1
|
Hao T, Jiang G, Lin C, Boyer C, Huang R. Advanced oral breviscapine sustained-release tablets for improved ischemic stroke treatment. Biomaterials 2025; 316:123030. [PMID: 39705923 DOI: 10.1016/j.biomaterials.2024.123030] [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: 08/23/2024] [Revised: 11/21/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
This study aimed to address the challenges associated with the low oral bioavailability and the necessity for frequent dosing of breviscapine (BRE), a mainstream drug in the treatment of cardiovascular and cerebrovascular diseases. The poor solubility and permeability of BRE in the gastrointestinal tract were identified as significant barriers to effective drug absorption, thereby impacting therapeutic efficacy and patient compliance. To enhance the gastrointestinal absorption of BRE, particles loaded with BRE were engineered utilizing Cremophor EL (CrEL), an absorption enhancer, in conjunction with mesoporous silica, a biocompatible drug delivery vector, formulating mesoporous silica particles loaded with BRE and CrEL (BRE-CrEL@SiO2). The solubility and mucosal permeability of BRE were ameliorated, facilitating transepithelial transport and improving absorption kinetics. BRE-CrEL@SiO2 were subsequently integrated to prepare sustained-release tablets. The finite element simulation method was utilized in the study of non-planar circular BRE tablet process to ensure tablet quality. The superior bioavailability and therapeutic index of the absorption-promoting sustained-release tablets, compared to commercial tablets, were validated through in vivo pharmacokinetic and pharmacodynamic assessments, while safety was maintained. The oral relative bioavailability of the absorption-enhancing sustained-release tablets was 160.7 % relative to the commercial tablets, demonstrated in Beagle dogs, indicating higher absorption. This innovative formulation represents a significant advancement in improving therapeutic efficacy of ischemic stroke and reducing the treatment burden on patients. The study provides new insights into the development of novel dosage forms for BRE and other drugs with poor solubility and permeability, suggesting a promising approach to enhance their therapeutic effectiveness and improve patient compliance in treatment.
Collapse
Affiliation(s)
- Tingting Hao
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education), Fudan University, Shanghai, 201203, China
| | - Guangwei Jiang
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education), Fudan University, Shanghai, 201203, China
| | - Chenteng Lin
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education), Fudan University, Shanghai, 201203, China
| | - Cyrille Boyer
- Australian Centre for Nanomedicine, School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Rongqin Huang
- School of Pharmacy, Key Laboratory of Smart Drug Delivery (Ministry of Education), Fudan University, Shanghai, 201203, China.
| |
Collapse
|
2
|
Jayanti S, Vitek L, Verde CD, Llido JP, Sukowati C, Tiribelli C, Gazzin S. Role of Natural Compounds Modulating Heme Catabolic Pathway in Gut, Liver, Cardiovascular, and Brain Diseases. Biomolecules 2024; 14:63. [PMID: 38254662 PMCID: PMC10813662 DOI: 10.3390/biom14010063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/24/2024] Open
Abstract
The crucial physiological process of heme breakdown yields biliverdin (BV) and bilirubin (BR) as byproducts. BV, BR, and the enzymes involved in their production (the "yellow players-YP") are increasingly documented as endogenous modulators of human health. Mildly elevated serum bilirubin concentration has been correlated with a reduced risk of multiple chronic pro-oxidant and pro-inflammatory diseases, especially in the elderly. BR and BV per se have been demonstrated to protect against neurodegenerative diseases, in which heme oxygenase (HMOX), the main enzyme in the production of pigments, is almost always altered. HMOX upregulation has been interpreted as a tentative defense against the ongoing pathologic mechanisms. With the demonstration that multiple cells possess YP, their propensity to be modulated, and their broad spectrum of activity on multiple signaling pathways, the YP have assumed the role of an adjustable system that can promote health in adults. Based on that, there is an ongoing effort to induce their activity as a therapeutic option, and natural compounds are an attractive alternative to the goal, possibly requiring only minimal changes in the life style. We review the most recent evidence of the potential of natural compounds in targeting the YP in the context of the most common pathologic condition of adult and elderly life.
Collapse
Affiliation(s)
- Sri Jayanti
- Liver brain Unit “Rita Moretti”, Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163,5, Basovizza, 34149 Trieste, Italy or (S.J.); (C.D.V.); (J.P.L.); or (C.S.); (C.T.)
