1
|
Ge H, Yan S, Yin M, Gao Y, Wang J, Wang Q, Xu G, Yang M. Gua Sha Alleviates Radiculitis-Induced Pain Via HIF-1α-Mediated Metabolic Reprogramming Pathway in Rats. Pain Res Manag 2025; 2025:9923147. [PMID: 40130025 PMCID: PMC11932754 DOI: 10.1155/prm/9923147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/27/2025] [Indexed: 03/26/2025]
Abstract
Background: Radiculitis-induced pain (RIP) results from dorsal root ganglion (DRG) sensitization due to inflammation. Hypoxia-inducible factor 1-alpha (HIF-1α) is linked to inflammatory responses through metabolic changes, but its role in RIP is not well understood. Gua Sha therapy has been shown to reduce inflammation and neural damage from lumbar disc herniation (LDH). This study investigates whether HIF-1α-mediated metabolic reprogramming contributes to the pain-relieving effects of Gua Sha in RIP. Methods: Male SD rats were subjected to LDH surgery and divided into six groups: sham, model, sham Gua Sha, Gua Sha, Gua Sha + DMOG, and Gua Sha + YC-1. Gua Sha was applied 5 days postsurgery, every other day for three sessions per course, totaling three courses. Changes in paw withdrawal threshold (PWT) and latency (PWL) were monitored, along with blood flow in the rats' backs. Levels of IL-1β, TNF-α, and NF-κB were assessed in serum and DRG tissue. Pathological changes and hypoxia in DRG tissues were observed using hematoxylin-eosin staining and immunofluorescence. Western blotting and qPCR measured HIF-1α, GLUT1, PFKM, and PDK1 expression, while lactic acid and ATP levels in DRG tissue were also evaluated. Results: Gua Sha increased PWT and PWL, reduced serum and DRG inflammatory factors, improved back microcirculation, alleviated DRG hypoxia, and decreased HIF-1α and related signaling factors. It also lowered lactic acid and raised ATP levels. DMOG, a HIF-1α activator, reversed these effects. HIF-1α activation did not affect serum inflammatory factors but partially improved PWT. Inhibition of HIF-1α with YC-1 did not significantly differ from Gua Sha alone. Conclusion: HIF-1α-mediated metabolic reprogramming is a pathogenic mechanism in RIP. Gua Sha alleviates RIP by enhancing microcirculation, improving DRG hypoxia, inhibiting HIF-1α-mediated reprogramming, and reducing DRG sensitization and inflammation. This study provides insights into the mechanisms of Gua Sha's therapeutic effects in RIP.
Collapse
Affiliation(s)
- Haotian Ge
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Shuxia Yan
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Mingwan Yin
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yujie Gao
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jiayi Wang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Qin Wang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- TCM Nursing Intervention Laboratory of Chronic Diseases, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Guihua Xu
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Min Yang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Nerush MO, Shevyrin VA, Golushko NI, Moskalenko AM, Rosemberg DB, De Abreu MS, Yang LE, Galstyan DS, Lim LW, Demin KA, Kalueff AV. Classics in Chemical Neuroscience: Deliriant Antihistaminic Drugs. ACS Chem Neurosci 2024; 15:3848-3862. [PMID: 39404616 DOI: 10.1021/acschemneuro.4c00505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024] Open
Abstract
Antihistaminic drugs are widely used clinically and have long been primarily known for their use to treat severe allergic conditions caused by histamine release. Antihistaminic drugs also exert central nervous system (CNS) effects, acting as anxiolytics, hypnotics, and neuroleptics. However, these drugs also have multiple serious neuropharmacological side-effects, inducing delirium, hyperarousal, disorganized behavior, and hallucinations. Due to their robust CNS effects, antihistamines are also increasingly abused, with occasional overdoses and life-threatening toxicity. Here, we discuss chemical and neuropharmacological aspects of antihistaminic drugs in both human and animal (experimental) models and outline their current societal and mental health importance as neuroactive substances.
