects were significant in those whose estimated on-admission HT risk was either moderate or high.
Collapse
Affiliation(s)
- Mikhail N. Kalinin
- Department of Neurology, Kazan State Medical University, Kazan, Russia
- Department of Neurology, Interregional Clinical Diagnostic Center, Kazan, Russia
| | - Dina R. Khasanova
- Department of Neurology, Kazan State Medical University, Kazan, Russia
- Department of Neurology, Interregional Clinical Diagnostic Center, Kazan, Russia
| |
Collapse
|
8
|
Wang Y, Wu Z, Yan G, Li S, Zhang Y, Li G, Wu C. The CREB1 inhibitor 666-15 maintains cartilage homeostasis and mitigates osteoarthritis progression. Bone Joint Res 2024; 13:4-18. [PMID: 38163445 PMCID: PMC10758301 DOI: 10.1302/2046-3758.131.bjr-2023-0016.r2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2024] Open
Abstract
Aims cAMP response element binding protein (CREB1) is involved in the progression of osteoarthritis (OA). However, available findings about the role of CREB1 in OA are inconsistent. 666-15 is a potent and selective CREB1 inhibitor, but its role in OA is unclear. This study aimed to investigate the precise role of CREB1 in OA, and whether 666-15 exerts an anti-OA effect. Methods CREB1 activity and expression of a disintegrin and metalloproteinase with thrombospondin motifs 4 (ADAMTS4) in cells and tissues were measured by immunoblotting and immunohistochemical (IHC) staining. The effect of 666-15 on chondrocyte viability and apoptosis was examined by cell counting kit-8 (CCK-8) assay, JC-10, and terminal deoxynucleotidyl transferase-mediated dUTP nick end-labelling (TUNEL) staining. The effect of 666-15 on the microstructure of subchondral bone, and the synthesis and catabolism of cartilage, in anterior cruciate ligament transection mice were detected by micro-CT, safranin O and fast green (S/F), immunohistochemical staining, and enzyme-linked immunosorbent assay (ELISA). Results CREB1 was hyperactive in osteoarthritic articular cartilage, interleukin (IL)-1β-treated cartilage explants, and IL-1β- or carbonyl cyanide 3-chlorophenylhydrazone (CCCP)-treated chondrocytes. 666-15 enhanced cell viability of OA-like chondrocytes and alleviated IL-1β- or CCCP-induced chondrocyte injury through inhibition of mitochondrial dysfunction-associated apoptosis. Moreover, inhibition of CREB1 by 666-15 suppressed expression of ADAMTS4. Additionally, 666-15 alleviated joint degeneration in an ACLT mouse model. Conclusion Hyperactive CREB1 played a critical role in OA development, and 666-15 exerted anti-IL-1β or anti-CCCP effects in vitro as well as joint-protective effects in vivo. 666-15 may therefore be used as a promising anti-OA drug.
Collapse
Affiliation(s)
- Ying Wang
- Department of Molecular Orthopedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Zhimin Wu
- Department of Molecular Orthopedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Guoqiang Yan
- National Center for Orthopaedics, Animal Laboratory, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Shan Li
- Department of Molecular Orthopedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Yanzhuo Zhang
- Department of Molecular Orthopedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Guangping Li
- National Center for Orthopaedics, Laboratory of Bone Tissue Engineering, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Chengai Wu
- Department of Molecular Orthopedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
9
|
Kalinin MN, Khasanova DR. [Cerebrolysin as an early add-on to reperfusion therapy: heterogeneous treatment effect analysis in ischemic stroke patients with varying risk of hemorrhagic transformation]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:55-66. [PMID: 38512096 DOI: 10.17116/jnevro202412403255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
OBJECTIVE The study goal was the assessment of heterogeneous treatment effects of Cerebrolysin as an early add-on to reperfusion therapy in stroke patients with varying risk of hemorrhagic transformation (HT). MATERIAL AND METHODS It was post hoc analysis of the CEREHETIS trial (ISRCTN87656744). Patients with middle cerebral artery infarction (n=238) were stratified by HT risk with the HTI score. The study outcomes were symptomatic and any HT, and functional outcome measured with the modified Rankin Scale (mRS) on day 90. Favorable outcome was defined as an mRS score of ≤2. Heterogeneous treatment effect analysis was performed using techniques of meta-analysis and the matching-smoothing method. RESULTS Heterogeneity of Cerebrolysin treatment effects was moderate (I2=36.98-69.3%, H2=1.59-3.26) and mild (I2=18.33-32.39%, H2=1.22-1.48) for symptomatic and any HT, respectively. A positive impact of the Cerebrolysin treatment on HT and functional outcome was observed in patients with moderate (HTI=1) and high (HTI≥2) HT risk. However, the effect was neutral in those with low risk (HTI=0). In high HT risk patients, there was a steady decline in the rate of symptomatic (HTI=0 vs. HTI≥2: by 3.8%, p=0.120 vs. 14.3%, p<0.001) and any HT (HTI=0 vs. HTI≥2: by 0.6%, p=0.864 vs. 19.5%, p<0.001). Likewise, Cerebrolysin treatment resulted in an overall decrease in the mRS scores (HTI=0 vs. HTI≥2: by 2.1%, p=0.893 vs. 63%, p<0.001) with a reciprocal increase of the fraction with favorable outcome (HTI=0 vs. HTI≥2: by 2% p=0.634 vs. 19.2%, p<0.001). CONCLUSION Clinically meaningful heterogeneity of Cerebrolysin treatment effects on HT and functional outcome was established in stroke patients. The Cerebrolysin positive impact was significant in those whose estimated on-admission HT risk was either moderate or high.
Collapse
Affiliation(s)
- M N Kalinin
- Kazan State Medical University, Kazan, Russia
- Interregional Clinical Diagnostic Center, Kazan, Russia
| | - D R Khasanova
- Kazan State Medical University, Kazan, Russia
- Interregional Clinical Diagnostic Center, Kazan, Russia
| |
Collapse
|
10
|
Fan XY, Shi G, Zhao YP, Yang JJ, Feng J. Neuroprotective effects of oxytocin against ischemic stroke in rats by blocking glutamate release and CREB-mediated DNA hypermethylation. Biomed Pharmacother 2023; 167:115520. [PMID: 37729734 DOI: 10.1016/j.biopha.2023.115520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023] Open
Abstract
Glutamate plays a crucial role in cognitive impairments after ischemic stroke. There is a scarcity of information about how glutamate-induced activation of cAMP-response element binding (CREB) signaling pathway regulates both the negative and positive regulators of synaptic plasticity. Recent studies have demonstrated the involvement of prominent epigenetic repressors, such as MeCP2 and DNMTs, in stroke. Neuroprotective effects of oxytocin against ischemia have been previously reported, while the underlying mechanism is still elusive. In this research, the possible role of CREB-mediated DNA hypermethylation and the potential mechanism of oxytocin in a rat model of permanent middle cerebral artery occlusion (pMCAO) were assessed. Adult male Sprague-Dawley rats were pretreated with intraperitoneal injection of oxytocin at the onset of pMCAO. The effects of oxytocin on spines and the expression levels of synaptic genes were determined. The regulatory effects of oxytocin on glutamate level, N-methyl-D-aspartate receptors (NMDARs), its downstream CREB pathway, and global or gene-specific DNA methylation status were evaluated by immunofluorescence, co-immunoprecipitation, and chromatin immunoprecipitation, respectively. We found that CREB could act as a common transcription factor for MeCP2 and DNMT3B after ischemic stroke. Oxytocin dose-dependently deactivated NR2B-related CaM-CREB pathway and inhibited DNA hypermethylation at the CpG islands of Ngf gene in pMCAO-operated rats. Moreover, oxytocin prevented pMCAO-induced reduction in the number of spines and neural cells. DNA hypermethylation in Ngf gene contributed to the cognitive deficits post-stroke. The neuroprotective effects of oxytocin against ischemia could be attributed to inhibiting glutamate release, providing additional evidence on the mechanism of oxytocin against ischemic stroke.
Collapse
Affiliation(s)
- Xin-Yu Fan
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China
| | - Guang Shi
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China; Department of Neurology, The People's Hospital of Liaoning Province, Shenyang, China
| | - Yun-Peng Zhao
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jing-Jing Yang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
11
|
Marghani BH, Rezk S, Ateya AI, Alotaibi BS, Othman BH, Sayed SM, Alshehri MA, Shukry M, Mansour MM. The Effect of Cerebrolysin in an Animal Model of Forebrain Ischemic-Reperfusion Injury: New Insights into the Activation of the Keap1/Nrf2/Antioxidant Signaling Pathway. Int J Mol Sci 2023; 24:12080. [PMID: 37569457 PMCID: PMC10418386 DOI: 10.3390/ijms241512080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Forebrain ischemia-reperfusion (IR) injury causes neurological impairments due to decreased cerebral autoregulation, hypoperfusion, and edema in the hours to days following the restoration of spontaneous circulation. This study aimed to examine the protective and/or therapeutic effects of cerebrolysin (CBL) in managing forebrain IR injury and any probable underlying mechanisms. To study the contribution of reperfusion to forebrain injury, we developed a transient dual carotid artery ligation (tDCAL/IR) mouse model. Five equal groups of six BLC57 mice were created: Group 1: control group (no surgery was performed); Group 2: sham surgery (surgery was performed without IR); Group 3: tDCAL/IR (surgery with IR via permanently ligating the left CA and temporarily closing the right CA for 30 min, followed by reperfusion for 72 h); Group 4: CBL + tDCAL/IR (CBL was given intravenously at a 60 mg/kg BW dose 30 min before IR); and Group 5: tDCAL/IR + CBL (CBL was administered i.v. at 60 mg/kg BW three hours after IR). At 72 h following IR, the mice were euthanized. CBL administration 3 h after IR improved neurological functional recovery, enhanced anti-inflammatory and antioxidant activities, alleviated apoptotic neuronal death, and inhibited reactive microglial and astrocyte activation, resulting in neuroprotection after IR injury in the tDCAL/IR + CBL mice group as compared to the other groups. Furthermore, CBL reduced the TLRs/NF-kB/cytokines while activating the Keap1/Nrf2/antioxidant signaling pathway. These results indicate that CBL may improve neurologic function in mice following IR.
Collapse
Affiliation(s)
- Basma H. Marghani
- Department of Physiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
- Department of Biochemistry, Physiology, and Pharmacology, Faculty of Veterinary Medicine, King Salman International University, El Tor 46612, Egypt
| | - Shaymaa Rezk
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed I. Ateya
- Department of Husbandry and Development of Animal Wealth, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Badriyah S. Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Basma H. Othman
- Medical Experimental Research Center, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Samy M. Sayed
- Faculty of Agriculture, Cairo University, Giza 12613, Egypt;
- Department of Science and Technology, Ranyah University College, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Mohammed Ali Alshehri
- Biology Department, College of Science, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Mustafa Shukry
- Physiology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Mohamed M. Mansour
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
12
|
Wach J, Vychopen M, Güresir A, Güresir E. Anti-Inflammatory Drug Therapy in Aneurysmal Subarachnoid Hemorrhage: A Systematic Review and Meta-Analysis of Prospective Randomized and Placebo-Controlled Trials. J Clin Med 2023; 12:4165. [PMID: 37373858 DOI: 10.3390/jcm12124165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/02/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
Emerging evidence suggests that neuroinflammation may play a potential role in aneurysmal subarachnoid hemorrhage (aSAH). We aim to analyze the influence of anti-inflammatory therapy on survival and outcome in aSAH. Eligible randomized placebo-controlled prospective trials (RCTs) were searched in PubMed until March 2023. After screening the available studies for inclusion and exclusion criteria, we strictly extracted the main outcome measures. Dichotomous data were determined and extracted by odds ratio (OR) with 95% confidence intervals (CIs). Neurological outcome was graded using the modified Rankin Scale (mRS). We created funnel plots to analyze publication bias. From 967 articles identified during the initial screening, we included 14 RCTs in our meta-analysis. Our results illustrate that anti-inflammatory therapy yields an equivalent probability of survival compared to placebo or conventional management (OR: 0.81, 95% CI: 0.55-1.19, p = 0.28). Generally, anti-inflammatory therapy trended to be associated with a better neurologic outcome (mRS ≤ 2) compared to placebo or conventional treatment (OR: 1.48, 95% CI: 0.95-2.32, p = 0.08). Our meta-analysis showed no increased mortality form anti-inflammatory therapy. Anti-inflammatory therapy in aSAH patients tends to improve the neurological outcome. However, multicenter, rigorous, designed, prospective randomized studies are still needed to investigate the effect of fighting inflammation in improving neurological functioning after aSAH.