- Eijkman Research Centre for Molecular Biology, Research Organization for Health, National Research and Innovation Agency, Cibinong 16915, Indonesia
| | - Libor Vitek
- Institute of Medical Biochemistry and Laboratory Diagnostics, and 4th Department of Internal Medicine, General University Hospital and 1st Faculty of Medicine, Charles University, 12000 Prague, Czech Republic;
| | - Camilla Dalla Verde
- Liver brain Unit “Rita Moretti”, Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163,5, Basovizza, 34149 Trieste, Italy or (S.J.); (C.D.V.); (J.P.L.); or (C.S.); (C.T.)
- Department of Life Sciences, University of Trieste, 34139 Trieste, Italy
| | - John Paul Llido
- Liver brain Unit “Rita Moretti”, Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163,5, Basovizza, 34149 Trieste, Italy or (S.J.); (C.D.V.); (J.P.L.); or (C.S.); (C.T.)
- Department of Life Sciences, University of Trieste, 34139 Trieste, Italy
- Department of Science and Technology, Philippine Council for Health Research and Development, Bicutan, Taguig City 1631, Philippines
| | - Caecilia Sukowati
- Liver brain Unit “Rita Moretti”, Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163,5, Basovizza, 34149 Trieste, Italy or (S.J.); (C.D.V.); (J.P.L.); or (C.S.); (C.T.)
- Eijkman Research Centre for Molecular Biology, Research Organization for Health, National Research and Innovation Agency, Cibinong 16915, Indonesia
| | - Claudio Tiribelli
- Liver brain Unit “Rita Moretti”, Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163,5, Basovizza, 34149 Trieste, Italy or (S.J.); (C.D.V.); (J.P.L.); or (C.S.); (C.T.)
| | - Silvia Gazzin
- Liver brain Unit “Rita Moretti”, Fondazione Italiana Fegato-Onlus, Bldg. Q, AREA Science Park, ss14, Km 163,5, Basovizza, 34149 Trieste, Italy or (S.J.); (C.D.V.); (J.P.L.); or (C.S.); (C.T.)
| |
Collapse
|
3
|
Wan C, Pei J, Wang D, Hu J, Tang Z, Zhao W. Identification of m 6A methylation-related genes in cerebral ischaemia‒reperfusion of Breviscapus therapy based on bioinformatics methods. BMC Med Genomics 2023; 16:210. [PMID: 37670341 PMCID: PMC10478429 DOI: 10.1186/s12920-023-01651-3] [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: 05/05/2023] [Accepted: 08/29/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND Cerebral ischaemia‒reperfusion (I/R) frequently causes late-onset neuronal damage. Breviscapine promotes autophagy in microvascular endothelial cells in I/R and can inhibit oxidative damage and apoptosis. However, the mediation mechanism of breviscapine on neuronal cell death is unclear. METHODS First, transcriptome sequencing was performed on three groups of mice: the neuronal normal group (Control group), the oxygen-glucose deprivation/ reoxygenation group (OGD/R group) and the breviscapine administration group (Therapy group). Differentially expressed genes (DEGs) between the OGD/R and control groups and between the Therapy and OGD/R groups were obtained by the limma package. N6-methyladenosine (m6A) methylation-related DEGs were selected by Pearson correlation analysis. Then, prediction and confirmation of drug targets were performed by Swiss Target Prediction and UniProt Knowledgebase (UniProtKB) database, and key genes were obtained by Pearson correlation analysis between m6A-related DEGs and drug target genes. Next, gene set enrichment analysis (GSEA) and Ingenuity pathway analysis (IPA) were used to obtain the pathways of key genes. Finally, a circRNA-miRNA‒mRNA network was constructed based on the mRNAs, circRNAs and miRNAs. RESULTS A total of 2250 DEGs between the OGD/R and control groups and 757 DEGs between the Therapy and OGD/R groups were selected by differential analysis. A total of 7 m6A-related DEGs, including Arl4d, Gm10653, Gm1113, Kcns3, Olfml2a, Stk26 and Tfcp2l1, were obtained by Pearson correlation analysis. Four key genes (Tfcp2l1, Kcns3, Olfml2a and Arl4d) were acquired, and GSEA showed that these key genes significantly participated in DNA repair, e2f targets and the g2m checkpoint. IPA revealed that Tfcp2l1 played a significant role in human embryonic stem cell pluripotency. The circRNA-miRNA‒mRNA network showed that mmu_circ_0001258 regulated Tfcp2l1 by mmu-miR-301b-3p. CONCLUSIONS In conclusion, four key genes, Tfcp2l1, Kcns3, Olfml2a and Arl4d, significantly associated with the treatment of OGD/R by breviscapine were identified, which provides a theoretical basis for clinical trials.
