1
|
Hu H, Cao B, Huang D, Lin Y, Zhou B, Ying J, Huang L, Zhang L. Withaferin a modulation of microglia autophagy mitigates neuroinflammation and enhances cognitive function in POCD. Sci Rep 2024; 14:26112. [PMID: 39478022 PMCID: PMC11525708 DOI: 10.1038/s41598-024-75284-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/03/2024] [Indexed: 11/02/2024] Open
Abstract
With the aging process of the global population and the development of medical technology, the cases of postoperative cognitive dysfunction (POCD) are also increasing. Due to the complexity of the pathogenesis, urgent treatment has been sought. Neuroinflammation induced by the accumulation of lipid droplets (LDs) in microglia has been closely watched in recent years and is also considered to be an important cause of nerve damage. Our study found that derived from Withania somnifera, Withaferin A (WA) could reduce the accumulation of LDs in the hippocampus of POCD mice, inhibit the expression of inflammatory factor interleukin-1β (IL-1β), and improve the cognitive ability of mice. Further in vitro experimental studies showed that WA increased the autophagy level of microglia, promoted the degradation of LDs, and reduced the production of inflammatory factors. In this regard, our comprehensive research endeavor holds the potential to furnish novel insights into therapeutic strategies aimed at addressing POCD and its associated neural impairments.
Collapse
Affiliation(s)
- Haijun Hu
- Department of Anesthesiology, The Second Affliated Hospital, Jiangxi Medical College, Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi Privince, People's Republic of China
| | - Bingbing Cao
- The First Hospital of Xiushui, Jiujiang, Jiangxi Province, People's Republic of China
| | - Dan Huang
- Department of Anesthesiology, The Second Affliated Hospital, Jiangxi Medical College, Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi Privince, People's Republic of China
| | - Yue Lin
- Department of Anesthesiology, The Second Affliated Hospital, Jiangxi Medical College, Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi Privince, People's Republic of China
| | - Bin Zhou
- Department of Anesthesiology, The Second Affliated Hospital, Jiangxi Medical College, Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi Privince, People's Republic of China
| | - Jun Ying
- Department of Anesthesiology, The Second Affliated Hospital, Jiangxi Medical College, Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi Privince, People's Republic of China
| | - Lelin Huang
- Department of Anesthesiology, Lushan Rehabilitation and Recuperation Center, PLA Joint Service Forces, Jiujiang, 3320000, People's Republic of China.
| | - Lieliang Zhang
- Department of Anesthesiology, The Second Affliated Hospital, Jiangxi Medical College, Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi Privince, People's Republic of China.
| |
Collapse
|
2
|
Zhou Z, Zhang P, Ya D, Liu J, Xu Y, Zhang Y, Tang W, Zhou D, Liao R, Liu L. Withaferin A protects against epilepsy by promoting LCN2-mediated astrocyte polarization to stopping neuronal ferroptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155892. [PMID: 39032282 DOI: 10.1016/j.phymed.2024.155892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 05/15/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Epilepsy is among the most frequent severe brain diseases, with few treatment options available. Neuronal ferroptosis is an important pathogenic mechanism in epilepsy. As a result, addressing ferroptosis appears to be a promising treatment approach for epilepsy. Withaferin A (WFA) is a C28 steroidal lactone that has a broad range of neuroprotective properties. Nonetheless, the antiepileptic action of WFA and the intrinsic mechanism by which it inhibits ferroptosis following epilepsy remain unknown. PURPOSE This study aimed at investigating to the antiepileptic potential of WFA in epilepsy, as well as to propose a potential therapeutic approach for epilepsy therapy. METHODS We conducted extensive research to examine the impacts of WFA on epilepsy and ferroptosis, using the kainic acid (KA)-treated primary astrocyte as an in vitro model and KA-induced temporal lobe epilepsy mice as an in vivo model. To analyze the neuroprotective effects of WFA on epileptic mice, electroencephalogram (EEG) recording, Nissl staining, and neurological function assessments such as the Morris water maze (MWM) test, Y-maze test, Elevated-plus maze (O-maze) test, and Open field test were used. Furthermore, the mechanism behind the neuroprotective effect of WFA in epilepsy was investigated using the transcriptomics analysis and verified on epileptic patient and epileptic mouse samples using Western blotting (WB) and immunofluorescence (IF) staining. In addition, WB, IF staining and specific antagonists/agonists were used to investigate astrocyte polarization and the regulatory signaling pathways involved. More critically, ferroptosis was assessed utilizing lipocalin-2 (LCN2) overexpression cell lines, siRNA knockdown, JC-1 staining, WB, IF staining, flow cytometry, electron microscopy (TEM), and ferroptosis-related GSH and MDA indicators. RESULTS In this study, we observed that WFA treatment reduced the number of recurrent seizures and time in seizure, and the loss of neurons in the hippocampal area in in epileptic mice, and even improved cognitive and anxiety impairment after epilepsy in a dose depend. Furthermore, WFA treatment was proven to enhance to the transformation of post-epileptic astrocytes from neurotoxic-type A1 to A2 astrocytes in both in vivo and in vitro experiments by inhibiting the phosphoinositide 3-kinase /AKT signaling pathway. At last, transcriptomics analysis in combination with functional experimental validation, it was discovered that WFA promoted astrocyte polarity transformation and then LCN2 in astrocytes, which inhibited neuronal ferroptosis to exert neuroprotective effects after epilepsy. In addition, we discovered significant astrocytic LCN2 expression in human TLE patient hippocampal samples. CONCLUSIONS Taken together, for the first, our findings suggest that WFA has neuroprotective benefits in epilepsy by modulating astrocyte polarization, and that LCN2 may be a novel potential target for the prevention and treatment of ferroptosis after epilepsy.