Collapse
Affiliation(s)
- Johannes Wach
- Department of Neurosurgery, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Martin Vychopen
- Department of Neurosurgery, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Agi Güresir
- Department of Neurosurgery, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Erdem Güresir
- Department of Neurosurgery, University Hospital Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
13
|
Kumar Nambi P, Kanna Sathyamoorthy Y, Kaliyappan K, Kumar Radhakrishnan R. Fucoidan (A sulfated polysaccharide) and Cerebroprotein in combination alleviate the neuroinflammation-mediated neural damage and functional deficits in the focal cerebral ischemia model of rat. Neuroscience 2023:S0306-4522(23)00207-5. [PMID: 37182836 DOI: 10.1016/j.neuroscience.2023.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
Cerebral ischemic reperfusion injury could emanate a cascade of events ensuing in neural death and severe neurobehavioural deficits. The currently available interventions have failed to target the multimodal, interlinked mechanisms that operate cerebral ischemia-induced damage and functional loss. So an integrative intervention has become a mandate to overcome the deleterious mechanisms involved in cerebral ischemic pathophysiology. In this study, adult male Sprague dawley rats were exposed to 2 hours of right middle cerebral artery occlusion (rMCAo) followed by reperfusion, and the intervention group received Fucoidan alone at a dose of 50mg/kg, i.p (intraperitoneal), Cerebrolysin alone at a dose of 2.5mg/kg body weight and the combination of both. The sham rats were exposed to surgical procedures, except for the rMCAo. The assessments of the groups were made 24 hours after the rMCAo. The stand-alone treatment with Fucoidan, Cerebrolysin has shown a better outcome in the neurobehavioral and, histopathological assessments and the combination has made a significant reduction in the neurological deficits and the infarct volume when compared to the standalone groups. The BBB integrity was well preserved in the combination group when compared with the lesion and standalone groups. Moreover, the combined intervention reduced the level of pro-inflammatory cytokines TNFα, NFkB, IL1α, IL1-β, IL-6, CD68, COX-2, and mRNA expression of inflammatory genes IL1α, IL1-β, IL-6, IBA-1, and COX-2 effectively. In conclusion, the present study suggests that rMCAo induced neuroinflammation and neurobehavioural alterations were attenuated by intervention with a combination of Fucoidan and cerebrolysin; Further, Fucoidan and Cerebrolysin combination improved the ischemic tolerance level by promoting the proteins and genes that regulate the inflammatory cytokines and in aiding better recovery after ischemic reperfusion injury.
Collapse
Affiliation(s)
- Pradeep Kumar Nambi
- Department of Anatomy, Dr. Arcot Lakshmanaswamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani campus Chennai 600 113, Tamil Nadu, India
| | - Yogesh Kanna Sathyamoorthy
- Department of Anatomy, Dr. Arcot Lakshmanaswamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani campus Chennai 600 113, Tamil Nadu, India
| | - Kathiravan Kaliyappan
- Department of Anatomy, Dr. Arcot Lakshmanaswamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani campus Chennai 600 113, Tamil Nadu, India
| | - Ramesh Kumar Radhakrishnan
- Department of Anatomy, Dr. Arcot Lakshmanaswamy Mudaliar Post Graduate Institute of Basic Medical Sciences, University of Madras, Taramani campus Chennai 600 113, Tamil Nadu, India.
| |
Collapse
|
14
|
Khasanova DR, Kalinin MN. Cerebrolysin as an Early Add-on to Reperfusion Therapy: Risk of Hemorrhagic Transformation after Ischemic Stroke (CEREHETIS), a prospective, randomized, multicenter pilot study. BMC Neurol 2023; 23:121. [PMID: 36973684 PMCID: PMC10041692 DOI: 10.1186/s12883-023-03159-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Cerebrolysin could mitigate reperfusion injury and hemorrhagic transformation (HT) in animal models of acute ischemic stroke. METHODS This was a prospective, randomized, open-label, parallel-group with active control, multicenter pilot study. Cerebrolysin (30 mL/day over 14 days) was administered concurrently with alteplase (0.9 mg/kg) in 126 patients, whereas 215 control patients received alteplase alone. The primary outcomes were the rate of any and symptomatic HT assessed from day 0 to 14. The secondary endpoints were drug safety and functional outcome measured with the National Institutes of Health Stroke Scale (NIHSS) on day 1 and 14, and the modified Rankin scale (mRS) on day 90. Advanced brain imaging analysis was applied on day 1 and 14 as a marker for in vivo pharmacology of Cerebrolysin. RESULTS Cerebrolysin treatment resulted in a substantial decrease of the symptomatic HT rate with an odds ratio (OR) of 0.248 (95% CI: 0.072-0.851; p = 0.019). No serious adverse events attributed to Cerebrolysin occurred. On day 14, the Cerebrolysin arm showed a significant decrease in the NIHSS score (p = 0.045). However, no difference in the mRS score was observed on day 90. A substantial improvement in the advanced brain imaging parameters of the infarcted area was evident in the Cerebrolysin group on day 14. CONCLUSIONS Early add-on of Cerebrolysin to reperfusion therapy was safe and significantly decreased the rate of symptomatic HT as well as early neurological deficit. No effect on day 90 functional outcome was detected. Improvements in the imaging metrics support the neuroprotective and blood-brain barrier stabilizing activity of Cerebrolysin. TRIAL REGISTRATION Name of Registry: ISRCTN. TRIAL REGISTRATION NUMBER ISRCTN87656744 . Trial Registration Date: 16/02/2021.
Collapse
Affiliation(s)
- Dina R Khasanova
- Department of Neurology and Neurosurgery for Postgraduate Training, Kazan State Medical University, Kazan, Russia
- Department of Neurology, Interregional Clinical Diagnostic Center, 12A Karbyshev St, Kazan, 420101, Russia
| | - Mikhail N Kalinin
- Department of Neurology and Neurosurgery for Postgraduate Training, Kazan State Medical University, Kazan, Russia.
- Department of Neurology, Interregional Clinical Diagnostic Center, 12A Karbyshev St, Kazan, 420101, Russia.
| |
Collapse
|
15
|
Kwon M, Lee JH, Yoon Y, Pleasure SJ, Yoon K. The CRHR1/CREB/REST signaling cascade regulates mammalian embryonic neural stem cell properties. EMBO Rep 2023; 24:e55313. [PMID: 36413000 PMCID: PMC9900344 DOI: 10.15252/embr.202255313] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 10/31/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022] Open
Abstract
Growing evidence suggests that the corticotropin-releasing hormone (CRH) signaling pathway, mainly known as a critical initiator of humoral stress responses, has a role in normal neuronal physiology. However, despite the evidence of CRH receptor (CRHR) expression in the embryonic ventricular zone, the exact functions of CRH signaling in embryonic brain development have not yet been fully determined. In this study, we show that CRHR1 is required for the maintenance of neural stem cell properties, as assessed by in vitro neurosphere assays and cell distribution in the embryonic cortical layers following in utero electroporation. Identifying the underlying molecular mechanisms of CRHR1 action, we find that CRHR1 functions are accomplished through the increasing expression of the master transcription factor REST. Furthermore, luciferase reporter and chromatin immunoprecipitation assays reveal that CRHR1-induced CREB activity is responsible for increased REST expression at the transcriptional level. Taken together, these findings indicate that the CRHR1/CREB/REST signaling cascade plays an important role downstream of CRH in the regulation of neural stem cells during embryonic brain development.
Collapse
Affiliation(s)
- Mookwang Kwon
- Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwonSouth Korea
| | - Ju Hyun Lee
- Department of Biopharmaceutical ConvergenceSungkyunkwan UniversitySuwonSouth Korea
| | - Youngik Yoon
- Department of Biopharmaceutical ConvergenceSungkyunkwan UniversitySuwonSouth Korea
| | - Samuel J Pleasure
- Department of NeurologyUniversity of California San FranciscoSan FranciscoCAUSA
| | - Keejung Yoon
- Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwonSouth Korea
- Department of Biopharmaceutical ConvergenceSungkyunkwan UniversitySuwonSouth Korea
| |
Collapse
|
16
|
d-allose protects brain microvascular endothelial cells from hypoxic/reoxygenated injury by inhibiting endoplasmic reticulum stress. Neurosci Lett 2023; 793:137000. [PMID: 36473686 DOI: 10.1016/j.neulet.2022.137000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/17/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Ischemic stroke is an acute brain disease with a high mortality rate. Currently, the only effective method is to restore the blood supply. But the inflammation and oxidative stress induced by this approach can damage the integrity of the endothelial system, which hampers the patient's outcome. d-allose has the biological activity to protect against ischemia-reperfusion injury, however, the underlying mechanism remains unclear. Here, brain microvascular endothelial cells (RBMECs) were used as the study material to establish an IR-injury model. Cell viability of RBMECs was suppressed after hypoxia/reoxygenation (H/R) treatment and significantly increased after d-allose supplementation. RNAseq results showed 180 differentially expressed genes (DEGs) between the therapy group (H/R + Dal) and the model group (H/R), of which 151 DEGs were restored to control levels by d-allose. Enrichment analysis revealed that DEGs were mainly involved in protein processing in endoplasmic reticulum. 6 DEGs in the unfolded protein response (UPR) pathway were verified by qRT-PCR. All of them were significantly down-regulated by d-allose, indicating that endoplasmic reticulum stress (ERS) was relieved. In addition, d-allose significantly inhibited the phosphorylation level of eIF2α, a marker of ERS. The downstream molecules of Phosphorylation of eIF2α, Gadd45a and Chac1, which trigger cycle arrest and apoptosis, respectively, were also significantly inhibited by d-allose. Thus, we conclude that d-allose inhibits the UPR pathway, attenuates eIF2α phosphorylation and ERS, restores the cell cycle, inhibits apoptosis, and thus enhances endothelial cell tolerance to H/R injury.
Collapse
|
17
|
Sun X, Wang T, Zhou L, Zhang C, Fu F. The effect of triple reuptake inhibitor toludesvenlafaxine on neurological function in cerebral ischemic rats. Front Pharmacol 2023; 14:1073099. [PMID: 37153779 PMCID: PMC10160376 DOI: 10.3389/fphar.2023.1073099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/12/2023] [Indexed: 05/10/2023] Open
Abstract
Purpose: The aim is to investigate the effect of toludesvenlafaxine (Tdv), a reuptake inhibitor of serotonin, norepinephrine, and dopamine, on the neurological function in cerebral ischemic rats and the underlying mechanisms. Material and Methods: Middle cerebral artery occlusion/reperfusion (MCAO/R) model was induced in rats and the neuroprotective effects of Tdv were evaluated by infarct size, Garcia test, and beam walking test. Neuronal apoptosis in the peri-infarct area was observed by TUNEL staining. And the apoptosis-related proteins were evaluated with Western blotting. The role of CREB pathway in effect of Tdv was also investigated using Western blotting and immunofluorescence. Results: In the MCAO/R model, administration of Tdv reduced the infarct size, promoted neural functional recovery, decreased the expression of Bax and Caspase-3, and increased the expression of Bcl-2 and BDNF. In addition, Tdv reduced neuronal apoptosis in the peri-infarct area. Tdv increased the expression of phosphorylated CREB. The application of the specific CREB inhibitor, compound 666-15, could reverse the anti-ischemic cerebral injury of Tdv in MCAO/R rats. Conclusion: Tdv ameliorated cerebral ischemic injury through reducing neuronal apoptosis and increasing the expression of BDNF via the activation of CREB pathway.
Collapse
|
18
|
Khasanova DR, Kalinin MN. [Effects of simultaneous use of Cerebrolysin and alteplase on hemorrhagic transformation of brain infarction and functional outcome in stroke patients: CEREHETIS, a randomized, multicenter pilot trial]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:60-69. [PMID: 37682097 DOI: 10.17116/jnevro202312308260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
OBJECTIVE The study aimed to assess effects of the simultaneous use of Cerebrolysin and intravenous thrombolysis (Alteplase) on hemorrhagic transformation (HT) and functional outcome as well as to analyze the treatment safety in acute stroke patients. MATERIAL AND METHODS It was a prospective, randomized, open-label, multicenter, parallel-group, active-controlled pilot study (Trial Registration Number: ISRCTN87656744, https://doi.org/10.1186/ISRCTN87656744, Trial registration date: 16/02/2021). The intervention group (n=126) was treated with Cerebrolysin infusion (30 mL) started simultaneously with Alteplase (0.9 mg/kg) via a separate IV line. The Cerebrolysin treatment continued for 14 consecutive days with the baseline therapy along. The control group (n=215) received only Alteplase and the baseline therapy. The primary endpoints were the rate of any and symptomatic hemorrhagic transformation (HT) from admission to day 14. Secondary endpoints were treatment safety and functional outcome measured with the National Institutes of Health stroke scale (NIHSS) in 24 h and on day 14, and with the modified Rankin scale (mRS) on day 90. RESULTS Treatment with Cerebrolysin resulted in a significant reduction of the symptomatic HT rate with an odds ratio of 0.248 (95% CI: 0.072-0.851; p=0.019). No serious adverse events related to Cerebrolysin were observed. On day 14, the intervention group showed a significant reduction in the NIHSS score (p=0.045). However, no difference in the mRS score was observed on day 90, but there was a trend towards its improvement. CONCLUSION The combination of Cerebrolysin and Alteplase was safe and significantly reduced the rate of symptomatic HT and improved early neurological deficit. However, no difference in functional outcome was found on day 90, but there was a trend towards favorable functional outcome.