Collapse
Affiliation(s)
- Cheng Wan
- Department of Interventional Radiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Jingchun Pei
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Dan Wang
- Department of Organ Transplantation Centre, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Jihong Hu
- Department of Interventional Radiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Zhiwei Tang
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China.
| | - Wei Zhao
- Department of Interventional Radiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China.
| |
Collapse
|
4
|
Niu P, Liu F, Lei F, Peng J, Wang Y, Zhao J, Gao Z, Gao Q, Gu J. Breviscapine regulates the proliferation, migration, invasion, and apoptosis of colorectal cancer cells via the PI3K/AKT pathway. Sci Rep 2023; 13:9674. [PMID: 37316553 DOI: 10.1038/s41598-023-33792-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 04/19/2023] [Indexed: 06/16/2023] Open
Abstract
Colorectal cancer (CRC) is ranked as one of the most common malignancies with a high death rate. It has been discovered that breviscapine can alter the progression and development of various cancers. Nevertheless, the function and mechanisms of breviscapine in CRC progression have not yet been described. The cell proliferation capacity of HCT116 and SW480 cells was assessed using the CCK-8 and EdU assays. Cell apoptosis was tested through flow cytometry, and cell migration and invasion were examined using the transwell assay. Moreover, protein expression was examined through a western blot. Tumor weight and volume were assessed using the nude mice in vivo assay, and the Ki-67 protein expression was verified through the IHC assay. This study discovered that an increased dose of breviscapine (0, 12.5, 25, 50, 100, 200, and 400 μM) gradually reduced cell proliferation and increased apoptosis in CRC. Additionally, breviscapine restricted the migration and invasion CRC cells. Moreover, it was revealed that breviscapine inactivated the PI3K/AKT pathway and inhibited CRC progression. Finally, an in vivo assay demonstrated that breviscapine restrained tumor growth in vivo. It affected the CRC cells' proliferation, migration, invasion, and apoptosis through the PI3K/AKT pathway. This discovery may offer new insights into CRC treatment.
Collapse
Affiliation(s)
- Pengfei Niu
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No. 9, Jinyuanzhuang Road, The Shijingshan District, Beijing, 100144, China
| | - Feng Liu
- Beijing Viewsolid Biotechnology Co., Ltd., Beijing, 100195, China
| | - Fuming Lei
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No. 9, Jinyuanzhuang Road, The Shijingshan District, Beijing, 100144, China
| | - Jisheng Peng
- Department of Traditional Chinese Medicine, Peking University Shougang Hospital, Beijing, 100144, China
| | - Yanzhao Wang
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No. 9, Jinyuanzhuang Road, The Shijingshan District, Beijing, 100144, China
| | - Jun Zhao
- Department of Traditional Chinese Medicine, Peking University Shougang Hospital, Beijing, 100144, China
| | - Zhaoya Gao
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No. 9, Jinyuanzhuang Road, The Shijingshan District, Beijing, 100144, China
| | - Qingkun Gao
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No. 9, Jinyuanzhuang Road, The Shijingshan District, Beijing, 100144, China
| | - Jin Gu
- Department of Gastrointestinal Surgery, Peking University Shougang Hospital, No. 9, Jinyuanzhuang Road, The Shijingshan District, Beijing, 100144, China.
| |
Collapse
|
5
|
King H, Reiber M, Philippi V, Stirling H, Aulehner K, Bankstahl M, Bleich A, Buchecker V, Glasenapp A, Jirkof P, Miljanovic N, Schönhoff K, von Schumann L, Leenaars C, Potschka H. Anesthesia and analgesia for experimental craniotomy in mice and rats: a systematic scoping review comparing the years 2009 and 2019. Front Neurosci 2023; 17:1143109. [PMID: 37207181 PMCID: PMC10188949 DOI: 10.3389/fnins.2023.1143109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/27/2023] [Indexed: 05/21/2023] Open
Abstract
Experimental craniotomies are a common surgical procedure in neuroscience. Because inadequate analgesia appears to be a problem in animal-based research, we conducted this review and collected information on management of craniotomy-associated pain in laboratory mice and rats. A comprehensive search and screening resulted in the identification of 2235 studies, published in 2009 and 2019, describing craniotomy in mice and/or rats. While key features were extracted from all studies, detailed information was extracted from a random subset of 100 studies/year. Reporting of perioperative analgesia increased from 2009 to 2019. However, the majority of studies from both years did not report pharmacologic pain management. Moreover, reporting of multimodal treatments remained at a low level, and monotherapeutic approaches were more common. Among drug groups, reporting of pre- and postoperative administration of non-steroidal anti-inflammatory drugs, opioids, and local anesthetics in 2019 exceeded that of 2009. In summary, these results suggest that inadequate analgesia and oligoanalgesia are persistent issues associated with experimental intracranial surgery. This underscores the need for intensified training of those working with laboratory rodents subjected to craniotomies. Systematic review registration https://osf.io/7d4qe.