Collapse
Affiliation(s)
- Maria O Nerush
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg 199034, Russia
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg 197341, Russia
| | | | - Nikita I Golushko
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg 199034, Russia
| | | | - Denis B Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria 97105-900, Brazil
| | - Murilo S De Abreu
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, Brazil
- Western Caspian University, Baku 1001, Azerbaijan
| | - Long-En Yang
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215000, P. R. China
- Suzhou Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215000, P. R. China
| | - David S Galstyan
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg 199034, Russia
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg 197341, Russia
| | - Lee Wei Lim
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215000, P. R. China
- Suzhou Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215000, P. R. China
| | - Konstantin A Demin
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg 199034, Russia
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg 197341, Russia
| | - Allan V Kalueff
- Institute of Translational Biomedicine (ITBM), St. Petersburg State University, St. Petersburg 199034, Russia
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg 197341, Russia
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215000, P. R. China
- Suzhou Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou 215000, P. R. China
| |
Collapse
|
3
|
Hu WJ, Wei H, Cai LL, Xu YH, Du R, Zhou Q, Zhu XL, Li YF. Magnetic targeting enhances the neuroprotective function of human mesenchymal stem cell-derived iron oxide exosomes by delivering miR-1228-5p. J Nanobiotechnology 2024; 22:665. [PMID: 39468528 PMCID: PMC11514807 DOI: 10.1186/s12951-024-02941-3] [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: 08/18/2024] [Accepted: 10/19/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Treating mitochondrial dysfunction is a promising approach for the treatment of post-stroke cognitive impairment (PSCI). HuMSC-derived exosomes (H-Ex) have shown powerful therapeutic effects in improving mitochondrial function, but the specific effects are unclear and its brain tissue targeting needs to be further optimized. RESULTS In this study, we found that H-Ex can improve mitochondrial dysfunction of neurons and significantly enhance the cognitive behavior performance of MCAO mice in OGD/R-induced SHSY5Y cells and MCAO mouse models. Based on this, we have developed an exosome delivery system loaded with superparamagnetic iron oxide nanoparticles (Spion-Ex) that can effectively penetrate the blood-brain barrier (BBB). The research results showed that under magnetic attraction, Spion-Ex can more effectively target the brain tissue and significantly improve mitochondrial function of neurons after stroke. Meanwhile, we further confirmed that miR-1228-5p is a key factor for H-Ex to improve mitochondrial function and cognitive behavior both in vivo and in vitro. The specific mechanism is that the increase of miR-1228-5p mediated by H-Ex can inhibit the expression of TRAF6 and activate the TRAF6-NADPH oxidase 1 (NOX1) pathway, thereby exerting protective effects against oxidative damage. More importantly, we found that under magnetic attraction, Spion-Ex exhibited excellent cognitive improvement effects by delivering miR-1228-5p. CONCLUSIONS Our research found that H-Ex has a good therapeutic effect on PSCI by increasing the expression of miR-1228-5p in PSCI, while H-Ex loaded with Spion-Ex exhibited more excellent effects on improving mitochondrial function and cognitive impairment under magnetic attraction, which can be used as a novel strategy for the treatment of PSCI.
Collapse
Affiliation(s)
- Wei-Jia Hu
- Department of Radiology, Affiliated People's Hospital of Jiangsu University, No. 8, Dianli Road, Zhenjiang, Jiangsu, 212001, China
- Department of Radiology, Affiliated Hospital of Jiangsu University, No. 438, Jiefang Road, Zhenjiang, Jiangsu, 212001, China
| | - Hong Wei
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang, Jiangsu, 212001, China
- Department of Neurology, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang, Jiangsu, 212001, China
| | - Li-Li Cai
- Medical College of Jiangsu University, No. 301, XueFu Road, Zhenjiang, Jiangsu, 212001, China
| | - Yu-Hao Xu
- Department of Radiology, Affiliated Hospital of Jiangsu University, No. 438, Jiefang Road, Zhenjiang, Jiangsu, 212001, China
| | - Rui Du
- Medical College of Jiangsu University, No. 301, XueFu Road, Zhenjiang, Jiangsu, 212001, China
| | - Qun Zhou
- Department of Radiology, Affiliated Hospital of Jiangsu University, No. 438, Jiefang Road, Zhenjiang, Jiangsu, 212001, China
| | - Xiao-Lan Zhu
- Department of Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, No. 20, Zhengdong Road, Zhenjiang, Jiangsu, 212001, China.