Collapse
Affiliation(s)
- D R Khasanova
- Kazan State Medical University, Kazan, Russia
- Interregional Clinical Diagnostic Center, Kazan, Russia
| | - M N Kalinin
- Kazan State Medical University, Kazan, Russia
- Interregional Clinical Diagnostic Center, Kazan, Russia
| |
Collapse
|
19
|
Morega S, Gresita A, Mitran SI, Musat MI, Boboc IKS, Gheorman V, Udristoiu I, Albu CV, Streba CT, Catalin B, Rogoveanu I. Cerebrolysin Use in Patients with Liver Damage-A Translational Study. Life (Basel) 2022; 12:1791. [PMID: 36362945 PMCID: PMC9695462 DOI: 10.3390/life12111791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/26/2022] [Accepted: 11/02/2022] [Indexed: 09/15/2024] Open
Abstract
The treatment of acute life-threatening events in patients suffering from chronic pathologies is problematic, as physicians need to consider multisystemic drug effects. Regarding Cerebrolysin, a Sonic Hedgehog signaling pathway amplifier and one of the few approved neurotrophic treatments for stroke patients, concerns of excessive Hedgehog pathway activation that could accelerate NAFLD progression to cirrhosis seem valid. We investigated stroke patients treated with Cerebrolysin that presented elevated levels of aspartate aminotransferase (AST) and/or alanine aminotransferase (ALT). We also investigated the efficiency of Cerebrolysin in reversing the neurogenesis inhibition within the hippocampus in a mouse model of NAFLD by evaluating behavior and histological outcomes. NeuN, BrdU and Iba1 positive signals in the cortex and hippocampus of the animals were also observed. Clinically, Cerebrolysin improved AST levels in a majority of stroke patients with hepatic damage. The same treatment in an experimental setup was able to reverse anxiety-like behavior in MCD mice, reducing their freezing time from 333.61 ± 21.81 s in MCD animals to 229.17 ± 26.28 in treated ones. The use of Cerebrolysin did not improve short-term memory nor rescued cell multiplication in the hippocampus after MCD food intake. Understanding the neuroprotective and neurotrophic effects that drugs have on NAFLD patients can significantly contribute to a suitable therapeutic approach.
Collapse
Affiliation(s)
- Shandiz Morega
- U.M.F. Doctoral School Craiova, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Andrei Gresita
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Biomedical Sciences, New York Institute of Technology, Old Westbury, NY 115680-8000, USA
| | - Smaranda Ioana Mitran
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Madalina Iuliana Musat
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ianis Kevyn Stefan Boboc
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Victor Gheorman
- Department of Psychiatry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ion Udristoiu
- Department of Psychiatry, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Carmen Valeria Albu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Costin Teodor Streba
- Research Center of Gastroenterology and Hepatology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Bogdan Catalin
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Ion Rogoveanu
- Gastroenterology Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
20
|
Cerebrolysin Alleviating Effect on Glutamate-Mediated Neuroinflammation Via Glutamate Transporters and Oxidative Stress. J Mol Neurosci 2022; 72:2292-2302. [PMID: 36333611 DOI: 10.1007/s12031-022-02078-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
Glutamate, one of the most important excitatory neurotransmitters, acts as a signal transducer in peripheral tissues and endocrine cells. Excessive glutamate secretion has been shown to cause excitotoxicity and neurodegenerative disease. Cerebrolysin is a mixture of enzymatically treated peptides derived from pig brain including neurotrophic factors, like brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF), nerve growth factor (NGF), and ciliary neurotrophic factor (CNTF). The present study investigated the protective effects of cerebrolysin on glutamate transporters (EAAT 1, EAAT 2) and cytokines (IL-1β and IL-10) activity in glutamate-mediated neurotoxicity. Primary cortex neuron culture was exposed to glutamate and successively treated with various cerebrolysin concentrations for 24 and 48 h. Our data showed that cerebrolysin primarily protects neurons by decreasing glutamate concentration in the synaptic cleft. In addition, Cerebrolysin can decrease oxidative stress and neuron cell damage by increasing antioxidant activity and decreasing inflammation cytokine levels.
Collapse
|
21
|
Lu W, Zhu Z, Shi D, Li X, Luo J, Liao X. Cerebrolysin alleviates early brain injury after traumatic brain injury by inhibiting neuroinflammation and apoptosis via TLR signaling pathway. Acta Cir Bras 2022; 37:e370605. [PMID: 36074398 PMCID: PMC9448247 DOI: 10.1590/acb370605] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 05/03/2022] [Indexed: 11/22/2022] Open
Abstract
Purpose: Traumatic brain injury (TBI) is a major cause of death and disability. Cerebrolysin (CBL) has been reported to be anti-inflammatory by reducing reactive oxygen species (ROS) production. However, the neuroprotection of CBL in TBI and the potential mechanism are unclear. We aimed to investigate the neuroprotection and mechanisms of CBL in TBI. Methods: The TBI model was established in strict accordance with the Feeney weight-drop model of focal injury. The neurological score, brain water content, neuroinflammatory cytokine levels, and neuronal damage were evaluated. The involvement of the early brain injury modulatory pathway was also investigated. Results: Following TBI, the results showed that CBL administration increased neurological scores and decreased brain edema by alleviating blood‑brain barrier (BBB) permeability, upregulating tight junction protein (ZO‑1) levels, and decreasing the levels of the inflammatory cytokines tumor necrosis factor‑α (TNF‑α), interleukin‑1β (IL‑1β), IL‑6, and NF‑κB. The TUNEL assay showed that CBL decreased hippocampal neuronal apoptosis after TBI and decreased the protein expression levels of caspase‑3 and Bax, increasing the levels of Bcl‑2. The levels of Toll‑like receptor 2 (TLR2) and TLR4 were significantly decreased after CBL treatment. In TBI patients, CBL can also decrease TNF‑α, IL‑1β, IL‑6, and NF‑κB levels. This result indicates that CBL‑mediated inhibition of neuroinflammation and apoptosis ameliorated neuronal death after TBI. The neuroprotective capacity of CBL is partly dependent on the TLR signaling pathway. Conclusions: Taken together, the results of this study indicate that CBL can improve neurological outcomes and reduce neuronal death against neuroinflammation and apoptosis via the TLR signaling pathway in mice.
Collapse
Affiliation(s)
- Weihong Lu
- BS. 904th Hospital of Joint Logistic Support Force of PLA - Department of Anesthesiology - Wuxi, China
| | - Zhonghua Zhu
- BS. 904th Hospital of Joint Logistic Support Force of PLA - Department of Anesthesiology - Wuxi, China
| | - Dongliang Shi
- MD. Anhui Medical University - Wuxi Clinical College - 904th Hospital of Joint Logistic Support Force of PLA - Department of Neurosurgery - Wuxi, China
| | - Xiaoyu Li
- BS. 904th Hospital of Joint Logistic Support Force of PLA - Department of Anesthesiology - Wuxi, China
| | - Jingzhi Luo
- BS. 904th Hospital of Joint Logistic Support Force of PLA - Department of Anesthesiology - Wuxi, China
| | - Xingzhi Liao
- BS. 904th Hospital of Joint Logistic Support Force of PLA - Department of Anesthesiology - Wuxi, China
| |
Collapse
|
22
|
Li K, Ran B, Wang Y, Liu L, Li W. PLCγ2 impacts microglia-related effectors revealing variants and pathways important in Alzheimer’s disease. Front Cell Dev Biol 2022; 10:999061. [PMID: 36147734 PMCID: PMC9485805 DOI: 10.3389/fcell.2022.999061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
Alzheimer’s disease (AD) is an irreversible neurodegenerative disease mainly characterized by memory loss and cognitive decline. The etiology of AD is complex and remains incompletely understood. In recent years, genome-wide association studies (GWAS) have increasingly highlighted the central role of microglia in AD pathology. As a trans-membrane receptor specifically present on the microglia in the central nervous system, phosphatidylinositol-specific phospholipase C gamma 2 (PLCγ2) plays an important role in neuroinflammation. GWAS data and corresponding pathological research have explored the effects of PLCG2 variants on amyloid burden and tau pathologies that underline AD. The link between PLCγ2 and other AD-related effectors in human and mouse microglia has also been established, placing PLCγ2 downstream of the triggering receptor expressed on myeloid cells 2 (TREM2), toll-like receptor 4 (TLR4), Bruton’s tyrosine kinase (BTK), and colony-stimulating factor 1 receptor (CSF1R). Because the research on PLCγ2’s role in AD is still in its early stages, few articles have been published, therefore in this paper, we integrate the relevant research published to date, review the structural features, expression patterns, and related pathways of PLCγ2, and summarize the recent studies on important PLCG2 variants related to AD. Furthermore, the possibility and challenge of using PLCγ2 to develop therapeutic drugs for AD are also discussed.
Collapse
|
23
|
Staszewski J, Stȩpień A, Piusińska-Macoch R, Dȩbiec A, Gniadek-Olejniczak K, Frankowska E, Maliborski A, Chadaide Z, Balo D, Król B, Namias R, Harston G, Mróz J, Piasecki P. Efficacy of Cerebrolysin Treatment as an Add-On Therapy to Mechanical Thrombectomy in Patients With Acute Ischemic Stroke Due to Large Vessel Occlusion: Study Protocol for a Prospective, Open Label, Single-Center Study With 12 Months of Follow-Up. Front Neurol 2022; 13:910697. [PMID: 35860483 PMCID: PMC9289167 DOI: 10.3389/fneur.2022.910697] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/30/2022] [Indexed: 12/18/2022] Open
Abstract
This study is designed to determine the efficacy of Cerebrolysin treatment as an add-on therapy to mechanical thrombectomy (MT) in reducing global disability in subjects with acute ischemic stroke (AIS). We have planned a single center, prospective, open-label, single-arm study with a 12-month follow-up of 50 patients with moderate to severe AIS, with a small established infarct core and with good collateral circulation who achieve significant reperfusion following MT and who receive additional Cerebrolysin within 8 h of stroke onset compared to 50 historical controls treated with MT alone, matched for age, clinical severity, occlusion location, baseline perfusion lesion volume, onset to reperfusion time, and use of iv thrombolytic therapy. The primary outcome measure will be the overall proportion of subjects receiving Cerebrolysin compared to the control group experiencing a favorable functional outcome (by modified Rankin Scale 0-2) at 90 days, following stroke onset. The secondary objectives are to determine the efficacy of Cerebrolysin as compared to the control group in reducing the risk of symptomatic secondary hemorrhagic transformation, improving neurological outcomes (NIHSS 0-2 at day 7, day 30, and 90), reducing mortality rates (over the 90-day and 12 months study period), and improving: activities of daily living (by Barthel Index), health-related quality of life (EQ-5D-5L) assessed at day 30, 90, and at 12 months. The other measures of efficacy in the Cerebrolysin group will include: assessment of final stroke volume and penumbral salvage (measured by CT/CTP at 30 days) and its change compared to baseline volume, changes over time in language function (by the 15-item Boston Naming Test), hemispatial neglect (by line bisection test), global cognitive function (by The Montreal Cognitive Assessment), and depression (by Hamilton Depression Rating Scale) between day 30 and day 90 assessments). The patients will receive 30 ml of Cerebrolysin within 8 h of AIS stroke onset and continue treatment once daily until day 21 (first cycle) and they will receive a second cycle of treatment (30 ml/d for 21 days given in the Outpatient Department or Neurorehabilitation Clinic) from day 69 to 90.