Collapse
Affiliation(s)
- Zixian Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Pengcheng Zhang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Dongshan Ya
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, PR China
| | - Jiao Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Yinchun Xu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Yu Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Wenfu Tang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Dong Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Rujia Liao
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, PR China.
| | - Ling Liu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
3
|
Ya D, Xiang W, Jiang Y, Zhang Y, Zhou Z, Li X, Deng J, Chen M, Yang B, Lin X, Liao R. Leptin combined with withaferin A boost posthemorrhagic neurogenesis via activation of STAT3/SOCS3 pathway. Exp Neurol 2024; 377:114809. [PMID: 38714285 DOI: 10.1016/j.expneurol.2024.114809] [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: 02/04/2024] [Revised: 04/10/2024] [Accepted: 05/03/2024] [Indexed: 05/09/2024]
Abstract
Neurogenesis as a potential strategy to improve the consequences of intracerebral hemorrhage (ICH). The current study investigates the effects of withaferin A (WFA) in combination with leptin (LEP) on ICH and neurogenesis mechanisms. LEP levels were dramatically reduced on days 7 and 14 following ICH insults in mice, but continuous WFA therapy significantly improved the potency of intrinsic LEP on day 14 after ICH. Furthermore, WFA combined with LEP enhances intrinsic neurogenesis and lessen motor deficits and long-term cognitive outcomes after ICH. In parallel, leptin deficiency in ob/ob mice limits enhancement of neurogenesis following ICH in response to WFA combined with LEP treatment. Importantly, the functional recovery conferred by WFA combined with LEP after ICH was inhibited by neurogenesis suppression. Mechanistically, this study unveiled that the signal transducer and activator of transcription-3 (STAT3) / suppressor of cytokine signaling-3 (SOCS3) pathway is a critical signaling pathway through which WFA combined with LEP treatment promotes intrinsic neurogenesis after ICH. Collectively, the results of this study elucidate the neuroprotective effects of WFA and LEP in ICH, and highlight a potential approach for ICH cell therapy.
Collapse
Affiliation(s)
- Dongshan Ya
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China; Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Wenjing Xiang
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China; Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Yanlin Jiang
- Department of Pharmacology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Yingmei Zhang
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China; Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Zixian Zhou
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China; Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Xiaoxia Li
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China; Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Jungang Deng
- Department of Pharmacology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Meiling Chen
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Bin Yang
- Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Xiaohui Lin
- Department of Geriatrics, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Rujia Liao
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China; Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China.
| |
Collapse
|
4
|
Mishra PS, Phaneuf D, Boutej H, Picher-Martel V, Dupre N, Kriz J, Julien JP. Inhibition of NF-κB with an Analog of Withaferin-A Restores TDP-43 Homeostasis and Proteome Profiles in a Model of Sporadic ALS. Biomedicines 2024; 12:1017. [PMID: 38790979 PMCID: PMC11118033 DOI: 10.3390/biomedicines12051017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/24/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
The current knowledge on pathogenic mechanisms in amyotrophic lateral sclerosis (ALS) has widely been derived from studies with cell and animal models bearing ALS-linked genetic mutations. However, it remains unclear to what extent these disease models are of relevance to sporadic ALS. Few years ago, we reported that the cerebrospinal fluid (CSF) from sporadic ALS patients contains toxic factors for disease transmission in mice via chronic intracerebroventricular (i.c.v.) infusion. Thus a 14-day i.c.v. infusion of pooled CSF samples from ALS cases in mice provoked motor impairment as well as ALS-like pathological features. This offers a unique paradigm to test therapeutics in the context of sporadic ALS disease. Here, we tested a new Withaferin-A analog (IMS-088) inhibitor of NF-κB that was found recently to mitigate disease phenotypes in mouse models of familial disease expressing TDP-43 mutant. Our results show that oral intake of IMS-088 ameliorated motor performance of mice infused with ALS-CSF and it alleviated pathological changes including TDP-43 proteinopathy, neurofilament disorganization, and neuroinflammation. Moreover, CSF infusion experiments were carried out with transgenic mice having neuronal expression of tagged ribosomal protein (hNfL-RFP mice), which allowed immunoprecipitation of neuronal ribosomes for analysis by mass spectrometry of the translational peptide signatures. The results indicate that treatment with IMS-088 prevented many proteomic alterations associated with exposure to ALS-CSF involving pathways related to cytoskeletal changes, inflammation, metabolic dysfunction, mitochondria, UPS, and autophagy dysfunction. The effective disease-modifying effects of this drug in a mouse model based on i.c.v. infusion of ALS-CSF suggest that the NF-κB signaling pathway represents a compelling therapeutic target for sporadic ALS.