| | - Yue-Feng Li
- Department of Radiology, Affiliated People's Hospital of Jiangsu University, No. 8, Dianli Road, Zhenjiang, Jiangsu, 212001, China.
- Medical College of Jiangsu University, No. 301, XueFu Road, Zhenjiang, Jiangsu, 212001, China.
| |
Collapse
|
4
|
Mao C, Liu X, Guo SW. Meclizine improves endometrial repair and reduces simulated menstrual bleeding in mice with induced adenomyosis. Am J Obstet Gynecol 2024; 231:113.e1-113.e13. [PMID: 38367751 DOI: 10.1016/j.ajog.2024.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/31/2024] [Accepted: 02/11/2024] [Indexed: 02/19/2024]
Abstract
BACKGROUND Adenomyosis is one of the structural causes of abnormal uterine bleeding, which often presents as heavy menstrual bleeding. Mostly because of the poor understanding of its pathophysiology, medical management of adenomyosis-induced heavy menstrual bleeding is still a challenge. We have previously reported that glycolysis is crucial to endometrial repair following menstruation and that suppressed glycolysis can cause heavy menstrual bleeding. OBJECTIVE This study aimed to test the hypothesis that meclizine, a drug with an excellent safety profile, alleviates heavy menstrual bleeding in mice with induced adenomyosis using a simulated menstruation model. STUDY DESIGN Adenomyosis was induced in 36 female C57BL/6 mice using endometrial-myometrial interface disruption. Three months after induction, the mice were randomly divided into the following 3 groups: low-dose meclizine, high-dose meclizine, and controls. Treatment with meclizine or vehicle started shortly before the simulated menstruation procedure and ended before progesterone withdrawal. The amount of blood loss was quantified and uterine tissue was harvested for histologic evaluation of the grade of endometrial repair. We performed immunohistochemistry analysis of 4 proteins critically involved in glycolysis: Glut1 (glucose transporter 1), Hk2 (hexokinase 2), Pfkfb3 (6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3), and Pkm2 (pyruvate kinase M2). The extent of tissue fibrosis in both ectopic and eutopic endometria was evaluated using Masson trichrome staining. RESULTS In mice with induced adenomyosis, meclizine accelerated endometrial repair in a dose-dependent manner and reduced the amount of menstrual bleeding. Meclizine administration raised endometrial immunoexpression of Hk2 and Pfkfb3 but not of Glut1 or Pkm2. The extent of endometrial fibrosis was reduced following the meclizine administration. Remarkably, these favorable changes were accompanied by the suppression of lesional progression, as evidenced by the dose-dependent reduction in the extent of fibrosis (a surrogate for lesional progression). CONCLUSION These encouraging results, taken together, suggest that glycolysis may be a promising therapeutic target and that meclizine may hold therapeutic potential as a nonhormonal treatment for adenomyosis-induced heavy menstrual bleeding without exacerbating the disease.