Collapse
Affiliation(s)
- Jacek Staszewski
- Clinic of Neurology, Military Institute of Medicine, Warsaw, Poland
| | - Adam Stȩpień
- Clinic of Neurology, Military Institute of Medicine, Warsaw, Poland
| | | | | | | | - Emilia Frankowska
- Department of Radiology, Military Institute of Medicine, Warsaw, Poland
| | - Artur Maliborski
- Department of Radiology, Military Institute of Medicine, Warsaw, Poland
| | - Zoltan Chadaide
- Brainomix Ltd., and Oxford University Hospitals NHSFT, Oxford, United Kingdom
| | - David Balo
- Brainomix Ltd., and Oxford University Hospitals NHSFT, Oxford, United Kingdom
| | - Beata Król
- Brainomix Ltd., and Oxford University Hospitals NHSFT, Oxford, United Kingdom
| | - Rafael Namias
- Brainomix Ltd., and Oxford University Hospitals NHSFT, Oxford, United Kingdom
| | - George Harston
- Brainomix Ltd., and Oxford University Hospitals NHSFT, Oxford, United Kingdom
| | - Józef Mróz
- Neurorehabilitation Clinic, Military Institute of Medicine, Warsaw, Poland
| | - Piotr Piasecki
- Department of Radiology, Military Institute of Medicine, Warsaw, Poland
| |
Collapse
|
24
|
Farajdokht F, Oghbaei F, Sadigh-Eteghad S, Majdi A, Aghsan SR, Farhoudi M, Vahidi-Eyrisofla N, Mahmoudi J. Cerebrolysin® and environmental enrichment, alone or in combination, ameliorate anxiety- and depressive-like behaviors in a post-ischemic depression model in mice. J Stroke Cerebrovasc Dis 2022; 31:106519. [DOI: 10.1016/j.jstrokecerebrovasdis.2022.106519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/18/2022] [Accepted: 04/14/2022] [Indexed: 11/27/2022] Open
|
25
|
Tran L, Alvarez XA, Le HA, Nguyen DA, Le T, Nguyen N, Nguyen T, Nguyen T, Vo T, Tran T, Duong C, Nguyen H, Nguyen S, Nguyen H, Le T, Nguyen M, Nguyen T. Clinical Efficacy of Cerebrolysin and Cerebrolysin plus Nootropics in the Treatment of Patients with Acute Ischemic Stroke in Vietnam. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 21:621-630. [PMID: 34414874 DOI: 10.2174/1871527320666210820091655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/29/2021] [Accepted: 07/11/2021] [Indexed: 01/13/2023]
Abstract
AIMS To investigate the efficacy and safety of Cerebrolysin and Cerebrolysin plus nootropics in the routine treatment of patients with acute ischemic stroke (AIS). BACKGROUND Acute ischemic stroke (AIS) is a leading cause of disability with unmet treatment needs lacking effective drug therapy. Multimodal drugs modulating stroke pathophysiology as Cerebrolysin constitute a good therapeutic option. OBJECTIVE In this study, we assessed the effects of Cerebrolysin and Cerebrolysin plus nootropics, in comparison with other nootropic drugs alone, on functional, neurological and cognitive recovery of patients with AIS in Vietnam. METHODS This non-interventional, controlled, open-label, prospective and multicenter study included 398 AIS patients (234 males) treated with Cerebrolysin (n=190; 20 i.v. infusions of 10 ml), other nootropics (comparator group; n=86), or a combination of both (n=122). The study primary endpoint was the modified Ranking Scale (mRS) score on day 90. Secondary endpoints included study-period change in NIHSS score; percentage of well-recovered (mRS 0-2) patients, the proportion of good NIHSS response (≥6 points) cases, and MoCA scores at day 90; and safety indicators. RESULTS Compared with other nootropics, both Cerebrolysin and combined therapy induced significant improvements (p<0.001) in: Functional recovery (mRS scores); percentage of well-recovered patients (Cerebrolysin: 81.6%; combination: 93.4%; comparator: 43.0%); neurological recovery (study- period NIHSS change); proportion of good NIHSS responders (Cerebrolysin: 77.5%; combination: 92.5%; comparator: 47.6%); and MoCA scores (Cerebrolysin: 23.3±4.8; combination: 23.7±4.1; comparator: 15.9±7.7). Compared to Cerebrolysin, combined therapy improved (p<0.01) mRS outcomes and NIHSS change, but not MoCA scores, in moderate-severe stroke (NIHSS>11) cases only. No drug-related adverse events were reported. CONCLUSION Cerebrolysin alone or combined with other nootropics was effective and safe in routine AIS treatment, during both acute and recovery phases, which supports its use in daily clinical practice. Others: According to the results of this multicenter study, the importance of reducing differences in the treatment regimens of AIS in Vietnam should be further emphasized.
Collapse
Affiliation(s)
- Luc Tran
- Outpatient Department, National Geriatric Hospital, Hanoi, Vietnam
| | - X Anton Alvarez
- Medinova Institute of Neurosciences, Clinica RehaSalud, A Coruña, Spain.,Clinical Research Department, QPS Holdings, A Coruña, Spain
| | | | | | - Thinh Le
- Neurology Department, Bach Mai Hospital, Hanoi, Vietnam
| | - Ngoc Nguyen
- Stroke centre, 108 Military Hospital, Hanoi, Vietnam
| | - Thang Nguyen
- Department of Cerebrovascular Disease, 115 People's Hospital, Hochiminh City, Vietnam
| | - Tai Nguyen
- Neurology Department, Cho Ray Hospital, Hochiminh City, Vietnam
| | - Tan Vo
- Neurology Department, Gia Dinh People's Hospital, Hochiminh City, Vietnam
| | - Tuan Tran
- Neurology Department, Thai Nguyen Central General Hosp, Thainguyen, Vietnam
| | - Chinh Duong
- Neurology Department, Nghe An General Hospital, Nghean, Vietnam
| | - Huyen Nguyen
- Neurology Department, Viet Tiep General Hospital, Haiphong, Vietnam
| | - Sam Nguyen
- Neurology Department, Thanh Hoa General Hospital, Thanhhoa, Vietnam
| | - Hien Nguyen
- Stroke unit, 103 Military Hospital, Hanoi, Vietnam
| | - Thanh Le
- Neurology Department, Thong Nhat Hospital, Hochiminh City, Vietnam
| | - Minh Nguyen
- Neurology Department, Trung Vuong Hospital, Hochiminh City, Vietnam
| | - Thang Nguyen
- Neurology Department, Hochiminh City Medicine and Pharmacy University Hospital, Hochiminh City, Vietnam
| |
Collapse
|
26
|
Chen X, Wei G, Li D, Fan Y, Zeng Y, Qian Z, Jia Z, Tang Y, Shi Y, Wu H, Li X. Sirtuin 1 alleviates microglia-induced inflammation by modulating the PGC-1α/Nrf2 pathway after traumatic brain injury in male rats. Brain Res Bull 2022; 185:28-38. [PMID: 35487384 DOI: 10.1016/j.brainresbull.2022.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/06/2022] [Accepted: 04/24/2022] [Indexed: 11/27/2022]
Abstract
Microglial activation and the subsequent inflammatory response play important roles in the central nervous system after traumatic brain injury (TBI). Activation of the PGC-1α pathway is responsible for microglial activation after TBI. Our previous study demonstrated that SIRT1 alleviates neuroinflammation-induced apoptosis after TBI, and activation of the PGC-1α/Nrf2 pathway extenuates TBI-induced neuronal apoptosis. However, no study has investigated whether SIRT1 can affect the PGC-1α/Nrf2 pathway to induce microglial excitation and the subsequent neuroinflammatory response. Microglial activation and the levels of pro-inflammatory factors, namely, tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) were assessed to evaluate the neuroinflammatory response after TBI. To examine the effects of SIRT1, immunohistochemical staining and western blot analysis were used to observe the nuclear translocation and secretion of PGC-1α, as well as the activation of the PGC-1α/Nrf2 pathway. Treatment with the SIRT1 inhibitor sirtinol promoted microglial activation and pro-inflammatory factor expression (TNF-α, IL-6, and IL-1β) and inhibited PGC-1α and Nrf2 nuclear translocation and secretion after TBI, while treatment with the SIRT1 activator A3 had the opposite effects. The results of this study suggest that microglial activation, the subsequent neuroinflammatory response, and the PGC-1α/Nrf2 pathway play essential roles in secondary injury after TBI. These results indicate that SIRT1 protects neurons after TBI by inhibiting microglial activation and the subsequent inflammatory response, possibly by activating the PGC-1α/Nrf2 pathway.
Collapse
Affiliation(s)
- Xiangrong Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China
| | - Guan Wei
- Department of Emergency, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China
| | - Ding Li
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China
| | - Youwu Fan
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu Province, China
| | - Yile Zeng
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou 362000, Fujian Province, China
| | - Zhengting Qian
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu Province, China
| | - Zhen Jia
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu Province, China
| | - Yong Tang
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu Province, China
| | - Yan Shi
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu Province, China
| | - Heming Wu
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu Province, China.
| | - Xiang Li
- Department of Neurosurgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, Jiangsu Province, China.
| |
Collapse
|
27
|
Cottini F, Rodriguez J, Hughes T, Sharma N, Guo L, Lozanski G, Liu B, Cocucci E, Yang Y, Benson D. Redefining CD56 as a biomarker and therapeutic target in Multiple Myeloma. Mol Cancer Res 2022; 20:1083-1095. [PMID: 35380709 DOI: 10.1158/1541-7786.mcr-21-0828] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 02/11/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022]
Abstract
Multiple myeloma (MM) cells aberrantly express surface antigens compared to normal plasma cells. Among others, CD56 is present at variable levels in approximately 70% of MM patients; however very little is known about CD56 role in MM. We demonstrated that MM patients with more than 10 percent of CD56-expressing clonal MM cells have inferior clinical outcomes. By gain-of and loss-of function models, we revealed that CD56 promotes MM cell growth, survival, and adhesion to stromal cells. These protumoral effects are induced by the activation of the RSK2/CREB1 signaling pathway, with increased mRNA and protein levels of the anti-apoptotic genes BCL2 and MCL1. Consequently, the genomic and pharmacological inhibition of RSK2 or CREB1 specifically induced MM cell death in CD56-expressing MM cells. Finally, we observed that CD56 signaling decreases CRBN expression, reducing responses to lenalidomide. RSK2 or CREB1 inhibition increased CRBN levels and were synergic with lenalidomide in inducing cell death, especially in CD56-expressing MM cells. In conclusion, our findings demonstrate that CD56 promotes MM cell growth, and pave the way to novel therapies based on targeting CD56, along with the use of CD56 as a predictive biomarker for MM therapies. Implications: Multiple myeloma (MM) is an incurable, genetically heterogeneous disease, without available tailored therapeutic approaches. CD56 signaling promotes MM growth and adhesion, by activating CREB1 target genes, MCL1 and BCL2. Inhibition of CREB1 alone or in combination with lenalidomide is an unexplored synthetic lethal approach in CD56-expressing MM patients.
Collapse
Affiliation(s)
- Francesca Cottini
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Jose Rodriguez
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Tiffany Hughes
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Nidhi Sharma
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Ling Guo
- Department of Pathology, OhioHealth, Columbus, Ohio
| | - Gerard Lozanski
- Department of Pathology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Bei Liu
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Emanuele Cocucci
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
- The Ohio State University, College of Pharmacy, Columbus, Ohio
| | - Yiping Yang
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Don Benson
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| |
Collapse
|
28
|
Zheng W, Guo J, Lu X, Qiao Y, Liu D, Pan S, Liang L, Liu C, Zhu H, Liu Z, Liu Z. cAMP-response element binding protein mediates podocyte injury in diabetic nephropathy by targeting lncRNA DLX6-AS1. Metabolism 2022; 129:155155. [PMID: 35093327 DOI: 10.1016/j.metabol.2022.155155] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/16/2022] [Accepted: 01/22/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND Progressive proteinuria is one of the earliest clinical features of diabetic nephropathy (DN). In our previous study, lncRNA DLX6-AS1 (DLX6-AS1, Dlx6os1 in the mouse) was found to be associated with the extent of albuminuria in DN patients. Furthermore, the lack of Dlx6os1 was pivotal in switching off the inflammatory response in db/db mouse model. However, the regulatory factors responsible for elevated DLX6-AS1 in DN remains unknown. METHODS To identify potential regulatory factors for DLX6-AS1, JASPAR database and DNA pull down combined subsequent liquid chromatography-tandem mass spectrometry were used. Dual-luciferase reporter assay and chromatin immunoprecipitation were then performed to confirm binding sites. We also investigated the effects of the regulatory factors on DN progression in db/db mouse model and cultured human podocytes. RESULTS Our analyses demonstrated that cAMP-response element binding protein (CREB) was highly expressed and closely associated with DLX6-AS1 in DN. In db/db mouse and in cultured podocytes, CREB silencing significantly reduced the level of DLX6-AS1 or Dlx6os1 and attenuated renal damage. Mechanistically, CREB overexpression aggravated renal inflammation and destroyed the structure of podocytes by targeting DLX6-AS1. The damaging role of CREB in podocyte injury was also inhibited by 666-15, a selective inhibitor, in a dose-dependent manner. In vivo, the inhibition of CREB by 666-15 significantly attenuated albuminuria and ameliorated inflammatory infiltration in podocytes. CONCLUSIONS Our findings indicated that CREB is a key mediator of podocyte injury and acts by regulating DLX6-AS1. Thus, CREB may be an effective and potential therapeutic target for the treatment of DN.