Collapse
Affiliation(s)
- Pooja Shree Mishra
- CERVO Brain Research Centre, 2601 Chemin de la Canardière, Quebec, QC G1J 2G3, Canada; (P.S.M.); (D.P.); (H.B.); (J.K.)
| | - Daniel Phaneuf
- CERVO Brain Research Centre, 2601 Chemin de la Canardière, Quebec, QC G1J 2G3, Canada; (P.S.M.); (D.P.); (H.B.); (J.K.)
| | - Hejer Boutej
- CERVO Brain Research Centre, 2601 Chemin de la Canardière, Quebec, QC G1J 2G3, Canada; (P.S.M.); (D.P.); (H.B.); (J.K.)
| | - Vincent Picher-Martel
- Division of Neurosciences, Centre Hospitalier Universitaire de Québec, Laval University, Quebec, QC G1V 4G2, Canada; (V.P.-M.); (N.D.)
| | - Nicolas Dupre
- Division of Neurosciences, Centre Hospitalier Universitaire de Québec, Laval University, Quebec, QC G1V 4G2, Canada; (V.P.-M.); (N.D.)
| | - Jasna Kriz
- CERVO Brain Research Centre, 2601 Chemin de la Canardière, Quebec, QC G1J 2G3, Canada; (P.S.M.); (D.P.); (H.B.); (J.K.)
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
| | - Jean-Pierre Julien
- CERVO Brain Research Centre, 2601 Chemin de la Canardière, Quebec, QC G1J 2G3, Canada; (P.S.M.); (D.P.); (H.B.); (J.K.)
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
| |
Collapse
|
5
|
Zhang Y, Ya D, Yang J, Jiang Y, Li X, Wang J, Tian N, Deng J, Yang B, Li Q, Liao R. EAAT3 impedes oligodendrocyte remyelination in chronic cerebral hypoperfusion-induced white matter injury. CNS Neurosci Ther 2024; 30:e14487. [PMID: 37803915 PMCID: PMC10805396 DOI: 10.1111/cns.14487] [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: 06/26/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Chronic cerebral hypoperfusion-induced demyelination causes progressive white matter injury, although the pathogenic pathways are unknown. METHODS The Single Cell Portal and PanglaoDB databases were used to analyze single-cell RNA sequencing experiments to determine the pattern of EAAT3 expression in CNS cells. Immunofluorescence (IF) was used to detect EAAT3 expression in oligodendrocytes and oligodendrocyte progenitor cells (OPCs). EAAT3 levels in mouse brains were measured using a western blot at various phases of development, as well as in traumatic brain injury (TBI) and intracerebral hemorrhage (ICH) mouse models. The mouse bilateral carotid artery stenosis (BCAS) model was used to create white matter injury. IF, Luxol Fast Blue staining, and electron microscopy were used to investigate the effect of remyelination. 5-Ethynyl-2-Deoxy Uridine staining, transwell chamber assays, and IF were used to examine the effects of OPCs' proliferation, migration, and differentiation in vivo and in vitro. The novel object recognition test, the Y-maze test, the rotarod test, and the grid walking test were used to examine the impact of behavioral modifications. RESULTS A considerable amount of EAAT3 was expressed in OPCs and mature oligodendrocytes, according to single-cell RNA sequencing data. During multiple critical phases of mouse brain development, there were no substantial changes in EAAT3 levels in the hippocampus, cerebral cortex, or white matter. Furthermore, neither the TBI nor ICH models significantly affected the levels of EAAT3 in the aforementioned brain areas. The chronic white matter injury caused by BCAS, on the other hand, resulted in a strikingly high level of EAAT3 expression in the oligodendroglia and white matter. Correspondingly, blocking EAAT3 assisted in the recovery of cognitive and motor impairment as well as the restoration of cerebral blood flow following BCAS. Furthermore, EAAT3 suppression was connected to improved OPCs' survival and proliferation in vivo as well as faster OPCs' proliferation, migration, and differentiation in vitro. Furthermore, this study revealed that the mTOR pathway is implicated in EAAT3-mediated remyelination. CONCLUSIONS Our findings provide the first evidence that abnormally high levels of oligodendroglial EAAT3 in chronic cerebral hypoperfusion impair OPCs' pro-remyelination actions, hence impeding white matter repair and functional recovery. EAAT3 inhibitors could be useful in the treatment of ischemia demyelination.
Collapse
Affiliation(s)
- Yingmei Zhang
- Laboratory of NeuroscienceAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
- Department of NeurologyAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
| | - Dongshan Ya
- Laboratory of NeuroscienceAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
- Department of NeurologyAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
| | - Jiaxin Yang
- Laboratory of NeuroscienceAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
- Department of NeurologyAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
| | - Yanlin Jiang
- Department of PharmacologyAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
| | - Xiaoxia Li
- Laboratory of NeuroscienceAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
- Department of NeurologyAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
| | - Jiawen Wang
- Laboratory of NeuroscienceAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
- Guangxi Clinical Research Center for Neurological DiseasesAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
| | - Ning Tian
- Laboratory of NeuroscienceAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
- Guangxi Clinical Research Center for Neurological DiseasesAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
| | - Jungang Deng
- Department of PharmacologyAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
| | - Bin Yang
- Guangxi Clinical Research Center for Neurological DiseasesAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
| | - Qinghua Li
- Laboratory of NeuroscienceAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
- Department of NeurologyAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
- Guangxi Clinical Research Center for Neurological DiseasesAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
| | - Rujia Liao
- Laboratory of NeuroscienceAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
- Department of NeurologyAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
- Guangxi Clinical Research Center for Neurological DiseasesAffiliated Hospital of Guilin Medical University, Guilin Medical UniversityGuilinChina
| |
Collapse
|
6
|
Biswas D, Chakraborty A, Mukherjee S, Ghosh B. Hairy root culture: a potent method for improved secondary metabolite production of Solanaceous plants. FRONTIERS IN PLANT SCIENCE 2023; 14:1197555. [PMID: 37731987 PMCID: PMC10507345 DOI: 10.3389/fpls.2023.1197555] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/13/2023] [Indexed: 09/22/2023]
Abstract
Secondary metabolites synthesized by the Solanaceous plants are of major therapeutic and pharmaceutical importance, many of which are commonly obtained from the roots of these plants. 'Hairy roots', mirroring the same phytochemical pattern of the corresponding root of the parent plant with higher growth rate and productivity, are therefore extensively studied as an effective alternative for the in vitro production of these metabolites. Hairy roots are the transformed roots, generated from the infection site of the wounded plants with Agrobacterium rhizogenes. With their fast growth, being free from pathogen and herbicide contamination, genetic stability, and autotrophic nature for plant hormones, hairy roots are considered as useful bioproduction systems for specialized metabolites. Lately, several elicitation methods have been employed to enhance the accumulation of these compounds in the hairy root cultures for both small and large-scale production. Nevertheless, in the latter case, the cultivation of hairy roots in bioreactors should still be optimized. Hairy roots can also be utilized for metabolic engineering of the regulatory genes in the metabolic pathways leading to enhanced production of metabolites. The present study summarizes the updated and modern biotechnological aspects for enhanced production of secondary metabolites in the hairy root cultures of the plants of Solanaceae and their respective importance.