Collapse
Affiliation(s)
- Hannah King
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Maria Reiber
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Vanessa Philippi
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Helen Stirling
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Katharina Aulehner
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Marion Bankstahl
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - André Bleich
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Verena Buchecker
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Aylina Glasenapp
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Paulin Jirkof
- Office for Animal Welfare and 3Rs, University of Zurich, Zurich, Switzerland
| | - Nina Miljanovic
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Katharina Schönhoff
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Lara von Schumann
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
| | - Cathalijn Leenaars
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Heidrun Potschka
- Institute of Pharmacology, Toxicology, and Pharmacy, Ludwig Maximilian University of Munich, Munich, Germany
- *Correspondence: Heidrun Potschka,
| |
Collapse
|
6
|
Silvestro S, Mazzon E. Nrf2 Activation: Involvement in Central Nervous System Traumatic Injuries. A Promising Therapeutic Target of Natural Compounds. Int J Mol Sci 2022; 24:199. [PMID: 36613649 PMCID: PMC9820431 DOI: 10.3390/ijms24010199] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Central nervous system (CNS) trauma, such as traumatic brain injury (TBI) and spinal cord injury (SCI), represents an increasingly important health burden in view of the preventability of most injuries and the complex and expensive medical care that they necessitate. These injuries are characterized by different signs of neurodegeneration, such as oxidative stress, mitochondrial dysfunction, and neuronal apoptosis. Cumulative evidence suggests that the transcriptional factor nuclear factor erythroid 2-related factor 2 (Nrf2) plays a crucial defensive role in regulating the antioxidant response. It has been demonstrated that several natural compounds are able to activate Nrf2, mediating its antioxidant response. Some of these compounds have been tested in experimental models of SCI and TBI, showing different neuroprotective properties. In this review, an overview of the preclinical studies that highlight the positive effects of natural bioactive compounds in SCI and TBI experimental models through the activation of the Nrf2 pathway has been provided. Interestingly, several natural compounds can activate Nrf2 through multiple pathways, inducing a strong antioxidant response against CNS trauma. Therefore, some of these compounds could represent promising therapeutic strategies for these pathological conditions.
Collapse
Affiliation(s)
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| |
Collapse
|
7
|
Targeting Nrf2-Mediated Oxidative Stress Response in Traumatic Brain Injury: Therapeutic Perspectives of Phytochemicals. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1015791. [PMID: 35419162 PMCID: PMC9001080 DOI: 10.1155/2022/1015791] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/22/2021] [Accepted: 03/19/2022] [Indexed: 02/07/2023]
Abstract
Traumatic brain injury (TBI), known as mechanical damage to the brain, impairs the normal function of the brain seriously. Its clinical symptoms manifest as behavioral impairment, cognitive decline, communication difficulties, etc. The pathophysiological mechanisms of TBI are complex and involve inflammatory response, oxidative stress, mitochondrial dysfunction, blood-brain barrier (BBB) disruption, and so on. Among them, oxidative stress, one of the important mechanisms, occurs at the beginning and accompanies the whole process of TBI. Most importantly, excessive oxidative stress causes BBB disruption and brings injury to lipids, proteins, and DNA, leading to the generation of lipid peroxidation, damage of nuclear and mitochondrial DNA, neuronal apoptosis, and neuroinflammatory response. Transcription factor NF-E2 related factor 2 (Nrf2), a basic leucine zipper protein, plays an important role in the regulation of antioxidant proteins, such as oxygenase-1(HO-1), NAD(P)H Quinone Dehydrogenase 1 (NQO1), and glutathione peroxidase (GPx), to protect against oxidative stress, neuroinflammation, and neuronal apoptosis. Recently, emerging evidence indicated the knockout (KO) of Nrf2 aggravates the pathology of TBI, while the treatment of Nrf2 activators inhibits neuronal apoptosis and neuroinflammatory responses via reducing oxidative damage. Phytochemicals from fruits, vegetables, grains, and other medical herbs have been demonstrated to activate the Nrf2 signaling pathway and exert neuroprotective effects in TBI. In this review, we emphasized the contributive role of oxidative stress in the pathology of TBI and the protective mechanism of the Nrf2-mediated oxidative stress response for the treatment of TBI. In addition, we summarized the research advances of phytochemicals, including polyphenols, terpenoids, natural pigments, and otherwise, in the activation of Nrf2 signaling and their potential therapies for TBI. Although there is still limited clinical application evidence for these natural Nrf2 activators, we believe that the combinational use of phytochemicals such as Nrf2 activators with gene and stem cell therapy will be a promising therapeutic strategy for TBI in the future.