Collapse
Affiliation(s)
- Chenyu Mao
- Department of General Gynecology, Obstetrics & Gynecology Hospital, Fudan University, Shanghai, China
| | - Xishi Liu
- Department of General Gynecology, Obstetrics & Gynecology Hospital, Fudan University, Shanghai, China
| | - Sun-Wei Guo
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China; Research Institute, Obstetrics & Gynecology Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
5
|
Harrison DE, Strong R, Reifsnyder P, Rosenthal N, Korstanje R, Fernandez E, Flurkey K, Ginsburg BC, Murrell MD, Javors MA, Lopez-Cruzan M, Nelson JF, Willcox BJ, Allsopp R, Watumull DM, Watumull DG, Cortopassi G, Kirkland JL, Tchkonia T, Choi YG, Yousefzadeh MJ, Robbins PD, Mitchell JR, Acar M, Sarnoski EA, Bene MR, Salmon A, Kumar N, Miller RA. Astaxanthin and meclizine extend lifespan in UM-HET3 male mice; fisetin, SG1002 (hydrogen sulfide donor), dimethyl fumarate, mycophenolic acid, and 4-phenylbutyrate do not significantly affect lifespan in either sex at the doses and schedules used. GeroScience 2024; 46:795-816. [PMID: 38041783 PMCID: PMC10828146 DOI: 10.1007/s11357-023-01011-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/07/2023] [Indexed: 12/03/2023] Open
Abstract
In genetically heterogeneous (UM-HET3) mice produced by the CByB6F1 × C3D2F1 cross, the Nrf2 activator astaxanthin (Asta) extended the median male lifespan by 12% (p = 0.003, log-rank test), while meclizine (Mec), an mTORC1 inhibitor, extended the male lifespan by 8% (p = 0.03). Asta was fed at 1840 ± 520 (9) ppm and Mec at 544 ± 48 (9) ppm, stated as mean ± SE (n) of independent diet preparations. Both were started at 12 months of age. The 90th percentile lifespan for both treatments was extended in absolute value by 6% in males, but neither was significant by the Wang-Allison test. Five other new agents were also tested as follows: fisetin, SG1002 (hydrogen sulfide donor), dimethyl fumarate, mycophenolic acid, and 4-phenylbutyrate. None of these increased lifespan significantly at the dose and method of administration tested in either sex. Amounts of dimethyl fumarate in the diet averaged 35% of the target dose, which may explain the absence of lifespan effects. Body weight was not significantly affected in males by any of the test agents. Late life weights were lower in females fed Asta and Mec, but lifespan was not significantly affected in these females. The male-specific lifespan benefits from Asta and Mec may provide insights into sex-specific aspects of aging.
Collapse
Affiliation(s)
- David E Harrison
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA.
| | - Randy Strong
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, TX, USA
- Education, and Clinical Center, Geriatric Research, San Antonio, TX, USA
- Research Service, South Texas Veterans Health Care System, San Antonio, TX, USA
- Department of Pharmacology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Peter Reifsnyder
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Nadia Rosenthal
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Ron Korstanje
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Elizabeth Fernandez
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, TX, USA
- Education, and Clinical Center, Geriatric Research, San Antonio, TX, USA
- Department of Pharmacology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Kevin Flurkey
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Brett C Ginsburg
- Department of Psychiatry, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Meredith D Murrell
- Department of Psychiatry, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Martin A Javors
- Department of Psychiatry, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Marisa Lopez-Cruzan
- Department of Psychiatry, The University of Texas Health Science Center, San Antonio, TX, USA
| | - James F Nelson
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, TX, USA
- Department of Physiology, The University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Bradley J Willcox
- John A. Burns School of Medicine, University of Hawai'I at Mānoa, Honolulu, HI, USA
| | - Richard Allsopp
- John A. Burns School of Medicine, University of Hawai'I at Mānoa, Honolulu, HI, USA
| | | | | | - Gino Cortopassi
- Department of Molecular Biosciences, University of California, Davis, CA, USA
| | | | | | | | | | | | | | - Murat Acar
- Department of Basic Medical Sciences, School of Medicine, Koç University, 34450, Istanbul, Turkey
| | - Ethan A Sarnoski
- Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Michael R Bene
- Department of Molecular Medicine, The University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Adam Salmon