Collapse
Affiliation(s)
- Diptesh Biswas
- Plant Biotechnology Laboratory, Post Graduate Department of Botany, Ramakrishna Mission Vivekananda Centenary College, Kolkata, India
| | - Avijit Chakraborty
- Plant Biotechnology Laboratory, Post Graduate Department of Botany, Ramakrishna Mission Vivekananda Centenary College, Kolkata, India
| | - Swapna Mukherjee
- Department of Microbiology, Dinabandhu Andrews College, Kolkata, India
| | - Biswajit Ghosh
- Plant Biotechnology Laboratory, Post Graduate Department of Botany, Ramakrishna Mission Vivekananda Centenary College, Kolkata, India
| |
Collapse
|
7
|
Khalil R, Green RJ, Sivakumar K, Varandani P, Bharadwaj S, Mohapatra SS, Mohapatra S. Withaferin A Increases the Effectiveness of Immune Checkpoint Blocker for the Treatment of Non-Small Cell Lung Cancer. Cancers (Basel) 2023; 15:3089. [PMID: 37370701 PMCID: PMC10295988 DOI: 10.3390/cancers15123089] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Treatment of late-stage lung cancers remains challenging with a five-year survival rate of 8%. Immune checkpoint blockers (ICBs) revolutionized the treatment of non-small cell lung cancer (NSCLC) by reactivating anti-tumor immunity. Despite achieving durable responses, ICBs are effective in only 20% of patients due to immune resistance. Therefore, synergistic combinatorial approaches that overcome immune resistance are currently under investigation. Herein, we studied the immunomodulatory role of Withaferin A (WFA)-a herbal compound-and its effectiveness in combination with an ICB for the treatment of NSCLC. Our in vitro results show that WFA induces immunogenic cell death (ICD) in NSCLC cell lines and increases expression of the programmed death ligand-1 (PD-L1). The administration of N-acetyl cysteine (NAC), a reactive oxygen species (ROS) scavenger, abrogated WFA-induced ICD and PD-L1 upregulation, suggesting the involvement of ROS in this process. Further, we found that a combination of WFA and α-PD-L1 significantly reduced tumor growth in an immunocompetent tumor model. Our results showed that WFA increases CD-8 T-cells and reduces immunosuppressive cells infiltrating the tumor microenvironment. Administration of NAC partially inhibited the anti-tumor response of the combination regimen. In conclusion, our results demonstrate that WFA sensitizes NSCLC to α-PD-L1 in part via activation of ROS.
Collapse
Affiliation(s)
- Roukiah Khalil
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Ryan J. Green
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Kavya Sivakumar
- Taneja School of Pharmacy, University of South Florida, Tampa, FL 33612, USA
| | - Payal Varandani
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Srinivas Bharadwaj
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Shyam S. Mohapatra
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Taneja School of Pharmacy, University of South Florida, Tampa, FL 33612, USA
- Department of Veterans Affairs, James A. Haley Veterans Hospital, Tampa, FL 33612, USA
| | - Subhra Mohapatra
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Department of Veterans Affairs, James A. Haley Veterans Hospital, Tampa, FL 33612, USA
| |
Collapse
|
8
|
Peng Y, Yin H, Li S, Yang H. Transcriptome of pituitary function changes in rat model of high altitude cerebral edema. Genomics 2022; 114:110519. [PMID: 36347325 DOI: 10.1016/j.ygeno.2022.110519] [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: 07/08/2022] [Revised: 10/22/2022] [Accepted: 11/04/2022] [Indexed: 11/06/2022]
Abstract
High altitude cerebral edema (HACE) is a serious subtype of acute mountain sickness (AMS). Studies have suggested that increased expression of corticotropin releasing hormone receptor 1 (CRFR1) in pituitary is related to the development of HACE, but no study has revealed the molecular landscape of pituitary function changes in this process. Rat model of HACE was established by simulating the high-altitude hypobaric hypoxia environment. Then RNA-sequencing was performed of rat pituitary gland (PG) in HACE and non-HACE groups. The function annotations, enrichment analysis, protein-protein interaction (PPI) network, chromosome location and drug repositioning of differentially expressed genes (DEGs) were explored based on the transcriptomic data. And we found pituitary secretion function was disordered in HACE, which was partly due to activated inflammation and oxidative stress. In addition, we identified potential biomarkers for early recognition of pituitary dysfunction and potential protective drugs for pituitary function in HACE.