Collapse
Affiliation(s)
- Wen Zheng
- Department of Integrated Traditional and Western Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China
| | - Jia Guo
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China
| | - Xiaoqing Lu
- Department of Integrated Traditional and Western Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China
| | - Yingjin Qiao
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China
| | - Dongwei Liu
- Department of Integrated Traditional and Western Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China
| | - Shaokang Pan
- Department of Integrated Traditional and Western Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou, PR China
| | - Lulu Liang
- Department of Integrated Traditional and Western Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China
| | - Chang Liu
- Department of Integrated Traditional and Western Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China
| | - Hongchao Zhu
- Department of Integrated Traditional and Western Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China
| | - Zhihong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, PR China.
| | - Zhangsuo Liu
- Department of Integrated Traditional and Western Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, PR China.
| |
Collapse
|
29
|
Isoform-selective HDAC Inhibitor Mocetinostat (MGCD0103) Alleviates Myocardial Ischemia/Reperfusion Injury via Mitochondrial Protection through the HDACs/CREB/PGC-1α Signaling Pathway. J Cardiovasc Pharmacol 2021; 79:217-228. [PMID: 34983914 DOI: 10.1097/fjc.0000000000001174] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/28/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Over the past decade, histone deacetylases (HDACs) has been proven to manipulate development and exacerbation of cardiovascular diseases, including myocardial ischemia/reperfusion injury (MIRI), cardiac hypertrophy, ventricular remodeling, myocardial fibrosis. Inhibition of histone deacetylases, especially class-I HDACs, is potent to protection of ischemic myocardium after ischemia/reperfusion. Herein, we examine whether mocetinostat (MGCD0103, MOCE), a class-I selective HDAC inhibitor in phase-II clinical trial, conducts cardioprotection under ischemia/reperfusion (I/R) in vivo and vitro, if so, reveal its potential pharmacological mechanism to provide an experimental and theoretical basis for mocetinostat usage in a clinical setting. HCMs were exposed to hypoxia and reoxygenation (H/R), with or without mocetinostat treatment. H/R reduced mitochondrial membrane potential (MMP) and induced HCMs apoptosis. Mocetinostat pre-treatment reversed these H/R-induced mitochondrial damage and cellular apoptosis and upregulated CREB, p-CREB and PGC-1α in HCMs during H/R. Transfection with siRNA against PGC-1α or CREB abolished the protective effects of mocetinostat on cardiomyocytes undergoing H/R. In vivo, mocetinostat was demonstrated to protect myocardial injury posed by myocardial ischemia/reperfusion (I/R) via activation of CREB and upregulation of PGC-1α. Mocetinostat (MGCD0103) can protect myocardium from ischemia/reperfusion injury through mitochondrial protection mediated by CREB/PGC-1α pathway. Therefore, activation of the CREB/PGC-1α signaling pathway via inhibition of Class-I HDACs may be a promising new therapeutic strategy for alleviating myocardial reperfusion injury.
Collapse
|
30
|
Yan J, Xu W, Lenahan C, Huang L, Wen J, Li G, Hu X, Zheng W, Zhang JH, Tang J. CCR5 Activation Promotes NLRP1-Dependent Neuronal Pyroptosis via CCR5/PKA/CREB Pathway After Intracerebral Hemorrhage. Stroke 2021; 52:4021-4032. [PMID: 34719258 PMCID: PMC8607924 DOI: 10.1161/strokeaha.120.033285] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 07/11/2021] [Accepted: 07/30/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND PURPOSE Neuronal pyroptosis is a type of regulated cell death triggered by proinflammatory signals. CCR5 (C-C chemokine receptor 5)-mediated inflammation is involved in the pathology of various neurological diseases. This study investigated the impact of CCR5 activation on neuronal pyroptosis and the underlying mechanism involving cAMP-dependent PKA (protein kinase A)/CREB (cAMP response element binding)/NLRP1 (nucleotide-binding domain leucine-rich repeat pyrin domain containing 1) pathway after experimental intracerebral hemorrhage (ICH). METHODS A total of 194 adult male CD1 mice were used. ICH was induced by autologous whole blood injection. Maraviroc (MVC)-a selective antagonist of CCR5-was administered intranasally 1 hour after ICH. To elucidate the underlying mechanism, a specific CREB inhibitor, 666-15, was administered intracerebroventricularly before MVC administration in ICH mice. In a set of naive mice, rCCL5 (recombinant chemokine ligand 5) and selective PKA activator, 8-Bromo-cAMP, were administered intracerebroventricularly. Short- and long-term neurobehavioral assessments, Western blot, Fluoro-Jade C, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and immunofluorescence staining were performed. RESULTS The brain expression of CCL5 (chemokine ligand 5), CCR5, PKA-Cα (protein kinase A-Cα), p-CREB (phospho-cAMP response element binding), and NLRP1 was increased, peaking at 24 hours after ICH. CCR5 was expressed on neurons, microglia, and astrocytes. MVC improved the short- and long-term neurobehavioral deficits and decreased neuronal pyroptosis in ipsilateral brain tissues at 24 hours after ICH, which were accompanied by increased PKA-Cα and p-CREB expression, and decreased expression of NLRP1, ASC (apoptosis-associated speck-like protein containing a CARD), C-caspase-1, GSDMD (gasdermin D), and IL (interleukin)-1β/IL-18. Such effects of MVC were abolished by 666-15. At 24 hours after injection in naive mice, rCCL5 induced neurological deficits, decreased PKA-Cα and p-CREB expression in the brain, and upregulated NLRP1, ASC, C-caspase-1, N-GSDMD, and IL-1β/IL-18 expression. Those effects of rCCL5 were reversed by 8-Bromo-cAMP. CONCLUSIONS CCR5 activation promoted neuronal pyroptosis and neurological deficits after ICH in mice, partially through the CCR5/PKA/CREB/NLRP1 signaling pathway. CCR5 inhibition with MVC may provide a promising therapeutic approach in managing patients with ICH.
Collapse
Affiliation(s)
- Jun Yan
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China (J.Y.)
| | - Weilin Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China (W.X.)
| | - Cameron Lenahan
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, NM (C.L.)
| | - Lei Huang
- Department of Neurosurgery (L.H., J.H.Z.), Loma Linda University, CA
- Department of Physiology and Pharmacology (L.H., J.H.Z., J.T.), Loma Linda University, CA
| | - Jing Wen
- Department of Rheumatism, First Affiliated Hospital of Guangxi Medical University, Nanning, China (J.W.)
| | - Gaigai Li
- Department of Neurology, Tongji Hospital, Tongji Medical college, Huazhong University of Science and Technology, Wuhan, China (G.L.)
| | - Xin Hu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China (X.H.)
| | - Wen Zheng
- Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha, China (W.Z.)
| | - John H. Zhang
- Department of Neurosurgery (L.H., J.H.Z.), Loma Linda University, CA
- Department of Physiology and Pharmacology (L.H., J.H.Z., J.T.), Loma Linda University, CA
- Department of Anesthesiology (J.H.Z.), Loma Linda University, CA
| | - Jiping Tang
- Department of Physiology and Pharmacology (L.H., J.H.Z., J.T.), Loma Linda University, CA
| |
Collapse
|
31
|
Xie J, Li X, Zhang L, Liu C, Leung JWH, Liu P, Yu Z, Liu R, Li L, Huang C, Huang Z. Genistein-3'-sodium sulfonate ameliorates cerebral ischemia injuries by blocking neuroinflammation through the α7nAChR-JAK2/STAT3 signaling pathway in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 93:153745. [PMID: 34634743 DOI: 10.1016/j.phymed.2021.153745] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/28/2021] [Accepted: 09/08/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Neuroinflammation plays a pivotal role in the acute progression of cerebral ischemia/reperfusion injury (I/RI). We previously reported that genistein-3'-sodium sulfonate (GSS), a derivative from the extract of the phytoestrogen genistein (Gen), protects cortical neurons against focal cerebral ischemia. However, the molecular mechanism underlying the neuroprotective effects exerted by GSS remains unclear. PURPOSE The present study focused on the anti-inflammatory effects of GSS following I/RI in rats. STUDY DESIGN Randomized controlled trial. METHODS The tMCAO rat model and LPS-stimulated BV2 in vitro model were used. Longa's scare was used to observe neurological function. TTC staining and Nissl staining were used to evaluate brain injury. ELISA, qRT-PCR, Western blotting and immunofluorescent staining methods were used to detect cytokine concentration, mRNA level, protein expression and location. RESULTS GSS treatment improves neurological function, reduces the volume of cerebral infarction, attenuates proinflammatory cytokines and inactivates the phosphorylation of JAK2 and STAT3 in I/RI rats. Furthermore, GSS increased the expression of α7nAChR. More importantly, the neuroprotective, anti-inflammatory and inhibiting JAK2/STAT3 signaling pathway effects of GSS were counteracted in the presence of alpha-bungarotoxin (α-BTX), an α7nAChR inhibitor, suggesting that α7nAChR is a potential target associated with the anti-inflammatory effects of GSS in the I/RI rats. GSS also inhibited BV2 cells from releasing IL-1β via the α7nAChR pathway after LPS stimulation. CONCLUSION GSS protects against cerebral I/RI through the expression of α7nAChR and inhibition of the JAK2/STAT3 pathway. Our findings provide evidence for the role of the cholinergic anti-inflammatory pathway in neuroinflammation and uncover a potential novel mechanism for GSS treatment in ischemic stroke. The downstream signals of GSS, α7nAChR- JAK2/STAT3 could also be potential targets for the treatment of I/RI.
Collapse
Affiliation(s)
- Jiali Xie
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Department of Physiology, Institute for Medical Sciences of Pain, Gannan Medical University, Ganzhou 341000, China; Department of Basic Medicine, Gannan Health Vocational College, Ganzhou, 341000, China
| | - Xiao Li
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Department of Physiology, Institute for Medical Sciences of Pain, Gannan Medical University, Ganzhou 341000, China; Department of Physiology, Basic Medicine School of Gannan Medical University, Ganzhou 341000, China
| | - Limei Zhang
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Department of Physiology, Institute for Medical Sciences of Pain, Gannan Medical University, Ganzhou 341000, China; Department of Physiology, Basic Medicine School of Gannan Medical University, Ganzhou 341000, China
| | - Chaoming Liu
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Department of Physiology, Institute for Medical Sciences of Pain, Gannan Medical University, Ganzhou 341000, China
| | - Joseph Wai-Hin Leung
- Department of Biology, University of Ottawa, Ottawa, K1N 6N5, Canada; Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, K1H 8L6, Canada
| | - Peiwen Liu
- The first clinical college of Lanzhou University, Nanzhou, 73000, China
| | - Zining Yu
- Graduate School, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Ruizhen Liu
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Department of Physiology, Institute for Medical Sciences of Pain, Gannan Medical University, Ganzhou 341000, China; Department of Physiology, Basic Medicine School of Gannan Medical University, Ganzhou 341000, China
| | - Liangdong Li
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Department of Physiology, Institute for Medical Sciences of Pain, Gannan Medical University, Ganzhou 341000, China; Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Cheng Huang
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Department of Physiology, Institute for Medical Sciences of Pain, Gannan Medical University, Ganzhou 341000, China; Department of Physiology, Basic Medicine School of Gannan Medical University, Ganzhou 341000, China
| | - Zhihua Huang
- Key Laboratory of Prevention and treatment of cardiovascular and cerebrovascular diseases of Ministry of Education, Department of Physiology, Institute for Medical Sciences of Pain, Gannan Medical University, Ganzhou 341000, China; Department of Physiology, Basic Medicine School of Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|
32
|
Tao Y, Xu Y, Shen M, Feng X, Wu Y, Wu Y, Shen L, Wang Y. The neuroprotection of cerebrolysin after spontaneous intracerebral hemorrhage through regulates necroptosis via Akt/ GSK3β signaling pathway. Acta Cir Bras 2021; 36:e361002. [PMID: 34817023 PMCID: PMC8610213 DOI: 10.1590/acb361002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/16/2021] [Indexed: 11/22/2022] Open
Abstract
PURPOSE Spontaneous intracerebral hemorrhage (ICH) is a major cause of death and disability with a huge economic burden worldwide. Cerebrolysin (CBL) has been previously used as a nootropic drug. Necroptosis is a programmed cell death mechanism that plays a vital role in neuronal cell death after ICH. However, the precise role of necroptosis in CBL neuroprotection following ICH has not been confirmed. METHODS In the present study, we aimed to investigate the neuroprotective effects and potential molecular mechanisms of CBL in ICH-induced early brain injury (EBI) by regulating neural necroptosis in the C57BL/6 mice model. Mortality, neurological score, brain water content, and neuronal death were evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, Evans blue extravasation, Western blotting, and quantitative real-time polymerase chain reaction (PCR). RESULTS The results show that CBL treatment markedly increased the survival rate, neurological score, and neuron survival, and downregulated the protein expression of RIP1 and RIP3, which indicated that CBL-mediated inhibition of necroptosis, and ameliorated neuronal death after ICH. The neuroprotective capacity of CBL is partly dependent on the Akt/GSK3β signaling pathway. CONCLUSIONS CBL improves neurological outcomes in mice and reduces neuronal death by protecting against neural necroptosis.
Collapse
Affiliation(s)
| | | | | | | | - Yan Wu
- Anhui Medical University, China
| | | | | | | |
Collapse
|
33
|
Widodo SS, Dinevska M, Furst LM, Stylli SS, Mantamadiotis T. IL-10 in glioma. Br J Cancer 2021; 125:1466-1476. [PMID: 34349251 PMCID: PMC8609023 DOI: 10.1038/s41416-021-01515-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/05/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
The prognosis for patients with glioblastoma (GBM), the most common and malignant type of primary brain tumour, is very poor, despite current standard treatments such as surgery, radiotherapy and chemotherapy. Moreover, the immunosuppressive tumour microenvironment hinders the development of effective immunotherapies for GBM. Cytokines such as interleukin-10 (IL-10) play a major role in modulating the activity of infiltrating immune cells and tumour cells in GBM, predominantly conferring an immunosuppressive action; however, in some circumstances, IL-10 can have an immunostimulatory effect. Elucidating the function of IL-10 in GBM is necessary to better strategise and improve the efficacy of immunotherapy. This review discusses the immunostimulatory and immunosuppressive roles of IL-10 in the GBM tumour microenvironment while considering IL-10-targeted treatment strategies. The molecular mechanisms that underlie the expression of IL-10 in various cell types are also outlined, and how this resulting information might provide an avenue for the improvement of immunotherapy in GBM is explored.