- Barshop Institute for Longevity and Aging Studies, The University of Texas Health Science Center, San Antonio, TX, USA
- Education, and Clinical Center, Geriatric Research, San Antonio, TX, USA
- Research Service, South Texas Veterans Health Care System, San Antonio, TX, USA
- Department of Molecular Medicine, The University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Navasuja Kumar
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
6
|
Liu J, Wu S, Zhang Y, Wang C, Liu S, Wan J, Yang L. SARS-CoV-2 viral genes Nsp6, Nsp8, and M compromise cellular ATP levels to impair survival and function of human pluripotent stem cell-derived cardiomyocytes. Stem Cell Res Ther 2023; 14:249. [PMID: 37705046 PMCID: PMC10500938 DOI: 10.1186/s13287-023-03485-3] [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: 03/08/2023] [Accepted: 08/30/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Cardiovascular complications significantly augment the overall COVID-19 mortality, largely due to the susceptibility of human cardiomyocytes (CMs) to SARS-CoV-2 virus. SARS-CoV-2 virus encodes 27 genes, whose specific impacts on CM health are not fully understood. This study elucidates the deleterious effects of SARS-CoV-2 genes Nsp6, M, and Nsp8 on human CMs. METHODS CMs were derived from human pluripotent stem cells (hPSCs), including human embryonic stem cells and induced pluripotent stem cells, using 2D and 3D differentiation methods. We overexpressed Nsp6, M, or Nsp8 in hPSCs and then applied whole mRNA-seq and mass spectrometry for multi-omics analysis. Co-immunoprecipitation mass spectrometry was utilized to map the protein interaction networks of Nsp6, M, and Nsp8 within host hiPSC-CMs. RESULTS Nsp6, Nsp8, and M globally perturb the transcriptome and proteome of hPSC-CMs. SARS-CoV-2 infection and the overexpression of Nsp6, Nsp8, or M coherently upregulated genes associated with apoptosis and immune/inflammation pathways, whereas downregulated genes linked to heart contraction and functions. Global interactome analysis revealed interactions between Nsp6, Nsp8, and M with ATPase subunits. Overexpression of Nsp6, Nsp8, or M significantly reduced cellular ATP levels, markedly increased apoptosis, and compromised Ca2+ handling in hPSC-CMs. Importantly, administration of FDA-approved drugs, ivermectin and meclizine, could restore ATP levels, thereby mitigating apoptosis and dysfunction in hPSC-CMs overexpressing Nsp6, Nsp8, or M. CONCLUSION Overall, our findings uncover the extensive damaging effects of Nsp6, Nsp8, and M on hPSC-CMs, underlining the crucial role of ATP homeostasis in CM death and functional abnormalities induced by these SARS-CoV-2 genes, and reveal the potential therapeutic strategies to alleviate these detrimental effects with FDA-approved drugs.
Collapse
Affiliation(s)
- Juli Liu
- Department of Pediatrics, Indiana University School of Medicine, Herman B Wells Center for Pediatric Research, Indianapolis, IN, 46202, USA.
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
| | - Shiyong Wu
- Department of Pediatrics, Indiana University School of Medicine, Herman B Wells Center for Pediatric Research, Indianapolis, IN, 46202, USA
| | - Yucheng Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Cheng Wang
- Department of Pediatrics, Indiana University School of Medicine, Herman B Wells Center for Pediatric Research, Indianapolis, IN, 46202, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Lei Yang
- Department of Pediatrics, Indiana University School of Medicine, Herman B Wells Center for Pediatric Research, Indianapolis, IN, 46202, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
7
|
Liu S, Yang X, Chen F, Cai ZY. Dysfunction of the neurovascular unit in brain aging. J Biomed Res 2022; 37:153-165. [PMID: 37198158 DOI: 10.7555/jbr.36.20220105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023] Open
Abstract
An emerging concept termed the neurovascular unit (NVU) underlines neurovascular coupling. It has been reported that NVU impairment can result in neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease. Aging is a complex and irreversible process caused by programmed and damage-related factors. Loss of biological functions and increased susceptibility to additional neurodegenerative diseases are major characteristics of aging. In this review, we describe the basics of the NVU and discuss the effect of aging on NVU basics. Furthermore, we summarize the mechanisms that increase NVU susceptibility to neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease. Finally, we discuss new treatments for neurodegenerative diseases and methods of maintaining an intact NVU that may delay or diminish aging.