Collapse
Affiliation(s)
- Yuyang Peng
- Multidisciplinary Center for Pituitary Adenomas of Chongqing, Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Huachun Yin
- Multidisciplinary Center for Pituitary Adenomas of Chongqing, Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Song Li
- Multidisciplinary Center for Pituitary Adenomas of Chongqing, Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China; Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China
| | - Hui Yang
- Multidisciplinary Center for Pituitary Adenomas of Chongqing, Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China; Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing, China.
| |
Collapse
|
9
|
Zhou ZX, Cui Q, Zhang YM, Yang JX, Xiang WJ, Tian N, Jiang YL, Chen ML, Yang B, Li QH, Liao RJ. Withaferin A inhibits ferroptosis and protects against intracerebral hemorrhage. Neural Regen Res 2022; 18:1308-1315. [PMID: 36453416 PMCID: PMC9838153 DOI: 10.4103/1673-5374.355822] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Recent studies have indicated that suppressing oxidative stress and ferroptosis can considerably improve the prognosis of intracerebral hemorrhage (ICH). Withaferin A (WFA), a natural compound, exhibits a positive effect on a number of neurological diseases. However, the effects of WFA on oxidative stress and ferroptosis-mediated signaling pathways to ICH remain unknown. In this study, we investigated the neuroprotective effects and underlying mechanism for WFA in the regulation of ICH-induced oxidative stress and ferroptosis. We established a mouse model of ICH by injection of autologous tail artery blood into the caudate nucleus and an in vitro cell model of hemin-induced ICH. WFA was injected intracerebroventricularly at 0.1, 1 or 5 µg/kg once daily for 7 days, starting immediately after ICH operation. WFA markedly reduced brain tissue injury and iron deposition and improved neurological function in a dose-dependent manner 7 days after cerebral hemorrhage. Through in vitro experiments, cell viability test showed that WFA protected SH-SY5Y neuronal cells against hemin-induced cell injury. Enzyme-linked immunosorbent assays in vitro and in vivo showed that WFA markedly decreased the level of malondialdehyde, an oxidative stress marker, and increased the activities of anti-oxidative stress markers superoxide dismutase and glutathione peroxidase after ICH. Western blot assay, quantitative polymerase chain reaction and immunofluorescence results demonstrated that WFA activated the nuclear factor E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling axis, promoted translocation of Nrf2 from the cytoplasm to nucleus, and increased HO-1 expression. Silencing Nrf2 with siRNA completely reversed HO-1 expression, oxidative stress and protective effects of WFA. Furthermore, WFA reduced hemin-induced ferroptosis. However, after treatment with an HO-1 inhibitor, the neuroprotective effects of WFA against hemin-induced ferroptosis were weakened. MTT test results showed that WFA combined with ferrostatin-1 reduced hemin-induced SH-SY5Y neuronal cell injury. Our findings reveal that WFA treatment alleviated ICH injury-induced ferroptosis and oxidative stress through activating the Nrf2/HO-1 pathway, which may highlight a potential role of WFA for the treatment of ICH.
Collapse
Affiliation(s)
- Zi-Xian Zhou
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China,Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Qi Cui
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China,Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Ying-Mei Zhang
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China,Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Jia-Xin Yang
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China,Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Wen-Jing Xiang
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China,Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Ning Tian
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China,Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Yan-Lin Jiang
- Department of Pharmacology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Mei-Ling Chen
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Bin Yang
- Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Qing-Hua Li
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China,Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China,Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Ru-Jia Liao
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China,Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China,Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China,Correspondence to: Ru-Jia Liao, .
| |
Collapse
|
10
|
Cui Q, Zhang Y, Tian N, Yang J, Ya D, Xiang W, Zhou Z, Jiang Y, Deng J, Yang B, Lin X, Li Q, Liao R. Leptin Promotes Angiogenesis via Pericyte STAT3 Pathway upon Intracerebral Hemorrhage. Cells 2022; 11:cells11172755. [PMID: 36078162 PMCID: PMC9454866 DOI: 10.3390/cells11172755] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 11/22/2022] Open
Abstract
Angiogenesis is a vital endogenous brain self-repair processes for neurological recovery after intracerebral hemorrhage (ICH). Increasing evidence suggests that leptin potentiates angiogenesis and plays a beneficial role in stroke. However, the proangiogenic effect of leptin on ICH has not been adequately explored. Moreover, leptin triggers post-ICH angiogenesis through pericyte, an important component of forming new blood vessels, which remains unclear. Here, we reported that exogenous leptin infusion dose-dependent promoted vascular endothelial cells survival and proliferation at chronic stage of ICH mice. Additionally, leptin robustly ameliorated pericytes loss, enhanced pericytes proliferation and migration in ICH mice in vivo, and in ICH human brain microvascular pericytes (HBVPC) in vitro. Notably, we showed that pericytes-derived pro-angiogenic factors were responsible for enhancing the survival, proliferation and tube formation followed leptin treatment in human brain microvascular endothelial cells (HCMEC/D3)/HBVPC co-culture models. Importantly, considerable improvements in neurobehavioral function and hostile microenvironment were observed in leptin treatment ICH mice, indicating that better vascular functionality post ICH improves outcome. Mechanistically, this study unveiled that leptin boost post-ICH angiogenesis potentially through modulation of leptin receptor (leptinR)/Signal Transducer and Activator of Transcription 3 (STAT3) signaling pathway in pericyte. Thus, leptin may be a lucrative option for the treatment of ICH.