Collapse
|
8
|
Feng Y, Ju Y, Yan Z, Ji M, Yang M, Wu Q, Wang L, Sun G. Protective role of wogonin following traumatic brain injury by reducing oxidative stress and apoptosis via the PI3K/Nrf2/HO‑1 pathway. Int J Mol Med 2022; 49:53. [PMID: 35179214 PMCID: PMC8904077 DOI: 10.3892/ijmm.2022.5109] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/25/2022] [Indexed: 01/15/2023] Open
Abstract
Traumatic brain injury (TBI) is usually caused by accidental injuries and traffic accidents, with a very high mortality rate. Treatment and management following TBI are essential to reduce patient injury and help improve long‑term prognosis. Wogonin is a flavonoid compound with an antioxidant effect extracted from Scutellaria baicalensis Georgi. However, the function and mechanism of wogonin in protecting brain injury remain to be elucidated. The present study established a TBI model of Sprague‑Dawley rats and treated them with wogonin following trauma. The results showed that wogonin treatment significantly reduced neurobehavioral disorders, brain edema and hippocampal neuron damage caused by TBI. It was found that in TBI rats, administration of wogonin increased the levels of antioxidant factors glutathione, superoxide dismutase and catalase in the CA1 region of the hippocampus and significantly inhibited the production of malondialdehyde and reactive oxygen species. western blotting data showed that wogonin exerted antioxidant activity by downregulating the level of NOX2 protein. In inhibiting cell apoptosis, wogonin upregulated the expression of Bcl‑2 protein in the hippocampal CA1 region of TBI rats and inhibited caspase‑3 and Bax proteins. Additionally, wogonin inhibited the progression of injury following TBI through the PI3K/Akt/nuclear factor‑erythroid factor 2‑related factor 2 (Nrf2)/heme oxygenase‑1 (HO‑1) signaling pathway. Wogonin increased the expression of phosphorylated Akt, Nrf2 and HO‑1 in the hippocampus of TBI rats. Following the administration of PI3K inhibitor LY294002, the upregulation of these proteins by wogonin was partly reversed. In addition, LY294002 partially reversed the regulation of wogonin on NOX2, caspase‑3, Bax and Bcl‑2 proteins. Therefore, wogonin exerts antioxidant and anti‑apoptotic properties to prevent hippocampal damage following TBI, which is accomplished through the PI3K/Akt/Nrf2/HO‑1 pathway.
Collapse
Affiliation(s)
- Yan Feng
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Yaru Ju
- Department of Obstetrics, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei 050011, P.R. China
| | - Zhongjie Yan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Mingjun Ji
- Department of Critical Care Medical, Linxi County People's Hospital, Xingtai, Hebei 054000, P.R. China
| | - Ming Yang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Qiang Wu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Liqun Wang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Guozhu Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
9
|
Li C, Yu TY, Gong LR, Mu R, Zhang Y, Yu JB. Involvement of Nrf-2/HO-1 pathway in sevoflurane-induced cognitive improvement in rats with traumatic brain injury. Behav Brain Res 2021; 405:113200. [PMID: 33636237 DOI: 10.1016/j.bbr.2021.113200] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/14/2021] [Accepted: 02/18/2021] [Indexed: 02/09/2023]
Abstract
Traumatic brain injury (TBI) is an increasingly common emergency disease that usually leads to prolonged physical and cognitive impairments. In this study, we investigated if sevoflurane could induce cognitive improvement in TBI rats. Rats were subjected to head trauma induced by a fluid percussion device. A two-hour exposure to 3% sevoflurane was performed in a chamber immediately after TBI. Sevoflurane inhalation reduced the neurological and cognitive deficits induced by TBI with ameliorated synaptic injuries in the hippocampus. Moreover, after sevoflurane treatment, the expression of nuclear factor erythroid-2-related factor-2 (Nrf-2) and hemeoxygenase-1 (HO-1) in the hippocampus was enhanced 1 d after TBI and maintained at high levels 14 days later, and oxidative stress induced by TBI was inhibited. However, the HO-1 inhibitor, Zinc protoporphyrin (ZnPP), used to demonstrate the involvement of HO-1, suppressed the protective effect of sevoflurane. These results indicate that sevoflurane administered immediately after TBI may protect against TBI-induced synaptic and cognitive impairments by promoting the antioxidant Nrf-2/HO-1 pathway. Sevoflurane may be a promising anesthetic for patients with TBI.