Collapse
Affiliation(s)
- Samuel S. Widodo
- grid.1008.90000 0001 2179 088XDepartment of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC Australia
| | - Marija Dinevska
- grid.1008.90000 0001 2179 088XDepartment of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC Australia
| | - Liam M. Furst
- grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC Australia
| | - Stanley S. Stylli
- grid.1008.90000 0001 2179 088XDepartment of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC Australia ,grid.416153.40000 0004 0624 1200Department of Neurosurgery, Royal Melbourne Hospital, Parkville, VIC Australia
| | - Theo Mantamadiotis
- grid.1008.90000 0001 2179 088XDepartment of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC Australia ,grid.418025.a0000 0004 0606 5526Florey Institute of Neuroscience and Mental Health, Parkville, VIC Australia
| |
Collapse
|
34
|
Wang J, Liu B, Xu Y, Luan H, Wang C, Yang M, Zhao R, Song M, Liu J, Sun L, You J, Wang W, Sun F, Yan H. Thioperamide attenuates neuroinflammation and cognitive impairments in Alzheimer's disease via inhibiting gliosis. Exp Neurol 2021; 347:113870. [PMID: 34563511 DOI: 10.1016/j.expneurol.2021.113870] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease, which characterized by deposition of amyloid-β (Aβ) plaques, neurofibrillary tangles, neuronal loss, and accompanied by neuroinflammation. Neuroinflammatory processes are well acknowledged to contribute to the progression of AD pathology. Histamine H3 receptor (H3R) is a presynaptic autoreceptor regulating histamine release via negative feedback way. Recently, studies show that H3R are highly expressed not only in neurons but also in microglia and astrocytes. H3R antagonist has been reported to have anti-inflammatory efficacy. However, whether inhibition of H3R is responsible for the anti-neuroinflammation in glial cells and neuroprotection on APPswe, PSEN1dE9 (APP/PS1 Tg) mice remain unclear. In this study, we found that inhibition of H3R by thioperamide reduced the gliosis and induced a phenotypical switch from A1 to A2 in astrocytes, and ultimately attenuated neuroinflammation in APP/PS1 Tg mice. Additionally, thioperamide rescued the decrease of cyclic AMP response element-binding protein (CREB) phosphorylation and suppressed the phosphorylated P65 nuclear factor kappa B (p-P65 NF-κB) in APP/PS1 Tg mice. H89, an inhibitor of CREB signaling, abolished these effects of thioperamide to suppress gliosis and proinflammatory cytokine release. Lastly, thioperamide alleviated the deposition of amyloid-β (Aβ) and cognitive dysfunction in APP/PS1 mice, which were both reversed by administration of H89. Taken together, these results suggested the H3R antagonist thioperamide improved cognitive impairment in APP/PS1 Tg mice via modulation of the CREB-mediated gliosis and inflammation inhibiting, which contributed to Aβ clearance. This study uncovered a novel mechanism involving inflammatory regulating behind the therapeutic effect of thioperamide in AD.
Collapse
Affiliation(s)
- Jiangong Wang
- Department of Pharmacology, College of Basic Medicine, Binzhou Medical University, Yantai, China; Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Bin Liu
- Department of Pharmacology, College of Basic Medicine, Binzhou Medical University, Yantai, China; Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Yong Xu
- Department of Pharmacology, College of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Haiyun Luan
- Department of Pharmacology, College of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Chaoyun Wang
- Department of Pharmacology, College of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Meizi Yang
- Department of Pharmacology, College of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Runming Zhao
- Department of Pharmacology, College of Basic Medicine, Binzhou Medical University, Yantai, China
| | - Mengmeng Song
- Department of Thyroid Breast Surgery, Dongying People's Hospital, Dongying, China
| | - Jing Liu
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Linshan Sun
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Jingjing You
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Wentao Wang
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Fengjiao Sun
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Haijing Yan
- Department of Pharmacology, College of Basic Medicine, Binzhou Medical University, Yantai, China; Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China.
| |
Collapse
|
35
|
Wang Y, Guan X, Gao CL, Ruan W, Zhao S, Kai G, Li F, Pang T. Medioresinol as a novel PGC-1α activator prevents pyroptosis of endothelial cells in ischemic stroke through PPARα-GOT1 axis. Pharmacol Res 2021; 169:105640. [PMID: 33915296 DOI: 10.1016/j.phrs.2021.105640] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/17/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023]
Abstract
AIM Brain microvascular endothelial cells (BMVECs), as the important structure of blood-brain barrier (BBB), play a vital role in ischemic stroke. Pyroptosis of different cells in the brain may aggravate cerebral ischemic injury, and PGC-1α plays a major role in pyroptosis. However, it is not known whether BMVECs undergo pyroptosis after ischemic stroke and whether PGC-1α activator Medioresinol (MDN) we discovered may be useful against pyroptosis of endothelial cells and ischemic brain injury. METHODS For in vitro experiments, the bEnd.3 cells and BMVECs under oxygen and glucose-deprivation (OGD) were treated with or without MDN, and the LDH release, tight junction protein degradation, GSDMD-NT membrane location and pyroptosis-associated proteins were evaluated. For in vivo experiments, mice underwent transient middle cerebral artery occlusion (tMCAO) for ischemia model, and the neuroprotective effects of MDN were measured by infarct volume, the permeability of BBB and pyroptosis of BMVECs. For mechanistic study, effects of MDN on the accumulation of phenylalanine, mitochondrial reactive oxygen species (mtROS) were tested by untargeted metabolomics and MitoSOX Red probe, respectively. RESULTS BMVECs underwent pyroptosis after ischemia. MDN dose-dependently activated PGC-1α, significantly reduced pyroptosis, mtROS and the expressions of pyroptosis-associated proteins (NLRP3, ASC, cleaved caspase-1, IL-1β, GSDMD-NT), and increased ZO-1 and Occludin protein expressions in BMVECs. In tMCAO mice, MDN remarkably reduced brain infarct volume and the permeability of BBB, inhibited pyroptosis of BMVECs, and promoted long-term neurobehavioral functional recovery. Mechanistically, MDN promoted the interaction of PGC-1α with PPARα to increase PPARα nuclear translocation and transcription activity, further increased the expression of GOT1 and PAH, resulting in enhanced phenylalanine metabolism to reduce the ischemia-caused phenylalanine accumulation and mtROS and further ameliorate pyroptosis of BMVECs. CONCLUSION In this study, we for the first time discovered that pyroptosis of BMVECs was involved in the pathogenesis of ischemic stroke and MDN as a novel PGC-1α activator could ameliorate the pyroptosis of endothelial cells and ischemic brain injury, which might attribute to reduction of mtROS through PPARα/GOT1 axis in BMVECs. Taken together, targeting endothelial pyroptosis by MDN may provide alternative therapeutics for brain ischemic stroke.
Collapse
Affiliation(s)
- Yunjie Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing 210009, PR China; College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 311402, PR China; Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, PR China; School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, PR China
| | - Xin Guan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing 210009, PR China
| | - Cheng-Long Gao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing 210009, PR China
| | - Wenchen Ruan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing 210009, PR China
| | - Shunyi Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing 210009, PR China
| | - Guoyin Kai
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 311402, PR China
| | - Fei Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Tao Pang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing 210009, PR China; Key Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing 210009, PR China.
| |
Collapse
|
36
|
Zhu L, Liu Y, Wu X, Ren Y, Zhang Q, Ren L, Guo Y. Cerebroprotein hydrolysate-I protects senescence-induced by D-galactose in PC12 cells and mice. Food Sci Nutr 2021; 9:3722-3731. [PMID: 34262731 PMCID: PMC8269606 DOI: 10.1002/fsn3.2333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/14/2021] [Accepted: 05/01/2021] [Indexed: 11/25/2022] Open
Abstract
Cerebroprotein hydrolysate-I (CH-I),a mixture of peptides extracted from porcine brain tissue,has shown a neuroprotective effect, but its role in brain senescence is unclear. In the present study, we established a senescence model of PC12 cells and mice to investigate the effect of CH-I on brain senescence via JAK2/STAT3 pathway. The results showed that CH-I could improve cell viability, inhibit the apoptosis of cells, and reduce the senescence-positive cells induced by D-galactose. In vivo, CH-I improved the learning ability and memory of aging mice, reduced neuronal damage in mice hippocampus. Mechanism studies showed that CH-I could adjust BDNF protein expressions, activate JAK2/STAT3 pathway, and finally enhance telomerase activity. All these findings indicated that CH-I showed a neuroprotective effect against brain senescence. These results might provide further reference and support for the application of CH-I in delaying aging.
Collapse
Affiliation(s)
- Lin Zhu
- Institute of Cerebrovascular DiseasesTaishan Scholars Construction Project Excellent Innovative Team of Shandong ProvinceMedical Research CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Yingjuan Liu
- Institute of Cerebrovascular DiseasesTaishan Scholars Construction Project Excellent Innovative Team of Shandong ProvinceMedical Research CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Xiaolin Wu
- Institute of Cerebrovascular DiseasesTaishan Scholars Construction Project Excellent Innovative Team of Shandong ProvinceMedical Research CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Yuqian Ren
- Institute of Cerebrovascular DiseasesTaishan Scholars Construction Project Excellent Innovative Team of Shandong ProvinceMedical Research CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Qinghua Zhang
- Department of NeurologyShandong Second Provincial General HospitalJinanChina
| | - Leiming Ren
- Institute of Chinese Integrative MedicineHebei Medical UniversityShijiazhuangChina
| | - Yunliang Guo
- Institute of Cerebrovascular DiseasesTaishan Scholars Construction Project Excellent Innovative Team of Shandong ProvinceMedical Research CenterThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| |
Collapse
|
37
|
Brain Protein Expression Profile Confirms the Protective Effect of the ACTH (4-7)PGP Peptide (Semax) in a Rat Model of Cerebral Ischemia-Reperfusion. Int J Mol Sci 2021; 22:ijms22126179. [PMID: 34201112 PMCID: PMC8226508 DOI: 10.3390/ijms22126179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/02/2021] [Accepted: 06/05/2021] [Indexed: 12/02/2022] Open
Abstract
The Semax (Met-Glu-His-Phe-Pro-Gly-Pro) peptide is a synthetic melanocortin derivative that is used in the treatment of ischemic stroke. Previously, studies of the molecular mechanisms underlying the actions of Semax using models of cerebral ischemia in rats showed that the peptide enhanced the transcription of neurotrophins and their receptors and modulated the expression of genes involved in the immune response. A genome-wide RNA-Seq analysis revealed that, in the rat transient middle cerebral artery occlusion (tMCAO) model, Semax suppressed the expression of inflammatory genes and activated the expression of neurotransmitter genes. Here, we aimed to evaluate the effect of Semax in this model via the brain expression profiling of key proteins involved in inflammation and cell death processes (MMP-9, c-Fos, and JNK), as well as neuroprotection and recovery (CREB) in stroke. At 24 h after tMCAO, we observed the upregulation of active CREB in subcortical structures, including the focus of the ischemic damage; downregulation of MMP-9 and c-Fos in the adjacent frontoparietal cortex; and downregulation of active JNK in both tissues under the action of Semax. Moreover, a regulatory network was constructed. In conclusion, the suppression of inflammatory and cell death processes and the activation of recovery may contribute to the neuroprotective action of Semax at both the transcriptome and protein levels.
Collapse
|
38
|
Khoramjouy M, Naderi N, Kobarfard F, Heidarli E, Faizi M. An Intensified Acrolein Exposure Can Affect Memory and Cognition in Rat. Neurotox Res 2021; 39:277-291. [PMID: 32876917 DOI: 10.1007/s12640-020-00278-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/22/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022]
Abstract
Acrolein is a clear, colorless liquid and a highly reactive α, β-unsaturated aldehyde. Acrolein, a byproduct and initiator of oxidative stress, has a major role in the pathogenesis of disorders including pulmonary, cardiovascular, atherosclerosis, and neurodegenerative diseases. Environmental or dietary exposure and endogenous production are common sources of acrolein. Widespread exposure to acrolein is a major risk for human health; therefore, we decided to investigate the neurological effects of acrolein. In this study, we used male Sprague-Dawley rats and exposed them orally to acrolein (0.5, 1, 3, and 5 mg/kg/day) for 90 days and investigated the neurobehavioral and electrophysiological disturbances. We also assessed the correlation between neurotoxicity and CSF concentration of acrolein in the rats. The results showed that chronic oral administration of acrolein at 5 mg/kg/day impaired learning and memory in the neurobehavioral tests. In addition, acrolein decreased the release of excitatory neurotransmitters such as glutamate in electrophysiological studies. Our data demonstrated that chronic oral exposure of acrolein at a dose of 5 mg/kg leads to a direct correlation between neurotoxicity and its CSF concentration. In conclusion, exposure to acrolein as a major pollutant in the environment may cause cognitive problems and may have serious neurocognitive effects on humans.