Collapse
Affiliation(s)
- Qi Cui
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Yingmei Zhang
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Ning Tian
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
- Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Jiaxin Yang
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Dongshan Ya
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Wenjing Xiang
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Zixian Zhou
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Yanlin Jiang
- Department of Pharmacology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Jungang Deng
- Department of Pharmacology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Bin Yang
- Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Xiaohui Lin
- Department of Geriatrics, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Qinghua Li
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
- Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
| | - Rujia Liao
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
- Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin 541004, China
- Correspondence: ; Tel.: +86-0773-2833025
| |
Collapse
|
11
|
Diao X, Cui Q, Tian N, Zhou Z, Xiang W, Jiang Y, Deng J, Liao H, Lin X, Li Q, Liao R. Hemorrhage-Induced Sphingosine Kinase 1 Contributes to Ferroptosis-Mediated Secondary Brain Injury in Intracerebral Hemorrhage. Mol Neurobiol 2022; 59:1381-1397. [PMID: 34993846 DOI: 10.1007/s12035-021-02605-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 10/15/2021] [Indexed: 12/18/2022]
Abstract
The pathogenic processes of brain injury after intracerebral hemorrhage (ICH) have not yet been fully elucidated. Increasing evidence suggests that ferroptosis activation aggravates injury after ICH, but the underlying mechanism remains unclear. Sphingosine kinase 1 (Sphk1) is a key enzyme in the regulation of sphingosine metabolism involved in the ferroptosis pathway, but its role in ICH needs clarification. In this study, transcriptional changes in ICH patients were assessed by microarray data, exposing Sphk1 as a highly upregulated gene during ICH. Furthermore, Sphk1 chemical inhibitors and siRNA were used to inhibit ICH-induced Sphk1 upregulation in in vivo and in vitro models, showing that Sphk1 inhibition after protects against ferroptosis and attenuates secondary brain injury and cell death. Mechanistically, this study unveiled that sphingosine kinase 1/sphingosine 1-phosphate/extracellular-regulated protein kinases/phosphorylated extracellular-regulated protein kinases (Sphk1/S1p/ERK/p-ERK) pathway is responsible for regulation of ferroptosis leading to secondary brain injury and cell death following ICH. Collectively, this study demonstrates that ferroptosis is closely associated with ICH, and that Sphk1 has a critical role in this lethal process. These results suggest a novel unique and effective therapeutic approach for ICH prevention and treatment.
Collapse
Affiliation(s)
- Xiaojun Diao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410000, China
- Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China
| | - Qi Cui
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China
| | - Ning Tian
- Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China
| | - Zixian Zhou
- Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541004, China
| | - Wenjing Xiang
- Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541004, China
| | - Yanlin Jiang
- Department of Pharmacology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China
| | - Jungang Deng
- Department of Pharmacology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China
| | - Hongzhan Liao
- Department of Neurosurgery, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China
| | - Xiaohui Lin
- Department of Neurology, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China
| | - Qinghua Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410000, China.
- Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China.
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China.
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541004, China.
| | - Rujia Liao
- Guangxi Clinical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China.
- Laboratory of Neuroscience, Affiliated Hospital of Guilin Medical University, Guilin Medical University, Guilin, 541004, China.
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541004, China.
| |
Collapse
|
12
|
Ben Bakrim W, El Bouzidi L, Manouze H, Hafsa J, Sobeh M, Ba-M'hamed S, Bekkouche K, Kouisni L. Anti-amnesic effects of withaferin A, a steroidal lactone isolated from Withania adpressa, on scopolamine-induced memory impairment in mice. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2021.103529] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
13
|
Das R, Rauf A, Akhter S, Islam MN, Emran TB, Mitra S, Khan IN, Mubarak MS. Role of Withaferin A and Its Derivatives in the Management of Alzheimer's Disease: Recent Trends and Future Perspectives. Molecules 2021; 26:3696. [PMID: 34204308 PMCID: PMC8234716 DOI: 10.3390/molecules26123696] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/12/2021] [Accepted: 06/15/2021] [Indexed: 01/02/2023] Open
Abstract
Globally, Alzheimer's disease (AD) is one of the most prevalent age-related neurodegenerative disorders associated with cognitive decline and memory deficits due to beta-amyloid deposition (Aβ) and tau protein hyperphosphorylation. To date, approximately 47 million people worldwide have AD. This figure will rise to an estimated 75.6 million by 2030 and 135.5 million by 2050. According to the literature, the efficacy of conventional medications for AD is statistically substantial, but clinical relevance is restricted to disease slowing rather than reversal. Withaferin A (WA) is a steroidal lactone glycowithanolides, a secondary metabolite with comprehensive biological effects. Biosynthetically, it is derived from Withania somnifera (Ashwagandha) and Acnistus breviflorus (Gallinero) through the mevalonate and non-mevalonate pathways. Mounting evidence shows that WA possesses inhibitory activities against developing a pathological marker of Alzheimer's diseases. Several cellular and animal models' particulates to AD have been conducted to assess the underlying protective effect of WA. In AD, the neuroprotective potential of WA is mediated by reduction of beta-amyloid plaque aggregation, tau protein accumulation, regulation of heat shock proteins, and inhibition of oxidative and inflammatory constituents. Despite the various preclinical studies on WA's therapeutic potentiality, less is known regarding its definite efficacy in humans for AD. Accordingly, the present study focuses on the biosynthesis of WA, the epidemiology and pathophysiology of AD, and finally the therapeutic potential of WA for the treatment and prevention of AD, highlighting the research and augmentation of new therapeutic approaches. Further clinical trials are necessary for evaluating the safety profile and confirming WA's neuroprotective potency against AD.