Collapse
Affiliation(s)
- Cui Li
- Department of Anesthesiology and Critical Care Medicine, Tianjin NanKai Hospital, Tianjin Medical University, Tianjin, 300100, China
| | - Tian-Yu Yu
- Tianjin Medical University, Tianjin, 300070, China
| | - Li-Rong Gong
- Department of Anesthesiology and Critical Care Medicine, Tianjin NanKai Hospital, Tianjin Medical University, Tianjin, 300100, China
| | - Rui Mu
- Department of Anesthesiology and Critical Care Medicine, Tianjin NanKai Hospital, Tianjin Medical University, Tianjin, 300100, China
| | - Yuan Zhang
- Department of Anesthesiology and Critical Care Medicine, Tianjin NanKai Hospital, Tianjin Medical University, Tianjin, 300100, China
| | - Jian-Bo Yu
- Department of Anesthesiology and Critical Care Medicine, Tianjin NanKai Hospital, Tianjin Medical University, Tianjin, 300100, China.
| |
Collapse
|
10
|
Zhang J, Wang RJ, Chen M, Liu XY, Ma K, Xu HY, Deng WS, Ye YC, Li WX, Chen XY, Sun HT. Collagen/heparan sulfate porous scaffolds loaded with neural stem cells improve neurological function in a rat model of traumatic brain injury. Neural Regen Res 2021; 16:1068-1077. [PMID: 33269752 PMCID: PMC8224125 DOI: 10.4103/1673-5374.300458] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
One reason for the poor therapeutic effects of stem cell transplantation in traumatic brain injury is that exogenous neural stem cells cannot effectively migrate to the local injury site, resulting in poor adhesion and proliferation of neural stem cells at the injured area. To enhance the targeted delivery of exogenous stem cells to the injury site, cell therapy combined with neural tissue engineering technology is expected to become a new strategy for treating traumatic brain injury. Collagen/heparan sulfate porous scaffolds, prepared using a freeze-drying method, have stable physical and chemical properties. These scaffolds also have good cell biocompatibility because of their high porosity, which is suitable for the proliferation and migration of neural stem cells. In the present study, collagen/heparan sulfate porous scaffolds loaded with neural stem cells were used to treat a rat model of traumatic brain injury, which was established using the controlled cortical impact method. At 2 months after the implantation of collagen/heparan sulfate porous scaffolds loaded with neural stem cells, there was significantly improved regeneration of neurons, nerve fibers, synapses, and myelin sheaths in the injured brain tissue. Furthermore, brain edema and cell apoptosis were significantly reduced, and rat motor and cognitive functions were markedly recovered. These findings suggest that the novel collagen/heparan sulfate porous scaffold loaded with neural stem cells can improve neurological function in a rat model of traumatic brain injury. This study was approved by the Institutional Ethics Committee of Characteristic Medical Center of Chinese People’s Armed Police Force, China (approval No. 2017-0007.2) on February 10, 2019.
Collapse
Affiliation(s)
- Jian Zhang
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Characteristic Medical Center of Chinese People's Armed Police Force; Graduate School, Logistics University of People's Armed Police Force, Tianjin, China
| | - Ren-Jie Wang
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, China
| | - Miao Chen
- Graduate School, Affiliated Hospital of Traditional Chinese Medicine, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Xiao-Yin Liu
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Ke Ma
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Characteristic Medical Center of Chinese People's Armed Police Force; Graduate School, Logistics University of People's Armed Police Force, Tianjin, China
| | - Hui-You Xu
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Characteristic Medical Center of Chinese People's Armed Police Force; Graduate School, Logistics University of People's Armed Police Force, Tianjin, China
| | - Wu-Sheng Deng
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, China
| | - Yi-Chao Ye
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Characteristic Medical Center of Chinese People's Armed Police Force; Graduate School, Logistics University of People's Armed Police Force, Tianjin, China
| | - Wei-Xin Li
- Graduate School, Logistics University of People's Armed Police Force, Tianjin, China
| | - Xu-Yi Chen
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, China
| | - Hong-Tao Sun
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Traumatic Brain Injury and Neuroscience, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, China
| |
Collapse
|
11
|
Liu X, Li M, Zhu J, Huang W, Song J. Sestrin2 protects against traumatic brain injury by reinforcing the activation of Nrf2 signaling. Hum Exp Toxicol 2020; 40:1095-1111. [PMID: 33375867 DOI: 10.1177/0960327120984224] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Sestrin2 (SESN2) is stress-inducible protein that confers cytoprotective effects against various noxious stimuli. Accumulating evidence has documented that SESN2 has potent anti-apoptosis and anti-oxidative stress functions. However, whether it provides neuroprotection in traumatic brain injury (TBI) models remains unexplored. The purpose of this study was to explore the regulatory effect of SESN2 on TBI using in vivo and in vitro models. We found that TBI resulted in a marked induction of SESN2 in the cerebral cortex tissues of mice. SESN2 overexpression in the brain by in vivo gene transfer significantly decreased neurological deficit, brain edema, and neuronal apoptosis of mice with TBI. Moreover, the overexpression of SESN2 significantly decreased the oxidative stress induced by TBI in mice. In vitro studies of TBI demonstrated that SESN2 overexpression decreased apoptosis and oxidative stress in scratch-injured cortical neurons. Notably, SESN2 overexpression increased the nuclear levels of nuclear factor-erythroid 2-related factor 2 (Nrf2) and enhanced the activation of Nrf2 antioxidant signaling in in vivo and in vitro models of TBI. In addition, the inhibition of Nrf2 significantly abolished SESN2-mediated neuroprotective effects in vivo and in vitro. In conclusion, these results of our work demonstrate that SESN2 protects against TBI by enhancing the activation of Nrf2 antioxidant signaling.