Collapse
Affiliation(s)
- Mona Khoramjouy
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, 2660 Vali-e-Asr Ave., Tehran, 19919-53381, Iran
| | - Nima Naderi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, 2660 Vali-e-Asr Ave., Tehran, 19919-53381, Iran
| | - Farzad Kobarfard
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elmira Heidarli
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, 2660 Vali-e-Asr Ave., Tehran, 19919-53381, Iran
| | - Mehrdad Faizi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, 2660 Vali-e-Asr Ave., Tehran, 19919-53381, Iran.
| |
Collapse
|
39
|
Dong Y, Hu C, Huang C, Gao J, Niu W, Wang D, Wang Y, Niu C. Interleukin-22 Plays a Protective Role by Regulating the JAK2-STAT3 Pathway to Improve Inflammation, Oxidative Stress, and Neuronal Apoptosis following Cerebral Ischemia-Reperfusion Injury. Mediators Inflamm 2021; 2021:6621296. [PMID: 33790691 PMCID: PMC7984880 DOI: 10.1155/2021/6621296] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/07/2021] [Accepted: 02/26/2021] [Indexed: 12/12/2022] Open
Abstract
The interleukins (ILs) are a pluripotent cytokine family that have been reported to regulate ischemic stroke and cerebral ischemia/reperfusion (I/R) injury. IL-22 is a member of the IL-10 superfamily and plays important roles in tissue injury and repair. However, the effects of IL-22 on ischemic stroke and cerebral I/R injury remain unclear. In the current study, we provided direct evidence that IL-22 treatment decreased infarct size, neurological deficits, and brain water content in mice subjected to cerebral I/R injury. IL-22 treatment remarkably reduced the expression of inflammatory cytokines, including IL-1β, monocyte chemotactic protein- (MCP-) 1, and tumor necrosis factor- (TNF-) α, both in serum and the ischemic cerebral cortex. In addition, IL-22 treatment also decreased oxidative stress and neuronal apoptosis in mice after cerebral I/R injury. Moreover, IL-22 treatment significantly increased Janus tyrosine kinase (JAK) 2 and signal transducer and activator of transcription (STAT) 3 phosphorylation levels in mice and PC12 cells, and STAT3 knockdown abolished the IL-22-mediated neuroprotective function. These findings suggest that IL-22 might be exploited as a potential therapeutic agent for ischemic stroke and cerebral I/R injury.
Collapse
Affiliation(s)
- Yongfei Dong
- Department of Neurosurgery, Anhui Provincial Hospital, Cheeloo College of Medicine, Shangdong University, Jinan, Shangdong, 250021, China
| | - Chengyun Hu
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Chunxia Huang
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Jie Gao
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Wanxiang Niu
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Di Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Yang Wang
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Chaoshi Niu
- Department of Neurosurgery, Anhui Provincial Hospital, Hefei, Anhui 230001, China
| |
Collapse
|
40
|
Shakova FM, Kirova YI, Silachev DN, Romanova GA, Morozov SG. Protective Effects of PGC-1α Activators on Ischemic Stroke in a Rat Model of Photochemically Induced Thrombosis. Brain Sci 2021; 11:325. [PMID: 33806692 PMCID: PMC8002020 DOI: 10.3390/brainsci11030325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 11/16/2022] Open
Abstract
The pharmacological induction and activation of peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α), a key regulator of ischemic brain tolerance, is a promising direction in neuroprotective therapy. Pharmacological agents with known abilities to modulate cerebral PGC-1α are scarce. This study focused on the potential PGC-1α-modulating activity of Mexidol (2-ethyl-6-methyl-3-hydroxypyridine succinate) and Semax (ACTH(4-7) analog) in a rat model of photochemical-induced thrombosis (PT) in the prefrontal cortex. Mexidol (100 mg/kg) was administered intraperitoneally, and Semax (25 μg/kg) was administered intranasally, for 7 days each. The expression of PGC-1α and PGC-1α-dependent protein markers of mitochondriogenesis, angiogenesis, and synaptogenesis was measured in the penumbra via immunoblotting at Days 1, 3, 7, and 21 after PT. The nuclear content of PGC-1α was measured immunohistochemically. The suppression of PGC-1α expression was observed in the penumbra from 24 h to 21 days following PT and reflected decreases in both the number of neurons and PGC-1α expression in individual neurons. Administration of Mexidol or Semax was associated with preservation of the neuron number and neuronal expression of PGC-1α, stimulation of the nuclear translocation of PGC-1α, and increased contents of protein markers for PGC-1α activation. This study opens new prospects for the pharmacological modulation of PGC-1α in the ischemic brain.
Collapse
Affiliation(s)
- Fatima M. Shakova
- Institute of General Pathology and Pathophysiology, Baltiyskaya Str. 8, 125315 Moscow, Russia; (Y.I.K.); (G.A.R.); (S.G.M.)
| | - Yuliya I. Kirova
- Institute of General Pathology and Pathophysiology, Baltiyskaya Str. 8, 125315 Moscow, Russia; (Y.I.K.); (G.A.R.); (S.G.M.)
| | - Denis N. Silachev
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, Leninskye Gory 1, Bldg. 40, 119992 Moscow, Russia;
- Histology, Embryology and Cytology Department, Peoples’ Friendship University of Russia, Miklukho-Maklaya Str. 6, 117198 Moscow, Russia
| | - Galina A. Romanova
- Institute of General Pathology and Pathophysiology, Baltiyskaya Str. 8, 125315 Moscow, Russia; (Y.I.K.); (G.A.R.); (S.G.M.)
| | - Sergey G. Morozov
- Institute of General Pathology and Pathophysiology, Baltiyskaya Str. 8, 125315 Moscow, Russia; (Y.I.K.); (G.A.R.); (S.G.M.)
| |
Collapse
|
41
|
De Lazzari F, Prag HA, Gruszczyk AV, Whitworth AJ, Bisaglia M. DJ-1: A promising therapeutic candidate for ischemia-reperfusion injury. Redox Biol 2021; 41:101884. [PMID: 33561740 PMCID: PMC7872972 DOI: 10.1016/j.redox.2021.101884] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/13/2021] [Accepted: 01/25/2021] [Indexed: 12/31/2022] Open
Abstract
DJ-1 is a multifaceted protein with pleiotropic functions that has been implicated in multiple diseases, ranging from neurodegeneration to cancer and ischemia-reperfusion injury. Ischemia is a complex pathological state arising when tissues and organs do not receive adequate levels of oxygen and nutrients. When the blood flow is restored, significant damage occurs over and above that of ischemia alone and is termed ischemia-reperfusion injury. Despite great efforts in the scientific community to ameliorate this pathology, its complex nature has rendered it challenging to obtain satisfactory treatments that translate to the clinic. In this review, we will describe the recent findings on the participation of the protein DJ-1 in the pathophysiology of ischemia-reperfusion injury, firstly introducing the features and functions of DJ-1 and, successively highlighting the therapeutic potential of the protein. DJ-1 has been shown to confer protection in ischemia-reperfusion injury models. DJ-1 protection relies on the activation of antioxidant signaling pathways. DJ-1 regulates mitochondrial homeostasis during ischemia and reperfusion. DJ-1 seems to modulate ion homeostasis during ischemia and reperfusion. DJ-1 may represent a promising therapeutic target for ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Federica De Lazzari
- Physiology, Genetics and Behaviour Unit, Department of Biology, University of Padova, 35131, Padova, Italy
| | - Hiran A Prag
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Anja V Gruszczyk
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Alexander J Whitworth
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Marco Bisaglia
- Physiology, Genetics and Behaviour Unit, Department of Biology, University of Padova, 35131, Padova, Italy.
| |
Collapse
|
42
|
Woo PYM, Ho JWK, Ko NMW, Li RPT, Jian L, Chu ACH, Kwan MCL, Chan Y, Wong AKS, Wong HT, Chan KY, Kwok JCK. Randomized, placebo-controlled, double-blind, pilot trial to investigate safety and efficacy of Cerebrolysin in patients with aneurysmal subarachnoid hemorrhage. BMC Neurol 2020; 20:401. [PMID: 33143640 PMCID: PMC7607674 DOI: 10.1186/s12883-020-01908-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 08/24/2020] [Indexed: 11/10/2022] Open
Abstract
Background There are limited neuroprotective treatment options for patients with aneurysmal subarachnoid hemorrhage (SAH). Cerebrolysin, a brain-specific proposed pleiotropic neuroprotective agent, has been suggested to improve global functional outcomes in ischemic stroke. We investigated the efficacy, safety and feasibility of administering Cerebrolysin for SAH patients. Methods This was a prospective, randomized, double-blind, placebo-controlled, single-center, parallel-group pilot study. Fifty patients received either daily Cerebrolysin (30 ml/day) or a placebo (saline) for 14 days (25 patients per study group). The primary endpoint was a favorable Extended Glasgow Outcome Scale (GOSE) of 5 to 8 (moderate disability to good recovery) at six-months. Secondary endpoints included the modified Ranking Scale (mRS), the Montreal Cognitive Assessment (MOCA) score, occurrence of adverse effects and the occurrence of delayed cerebral ischemia (DCI). Results No severe adverse effects or mortality attributable to Cerebrolysin were observed. No significant difference was detected in the proportion of patients with favorable six-month GOSE in either study group (odds ratio (OR): 1.49; 95% confidence interval (CI): 0.43–5.17). Secondary functional outcome measures for favorable six-month recovery i.e. a mRS of 0 to 3 (OR: 3.45; 95% CI 0.79–15.01) were comparable for both groups. Similarly, there was no difference in MOCA neurocognitive performance (p-value: 0.75) and in the incidence of DCI (OR: 0.85 95% CI: 0.28–2.59). Conclusions Use of Cerebrolysin in addition to standard-of-care management of aneurysmal SAH is safe, well tolerated and feasible. However, the neutral results of this trial suggest that it does not improve the six-month global functional performance of patients. Clinical trial registration Name of Registry: ClinicalTrials.gov Trial Registration Number: NCT01787123. Date of Registration: 8th February 2013.
Collapse
Affiliation(s)
- Peter Y M Woo
- Department of Neurosurgery, Kwong Wah Hospital, Room CS11-01, 11th Floor, 25 Waterloo Road, Yaumatei, Hong Kong, China.
| | - Joanna W K Ho
- Department of Neurosurgery, Kwong Wah Hospital, Room CS11-01, 11th Floor, 25 Waterloo Road, Yaumatei, Hong Kong, China
| | - Natalie M W Ko
- Department of Neurosurgery, Kwong Wah Hospital, Room CS11-01, 11th Floor, 25 Waterloo Road, Yaumatei, Hong Kong, China
| | - Ronald P T Li
- Department of Neurosurgery, Kwong Wah Hospital, Room CS11-01, 11th Floor, 25 Waterloo Road, Yaumatei, Hong Kong, China
| | - Leo Jian
- Department of Neurosurgery, Kwong Wah Hospital, Room CS11-01, 11th Floor, 25 Waterloo Road, Yaumatei, Hong Kong, China
| | - Alberto C H Chu
- Department of Neurosurgery, Kwong Wah Hospital, Room CS11-01, 11th Floor, 25 Waterloo Road, Yaumatei, Hong Kong, China
| | - Marco C L Kwan
- Department of Neurosurgery, Kwong Wah Hospital, Room CS11-01, 11th Floor, 25 Waterloo Road, Yaumatei, Hong Kong, China
| | - Yung Chan
- Department of Neurosurgery, Kwong Wah Hospital, Room CS11-01, 11th Floor, 25 Waterloo Road, Yaumatei, Hong Kong, China
| | - Alain K S Wong
- Department of Neurosurgery, Kwong Wah Hospital, Room CS11-01, 11th Floor, 25 Waterloo Road, Yaumatei, Hong Kong, China
| | - Hoi-Tung Wong
- Department of Neurosurgery, Kwong Wah Hospital, Room CS11-01, 11th Floor, 25 Waterloo Road, Yaumatei, Hong Kong, China
| | - Kwong-Yau Chan
- Department of Neurosurgery, Kwong Wah Hospital, Room CS11-01, 11th Floor, 25 Waterloo Road, Yaumatei, Hong Kong, China
| | - John C K Kwok
- Department of Neurosurgery, Kwong Wah Hospital, Room CS11-01, 11th Floor, 25 Waterloo Road, Yaumatei, Hong Kong, China
| |
Collapse
|
43
|
Sapio L, Salzillo A, Ragone A, Illiano M, Spina A, Naviglio S. Targeting CREB in Cancer Therapy: A Key Candidate or One of Many? An Update. Cancers (Basel) 2020; 12:cancers12113166. [PMID: 33126560 PMCID: PMC7693618 DOI: 10.3390/cancers12113166] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Only 5% of all drug-related targets currently move from preclinical to clinical in cancer, and just some of them achieve patient’s bedside. Among others, intratumor heterogeneity and preclinical cancer model limitations actually represent the main reasons for this failure. Cyclic-AMP response element-binding protein (CREB) has been defined as a proto-oncogene in different tumor types, being involved in maintenance and progression. Due to its relevance in tumor pathophysiology, many CREB inhibitor compounds have been developed and tested over the years. Herein, we examine the current state-of-the-art of both CREB and CREB inhibitors in cancer, retracing some of the most significant findings of the last years. While the scientific statement confers on CREB a proactive role in cancer, its therapeutic potential is still stuck at laboratory bench. Therefore, pursuing every concrete result to achieve CREB inhibition in clinical might give chance and future to cancer patients worldwide. Abstract Intratumor heterogeneity (ITH) is considered the major disorienting factor in cancer treatment. As a result of stochastic genetic and epigenetic alterations, the appearance of a branched evolutionary shape confers tumor plasticity, causing relapse and unfavorable clinical prognosis. The growing evidence in cancer discovery presents to us “the great paradox” consisting of countless potential targets constantly discovered and a small number of candidates being effective in human patients. Among these, cyclic-AMP response element-binding protein (CREB) has been proposed as proto-oncogene supporting tumor initiation, progression and metastasis. Overexpression and hyperactivation of CREB are frequently observed in cancer, whereas genetic and pharmacological CREB downregulation affects proliferation and apoptosis. Notably, the present review is designed to investigate the feasibility of targeting CREB in cancer therapy. In particular, starting with the latest CREB evidence in cancer pathophysiology, we evaluate the advancement state of CREB inhibitor design, including the histone lysine demethylases JMJD3/UTX inhibitor GSKJ4 that we newly identified as a promising CREB modulator in leukemia cells. Moreover, an accurate analysis of strengths and weaknesses is also conducted to figure out whether CREB can actually represent a therapeutic candidate or just one of the innumerable preclinical cancer targets.