Collapse
Affiliation(s)
- Rajib Das
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh; (R.D.); (S.M.)
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar 23561, Pakistan;
| | - Saima Akhter
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh;
| | - Mohammad Nazmul Islam
- Department of Pharmacy, International Islamic University Chittagong, Chittagong 4318, Bangladesh;
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh; (R.D.); (S.M.)
| | - Ishaq N. Khan
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar 25100, Pakistan;
| | | |
Collapse
|
14
|
Saggam A, Limgaokar K, Borse S, Chavan-Gautam P, Dixit S, Tillu G, Patwardhan B. Withania somnifera (L.) Dunal: Opportunity for Clinical Repurposing in COVID-19 Management. Front Pharmacol 2021; 12:623795. [PMID: 34012390 PMCID: PMC8126694 DOI: 10.3389/fphar.2021.623795] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/30/2021] [Indexed: 12/13/2022] Open
Abstract
As the COVID-19 pandemic is progressing, the therapeutic gaps in conventional management have highlighted the need for the integration of traditional knowledge systems with modern medicine. Ayurvedic medicines, especially Ashwagandha (Withania somnifera (L.) Dunal, WS), may be beneficial in the management of COVID-19. WS is a widely prescribed Ayurvedic botanical known as an immunomodulatory, antiviral, anti-inflammatory, and adaptogenic agent. The chemical profile and pharmacological activities of WS have been extensively reported. Several clinical studies have reported its safety for use in humans. This review presents a research synthesis of in silico, in vitro, in vivo, and clinical studies on Withania somnifera (L.) Dunal (WS) and discusses its potential for prophylaxis and management of COVID-19. We have collated the data from studies on WS that focused on viral infections (HIV, HSV, H1N1 influenza, etc.) and noncommunicable diseases (hypertension, diabetes, cancer, etc.). The experimental literature indicates that WS has the potential for 1) maintaining immune homeostasis, 2) regulating inflammation, 3) suppressing pro-inflammatory cytokines, 4) organ protection (nervous system, heart, lung, liver, and kidney), and 5) anti-stress, antihypertensive, and antidiabetic activities. Using these trends, the review presents a triangulation of Ayurveda wisdom, pharmacological properties, and COVID-19 pathophysiology ranging from viral entry to end-stage acute respiratory distress syndrome (ARDS). The review proposes WS as a potential therapeutic adjuvant for various stages of COVID-19 management. WS may also have beneficial effects on comorbidities associated with the COVID-19. However, systematic studies are needed to realize the potential of WS for improving clinical outcome of patients with COVID-19.
Collapse
Affiliation(s)
- Akash Saggam
- AYUSH Center of Excellence, Center for Complementary and Integrative Health, Interdisciplinary School of Health Sciences, Savitribai Phule Pune University, Pune, India
| | - Kirti Limgaokar
- Division of Biochemistry, Department of Chemistry, Fergusson College (Autonomous), Pune, India
| | - Swapnil Borse
- AYUSH Center of Excellence, Center for Complementary and Integrative Health, Interdisciplinary School of Health Sciences, Savitribai Phule Pune University, Pune, India
| | - Preeti Chavan-Gautam
- AYUSH Center of Excellence, Center for Complementary and Integrative Health, Interdisciplinary School of Health Sciences, Savitribai Phule Pune University, Pune, India
| | | | - Girish Tillu
- AYUSH Center of Excellence, Center for Complementary and Integrative Health, Interdisciplinary School of Health Sciences, Savitribai Phule Pune University, Pune, India
| | - Bhushan Patwardhan
- AYUSH Center of Excellence, Center for Complementary and Integrative Health, Interdisciplinary School of Health Sciences, Savitribai Phule Pune University, Pune, India
| |
Collapse
|
15
|
Zhao X, Wang J, Tang L, Li P, Ru J, Bai Y. Withaferin A protects against hyperuricemia induced kidney injury and its possible mechanisms. Bioengineered 2021; 12:589-600. [PMID: 33517833 PMCID: PMC8806220 DOI: 10.1080/21655979.2021.1882761] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The study was designed to explore the effects of Withaferin A (WFA) on hyperuricemia-induced kidney injury and its action mechanism. Potassium oxonate (PO) was employed to establish the hyperuricemic mouse model. The pathological changes of renal tissue were evaluated by hematoxylin-eosin and masson trichrome staining. The levels of creatinine, blood urea nitrogen (BUN), uric acid (UA) and xanthine oxidase (XOD) were detected using corresponding commercial kits. Expressions of collagen-related and apoptosis-associated proteins in renal tissues were, respectively, evaluated by immunofluorescence and western blotting. Cell apoptosis was detected by TUNEL assay, and transporter expressions using western blotting. Followed by WFA, NRK-52E cells were treated with UA before evaluation of apoptosis and fibrosis. Results indicated that WFA ameliorated renal damage, improved kidney function, and decreased levels of creatinine, BUN, UA, and XOD in PO-induced hyperuricemic mice. Furthermore, WFA significantly prevented renal fibrosis and increased the expression of collagen-related proteins. Similarly, WFA markedly inhibited renal apoptosis, accompanied by changes of apoptosis-related proteins. Importantly, expression of transporters responsible for the secretion of organic anion transporter 1 (OAT1), OAT3, ATP-binding cassette subfamily G member 2 (ABCG2) was remarkably enhanced whereas that of urate transporter 1 (URAT1) and glucose transporter 9 (GLUT9) was reduced in renal tissues of mice with hyperuricemia. In vitro study revealed that WFA notably ameliorated UA-induced cell fibrosis and apoptosis. Taken together, WFA improves kidney function by decreasing UA via regulation of XOD and transporter genes in renal tubular cells.