Collapse
Affiliation(s)
- Xiaobin Liu
- Department of Neurosurgery, 162798The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Department of Neurosurgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China.,These authors (Xiaobin Liu and Min Li) contributed equally to this work and shared the first authorship
| | - Min Li
- Department of Neurosurgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China.,These authors (Xiaobin Liu and Min Li) contributed equally to this work and shared the first authorship
| | - Jiabao Zhu
- Department of Neurosurgery, Yuncheng Central Hospital of Shanxi Medical University, Yucheng, Shanxi, China
| | - Weidong Huang
- Department of Neurosurgery, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Jinning Song
- Department of Neurosurgery, 162798The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
12
|
Chen Y, Liu Y, Xie J, Zheng Q, Yue P, Chen L, Hu P, Yang M. Nose-to-Brain Delivery by Nanosuspensions-Based in situ Gel for Breviscapine. Int J Nanomedicine 2020; 15:10435-10451. [PMID: 33380794 PMCID: PMC7767747 DOI: 10.2147/ijn.s265659] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/25/2020] [Indexed: 12/19/2022] Open
Abstract
Purpose Nose-to-brain drug delivery is an effective approach for poorly soluble drugs to bypass the blood–brain barrier. A new drug intranasal delivery system, a nanosuspension-based in situ gel, was developed and evaluated to improve the solubility and bioavailability of the drug and to prolong its retention time in the nasal cavity. Materials and Methods Breviscapine (BRE) was chosen as the model drug. BRE nanosuspensions (BRE-NS) were converted into BRE nanosuspension powders (BRE-NP). A BRE nanosuspension in situ gelling system (BRE-NG) was prepared by mixing BRE-NP and 0.5% gellan gum (m/v). First, the BRE-NP were evaluated in terms of particle size and by differential scanning calorimetry (DSC), powder X-ray diffraction (PXRD), scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Subsequently, the critical ionic concentration of the gellan gum phase transition, influence of the deacetylated gellan gum (DGG) concentration on the expansion coefficient (S%), water-holding capacity, rheological properties and in vitro release behaviour of the BRE-NG were investigated. The pharmacokinetics and brain distribution of the BRE-NG after intranasal administration were compared with those of the intravenously injected BRE-NP nanosuspensions in rats. Results The rheology results demonstrated that BRE-NG was a non-Newtonian fluid with good spreadability and bioadhesion performance. Moreover, the absolute bioavailability estimated for BRE-NG after intranasal administration was 57.12%. The drug targeting efficiency (DTE%) of BRE in the cerebrum, cerebellum and olfactory bulb was 4006, 999 and 3290, respectively. The nose-to-brain direct transport percentage (DTP%) of the cerebrum, cerebellum and olfactory bulb was 0.975, 0.950 and 0.970, respectively. Conclusion It was concluded that the in situ gel significantly increased the drug retention time at the administration site. Therefore, the nanosuspension-based in situ gel could be a convenient and effective intranasal formulation for the administration of BRE.