Collapse
|
44
|
Wang SW, Wang W, Sheng H, Bai YF, Weng YY, Fan XY, Zheng F, Zhu XT, Xu ZC, Zhang F. Hesperetin, a SIRT1 activator, inhibits hepatic inflammation via AMPK/CREB pathway. Int Immunopharmacol 2020; 89:107036. [PMID: 33068864 DOI: 10.1016/j.intimp.2020.107036] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/05/2020] [Accepted: 09/21/2020] [Indexed: 12/31/2022]
Abstract
Silent mating type information regulation 2 homolog 1 (SIRT1) is an important inflammatory regulator, which epigenetically reprograms inflammation by altering the acetylation of NF-κB. Hesperetin, as a common flavonoid, has been proven to have a significant effect on acute inflammatory diseases. However, the detailed molecular mechanism by which hesperetin alleviates inflammatory response and accompanied tissue injury is poorly understood. Our results show that SIRT1 is required for the inhibitory effect of hesperetin on inflammation. Hesperetin suppresses the acetylation of RelA/p65 to reduce NF-κB activity by inducing SIRT1 expression. Mechanistically, hesperetin increases SIRT1 expression through AMPK/CREB pathway. Additionally, the protective effect of hesperetin against LPS/D-GalN-induced hepatitis in mice is also dependent on SIRT1. Our study suggests that hesperetin is an SIRT1 activator and could be potential candidates for the treatments of inflammatory conditions.
Collapse
Affiliation(s)
- Si-Wei Wang
- Department of Core Facility, Affiliated Quzhou Central Hospital, Zhejiang Chinese Medical University, Quzhou 324000, China
| | - Wen Wang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, China; Department of Acupuncture, Quzhou Municiple Hospital of Traditonal Chinese Medicine, Quzhou 324000, China
| | - Hao Sheng
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yong-Feng Bai
- Department of Clinical Laboratory, Affiliated Quzhou Central Hospital, Zhejiang Chinese Medical University, Quzhou 324000, China
| | - Yuan-Yuan Weng
- Department of Clinical Laboratory, Affiliated Quzhou Central Hospital, Zhejiang Chinese Medical University, Quzhou 324000, China
| | - Xue-Yu Fan
- Department of Clinical Laboratory, Affiliated Quzhou Central Hospital, Zhejiang Chinese Medical University, Quzhou 324000, China
| | - Fang Zheng
- Department of Core Facility, Affiliated Quzhou Central Hospital, Zhejiang Chinese Medical University, Quzhou 324000, China
| | - Xin-Tian Zhu
- Wenzhou Medical University, Wenzhou 325000, China
| | - Zheng-Cai Xu
- Department of Clinical Laboratory, Affiliated Quzhou Central Hospital, Zhejiang Chinese Medical University, Quzhou 324000, China
| | - Feng Zhang
- Department of Core Facility, Affiliated Quzhou Central Hospital, Zhejiang Chinese Medical University, Quzhou 324000, China; Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Clinical Laboratory, Affiliated Quzhou Central Hospital, Zhejiang Chinese Medical University, Quzhou 324000, China.
| |
Collapse
|
45
|
Gavrilova SI, Alvarez A. Cerebrolysin in the therapy of mild cognitive impairment and dementia due to Alzheimer's disease: 30 years of clinical use. Med Res Rev 2020; 41:2775-2803. [PMID: 32808294 DOI: 10.1002/med.21722] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/01/2020] [Accepted: 08/05/2020] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is the most common neurocognitive disorder and a global health problem. The prevalence of AD is growing dramatically, especially in low- and middle-income countries, and will reach 131.5 million cases worldwide by 2050. Therefore, developing a disease-modifying therapy capable of delaying or even preventing the onset and progression of AD has become a world priority, and is an unmet need. The pathogenesis of AD, considered as the result of an imbalance between resilience and risk factors, begins many years before the typical clinical picture develops and involves multiple pathophysiological mechanisms. Since the pathophysiology of AD is multifactorial, it is not surprising that all attempts done to modify the disease course with drugs directed towards a single therapeutic target have been unsuccessful. Thus, combined modality therapy, using multiple drugs with a single mechanism of action or multi-target drugs, appears as the most promising strategy for both effective AD therapy and prevention. Cerebrolysin, acting as a multitarget peptidergic drug with a neurotrophic mode of action, exerts long-lasting therapeutic effects on AD that could reflect its potential utility for disease modification. Clinical trials demonstrated that Cerebrolysin is safe and efficacious in the treatment of AD, and may enhance and prolong the efficacy of cholinergic drugs, particularly in moderate to advanced AD patients. In this review, we summarize advances of therapeutic relevance in the pathogenesis and the biomarkers of AD, paying special attention to neurotrophic factors, and present results of preclinical and clinical investigations with Cerebrolysin in AD.
Collapse
Affiliation(s)
- Svetlana I Gavrilova
- Department of Geriatric Psychiatry, Cognitive Disorders and Alzheimer's Disease Unit, Mental Health Research Center, Moscow, Russia
| | - Anton Alvarez
- Department of Neuropsychiatry, Medinova Institute of Neurosciences, Clinica RehaSalud, A Coruña, Spain.,Clinical Research Department, QPS Holdings, A Coruña, Spain
| |
Collapse
|
46
|
Sepehr A, Taheri F, Heidarian S, Motaghinejad M, Safari S. Neuroprotective and neuro-survival properties of safinamide against methamphetamine-induced neurodegeneration: Hypothetic possible role of BDNF/TrkB/PGC-1α signaling pathway and mitochondrial uncoupling protein -2(UCP-2). Med Hypotheses 2020; 143:110094. [PMID: 32682215 DOI: 10.1016/j.mehy.2020.110094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 06/19/2020] [Accepted: 07/07/2020] [Indexed: 12/16/2022]
Abstract
Methamphetamine is a behavioral psychostimulant that has a high potential for misuse and induction of neurotoxicity. Safinamide is a novel inhibitor of monoamine oxidase B (MAOB) with neuroprotective properties. Methamphetamine abuse causes dysfunction in the respiratory chain of the mitochondria, but the specific signaling mechanism and role of the uncoupling protein-2(UCP-2) remain unclear. As we know, some indirect evidence indicates that neurodegeneration can be caused by inhibition of the brain-derived neurotrophic factor (BDNF) receptor, TrkB and its downstream signaling pathway, such as the PGC-1α protein. Neuroprotective strategies and approaches to the management, treatment or prevention of methamphetamine-induced neurodegeneration by modulating BDNF / TrkB / PGC-1α-UCP-2 can be considered as novel therapeutic approaches to these psychostimulant neurochemical and neurobehavioral approaches. Previous studies have shown that safinamide, a monoamine oxidase-B (MAOB) inhibitor, can function as a neuroprotective agent and inhibit the neurodegenerative process especially in Parkinson's disease but its impact on other neurodegenerative processes and drug-induced neurotoxicity remain unclear. Although there is some evidence that BDNF / TrkB / PGC-1α-UCP-2 signaling pathway and mitochondrial UCP-2 mediated safinamide induced neuroprotection but it's exact and precise mechanism of action and neuroprotective effects in neurodegenerative disorder and the protective properties against methamphetamine induced neurodegeneration and the role of BDNF / TrkB / PGC-1α signaling pathway and role of mitochondrial UCP-2 in this process have not yet been clarified. Therefore, in subjects addicted to methamphetamine, we hypothesized that safinamide will provide neuroprotection against methamphetamine-prompted neurodegeneration, and it appears that BDNF / TrkB / PGC-1α signaling pathway and mitochondrial UCP-2 are likely to play a critical role.
Collapse
Affiliation(s)
- Afrah Sepehr
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Taheri
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Heidarian
- Department of chemistry, Zagros Institute of Higher Education, Kermanshah, Iran
| | - Majid Motaghinejad
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Sepideh Safari
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
47
|
Hu X, Li S, Doycheva DM, Huang L, Lenahan C, Liu R, Huang J, Xie S, Tang J, Zuo G, Zhang JH. Rh-CSF1 attenuates neuroinflammation via the CSF1R/PLCG2/PKCε pathway in a rat model of neonatal HIE. J Neuroinflammation 2020; 17:182. [PMID: 32522286 PMCID: PMC7285566 DOI: 10.1186/s12974-020-01862-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/29/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Hypoxic-ischemic encephalopathy (HIE) is a life-threatening cerebrovascular disease. Neuroinflammation plays an important role in the pathogenesis of HIE, in which microglia are key cellular mediators in the regulation of neuroinflammatory processes. Colony-stimulating factor 1 (CSF1), a specific endogenous ligand of CSF1 receptor (CSF1R), is crucial in microglial growth, differentiation, and proliferation. Recent studies showed that the activation of CSF1R with CSF1 exerted anti-inflammatory effects in a variety of nervous system diseases. This study aimed to investigate the anti-inflammatory effects of recombinant human CSF1 (rh-CSF1) and the underlying mechanisms in a rat model of HIE. METHODS A total of 202 10-day old Sprague Dawley rat pups were used. HI was induced by the right common carotid artery ligation with subsequent exposure of 2.5-h hypoxia. At 1 h and 24 h after HI induction, exogenous rh-CSF1 was administered intranasally. To explore the underlying mechanism, CSF1R inhibitor, BLZ945, and phospholipase C-gamma 2 (PLCG2) inhibitor, U73122, were injected intraperitoneally at 1 h before HI induction, respectively. Brain infarct area, brain water content, neurobehavioral tests, western blot, and immunofluorescence staining were performed. RESULTS The expressions of endogenous CSF1, CSF1R, PLCG2, protein kinase C epsilon type (PKCε), and cAMP response element-binding protein (CREB) were gradually increased after HIE. Rh-CSF1 significantly improved the neurological deficits at 48 h and 4 weeks after HI, which was accompanied by a reduction in the brain infarct area, brain edema, brain atrophy, and neuroinflammation. Moreover, activation of CSF1R by rh-CSF1 significantly increased the expressions of p-PLCG2, p-PKCε, and p-CREB, but inhibited the activation of neutrophil infiltration, and downregulated the expressions of IL-1β and TNF-α. Inhibition of CSF1R and PLCG2 abolished these neuroprotective effects of rh-CSF1 after HI. CONCLUSIONS Our findings demonstrated that the activation of CSF1R by rh-CSF1 attenuated neuroinflammation and improved neurological deficits after HI. The anti-inflammatory effects of rh-CSF1 partially acted through activating the CSF1R/PLCG2/PKCε/CREB signaling pathway after HI. These results suggest that rh-CSF1 may serve as a potential therapeutic approach to ameliorate injury in HIE patients.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, 550002, China.,Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Shirong Li
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, 550002, China.,Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Desislava Met Doycheva
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Lei Huang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Cameron Lenahan
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA.,Bvrrell College of Osteopathic Medicine, Las Cruces, NM, 88003, USA
| | - Rui Liu
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, 550002, China.,Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Juan Huang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA.,Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China
| | - Shucai Xie
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA.,Department of Hepatobiliary Surgery, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, 570208, Hainan, China
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA
| | - Gang Zuo
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA. .,Department of Neurosurgery, Taicang Hospital Affiliated to Soochow University, Taicang, Suzhou, 215400, Jiangsu, China.
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, Room 219, 11041 Campus Street, Loma Linda, CA, 92350, USA. .,Department of Neurosurgery, Loma Linda University, Loma Linda, CA, 92350, USA. .,Department of Anesthesiology, Loma Linda University, Loma Linda, CA, 92350, USA.
| |
Collapse
|