Collapse
Affiliation(s)
- Xia Zhao
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University , Beijing, China
| | - Jing Wang
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University , Beijing, China
| | - Liying Tang
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University , Beijing, China
| | - Pei Li
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University , Beijing, China
| | - Jing Ru
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University , Beijing, China
| | - Yuzhi Bai
- Department of Geriatrics, Beijing Chaoyang Hospital Affiliated to Capital Medical University , Beijing, China
| |
Collapse
|
16
|
Kumar S, Phaneuf D, Julien JP. Withaferin-A Treatment Alleviates TAR DNA-Binding Protein-43 Pathology and Improves Cognitive Function in a Mouse Model of FTLD. Neurotherapeutics 2021; 18:286-296. [PMID: 33078279 PMCID: PMC8116414 DOI: 10.1007/s13311-020-00952-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2020] [Indexed: 11/26/2022] Open
Abstract
Withaferin-A, an active withanolide derived from the medicinal herbal plant Withania somnifera induces autophagy, reduces TDP-43 proteinopathy, and improves cognitive function in transgenic mice expressing mutant TDP-43 modelling FTLD. TDP-43 is a nuclear DNA/RNA-binding protein with cellular functions in RNA transcription and splicing. Abnormal cytoplasmic aggregates of TDP-43 occur in several neurodegenerative diseases including amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration (FTLD), and limbic-predominant age-related TDP-43 encephalopathy (LATE). To date, no effective treatment is available for TDP-43 proteinopathies. Here, we tested the effects of withaferin-A (WFA), an active withanolide extracted from the medicinal herbal plant Withania somnifera, in a transgenic mouse model of FTLD expressing a genomic fragment encoding mutant TDP-43G348C. WFA treatment ameliorated the cognitive performance of the TDP-43G348C mice, and it reduced NF-κB activity and neuroinflammation in the brain. WFA alleviated TDP-43 pathology while it boosted the levels of the autophagic marker LC3BII in the brain. These data suggest that WFA and perhaps other autophagy inducers should be considered as potential therapy for neurodegenerative diseases with TDP-43 pathology.
Collapse
Affiliation(s)
- Sunny Kumar
- CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Daniel Phaneuf
- CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada
| | - Jean-Pierre Julien
- CERVO Brain Research Centre, Laval University, Quebec City, QC, Canada.
- Department of Psychiatry and Neuroscience, Canada Research Chair in Neurodegeneration, Université Laval, 2601, Chemin de la Canardière, Québec City, Québec, G1J 2G3, Canada.
| |
Collapse
|
17
|
Behl T, Sharma A, Sharma L, Sehgal A, Zengin G, Brata R, Fratila O, Bungau S. Exploring the Multifaceted Therapeutic Potential of Withaferin A and Its Derivatives. Biomedicines 2020; 8:E571. [PMID: 33291236 PMCID: PMC7762146 DOI: 10.3390/biomedicines8120571] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022] Open
Abstract
Withaferin A (WA), a manifold studied, C28-steroidal lactone withanolide found in Withania somnifera. Given its unique beneficial effects, it has gathered attention in the era of modern science. Cancer, being considered a "hopeless case and the leading cause of death worldwide, and the available conventional therapies have many lacunae in the form of side effects. The poly pharmaceutical natural compound, WA treatment, displayed attenuation of various cancer hallmarks by altering oxidative stress, promoting apoptosis, and autophagy, inhibiting cell proliferation, reducing angiogenesis, and metastasis progression. The cellular proteins associated with antitumor pathways were also discussed. WA structural modifications attack multiple signal transduction pathways and enhance the therapeutic outcomes in various diseases. Moreover, it has shown validated pharmacological effects against multiple neurodegenerative diseases by inhibiting acetylcholesterinases and butyrylcholinesterases enzyme activity, antidiabetic activity by upregulating adiponectin and preventing the phosphorylation of peroxisome proliferator-activated receptors (PPARγ), cardioprotective activity by AMP-activated protein kinase (AMPK) activation and suppressing mitochondrial apoptosis. The current review is an extensive survey of various WA associated disease targets, its pharmacokinetics, synergistic combination, modifications, and biological activities.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Aditi Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh 173229, India; (A.S.); (L.S.)
| | - Lalit Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh 173229, India; (A.S.); (L.S.)
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University Campus, Konya 42250, Turkey;
| | - Roxana Brata
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (R.B.); (O.F.)
| | - Ovidiu Fratila
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (R.B.); (O.F.)
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| |
Collapse
|