Collapse
Affiliation(s)
- Yingchong Chen
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China
| | - Yuling Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, People's Republic of China
| | - Jin Xie
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China
| | - Qin Zheng
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China
| | - Pengfei Yue
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China
| | - Liru Chen
- Beijing Hospital, Beijing 100730, People's Republic of China
| | - Pengyi Hu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China
| | - Ming Yang
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, People's Republic of China
| |
Collapse
|
13
|
Salman M, Tabassum H, Parvez S. Nrf2/HO-1 mediates the neuroprotective effects of pramipexole by attenuating oxidative damage and mitochondrial perturbation after traumatic brain injury in rats. Dis Model Mech 2020; 13:dmm045021. [PMID: 32540990 PMCID: PMC7449795 DOI: 10.1242/dmm.045021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 05/26/2020] [Indexed: 12/27/2022] Open
Abstract
Pramipexole (PPX), a D2-like receptor agonist, is generally used in the treatment of Parkinson's disease and restless leg syndrome. Its neuroprotective effects have been shown against various neurological disorders. Recent research work has demonstrated that PPX exerts neuroprotection through mitochondria. However, the neuromodulator-related effects of PPX against traumatic brain injury (TBI) remain unexplored. The present study, therefore, investigated the mechanism of neuroprotection by PPX against oxidative stress, mitochondrial dysfunction and neuronal damage following TBI in rats. We hypothesized that the neuroprotection by PPX in TBI-subjected rats might involve activation of the Nrf2/HO-1 (also known as Nfe2l2/Hmox1) signaling pathway. PPX was injected intraperitoneally (0.25 mg/kg body weight and 1.0 mg/kg body weight) at different time intervals post-TBI. Several neurobehavioral parameters were assessed at 48 h post-TBI, and the brain was isolated for molecular and biochemical analysis. The results demonstrated that PPX treatment significantly improved the behavioral deficits, decreased the lipid peroxidation rate, increased glutathione levels and decreased 4-hydroxynonenal levels in TBI-subjected rats. PPX also increased the activities of glutathione peroxidase and superoxide dismutase enzymes. In addition, PPX treatment inhibited mitochondrial reactive oxygen species production, restored mitochondrial membrane potential and increased ATP levels after a TBI. Further, PPX treatment reduced the Bax/Bcl2 ratio and translocation of Bax to mitochondria and cytochrome-c to the cytosol. Finally, PPX treatment greatly accelerated the translocation of Nrf2 to the nucleus and upregulated HO-1 protein expression. We conclude that the neuroprotective effects of PPX are mediated by activation of the Nrf2/HO-1 signaling pathway following TBI.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Mohd Salman
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Heena Tabassum
- Division of Basic Medical Sciences, Indian Council of Medical Research, Ministry of Health and Family Welfare, Government of India, V. Ramalingaswamy Bhawan, New Delhi 110 029, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
14
|
Salman M, Tabassum H, Parvez S. Tannic Acid Provides Neuroprotective Effects Against Traumatic Brain Injury Through the PGC-1α/Nrf2/HO-1 Pathway. Mol Neurobiol 2020; 57:2870-2885. [PMID: 32399817 DOI: 10.1007/s12035-020-01924-3] [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: 02/03/2020] [Accepted: 04/22/2020] [Indexed: 12/30/2022]
Abstract
The present research was conducted to elucidate a possible molecular mechanism related to neuromodulatory effects of tannic acid (TA) supplementation against traumatic brain injury (TBI) in a rodent model. Oxidative damage and neuroinflammation play a critical role in TBI and lead to behavioral alterations and neuronal dysfunction and death. These changes suggest a potential avenue in neurotherapeutic intervention. The aim of the present study was to investigate the neuroprotective effects of TA and potential mechanism of these effects in a controlled cortical impact injury model of TBI in Wistar rats that were treated with TA (50 mg/kg body weight. i.p.) before 30 min and 6 and 18 h after TBI. TBI-induced rats were examined after 24 h for behavioral dysfunction, Nissl stain, lipid peroxidation rate, glutathione level, activities of antioxidant enzymes (catalase, glutathione S-transferase, glutathione peroxidase, and superoxide dismutase), the expression level of 4-hydroxynonenal, pro-inflammatory cytokines such as tumor necrosis factor alpha and interleukin-1 beta, as well as brain edema and immunoreactivity of glial fibrillary acidic protein. Results indicated that TA supplementation significantly modulated above mentioned alterations. Moreover, TA treatment effectively upregulated the protein expression of peroxisome proliferator-activated receptor gamma co-activator 1 alpha (PGC-1α) and nuclear factor-E2-related factor-2 (Nrf2) as well as mitochondrial transcription factor A and heme oxygenase-1 (HO-1) following TBI. Overall, our results suggest that TA effectively ameliorates the behavioral alterations, oxidative damage, mitochondrial impairment, and inflammation against TBI that may be attributed to activation of PGC-1α/Nrf-2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Mohd Salman
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Heena Tabassum
- Division of Biomedical Sciences, Indian Council of Medical Research, Ministry of Health and Family Welfare, Govt. of India, V. Ramalingaswamy Bhawan, P.O. Box No. 4911, New Delhi, 110029, India.
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|