Collapse
Affiliation(s)
- Shu Liu
- Chongqing Medical University, Chongqing 400042, China
- Chongqing Institute Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing 400714, China
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Xu Yang
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Fei Chen
- Chongqing Medical University, Chongqing 400042, China
- Chongqing Institute Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Zhi-You Cai
- Chongqing Medical University, Chongqing 400042, China
- Chongqing Institute Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing 400714, China
- Department of Neurology, Chongqing General Hospital, Chongqing 400013, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| |
Collapse
|
8
|
Shannonhouse J, Bernabucci M, Gomez R, Son H, Zhang Y, Ai CH, Ishida H, Kim YS. Meclizine and Metabotropic Glutamate Receptor Agonists Attenuate Severe Pain and Ca 2+ Activity of Primary Sensory Neurons in Chemotherapy-Induced Peripheral Neuropathy. J Neurosci 2022; 42:6020-6037. [PMID: 35772967 PMCID: PMC9351649 DOI: 10.1523/jneurosci.1064-21.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/02/2022] [Accepted: 06/22/2022] [Indexed: 02/05/2023] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) affects ∼68% of patients undergoing chemotherapy, causing debilitating neuropathic pain and reducing quality of life. Cisplatin is a commonly used platinum-based chemotherapeutic drug known to cause CIPN, possibly by causing oxidative stress damage to primary sensory neurons. Metabotropic glutamate receptors (mGluRs) are widely hypothesized to be involved in pain processing and pain mitigation. Meclizine is an H1 histamine receptor antagonist known to have neuroprotective effects, including an anti-oxidative effect. Here, we used a mouse model of cisplatin-induced CIPN using male and female mice to test agonists of mGluR8 and Group II mGluR as well as meclizine as interventions to reduce cisplatin-induced pain. We performed behavioral pain tests, and we imaged Ca2+ activity of the large population of dorsal root ganglia (DRG) neurons in vivo For the latter, we used a genetically-encoded Ca2+ indicator, Pirt-GCaMP3, which enabled us to monitor different drug interventions at the level of the intact DRG neuronal ensemble. We found that CIPN increased spontaneous Ca2+ activity in DRG neurons, increased number of Ca2+ transients, and increased hyper-responses to mechanical, thermal, and chemical stimuli. We found that mechanical and thermal pain caused by CIPN was significantly attenuated by the mGluR8 agonist, (S)-3,4-DCPG, the Group II mGluR agonist, LY379268, and the H1 histamine receptor antagonist, meclizine. DRG neuronal Ca2+ activity elevated by CIPN was attenuated by LY379268 and meclizine, but not by (S)-3,4-DCPG. Furthermore, meclizine and LY379268 attenuated cisplatin-induced weight loss. These results suggest that Group II mGluR agonist, mGluR8 agonist, and meclizine are promising candidates as new treatment options for CIPN, and studies of their mechanisms are warranted.SIGNIFICANCE STATEMENT Chemotherapy-induced peripheral neuropathy (CIPN) is a painful condition that affects most chemotherapy patients and persists several months or longer after treatment ends. Research on CIPN mechanism is extensive but has produced only few clinically useful treatments. Using in vivo GCaMP Ca2+ imaging in live animals over 1800 neurons/dorsal root ganglia (DRG) at once, we have characterized the effects of the chemotherapeutic drug, cisplatin and three treatments that decrease CIPN pain. Cisplatin increases sensory neuronal Ca2+ activity and develops various sensitization. Metabotropic glutamate receptor (mGluR) agonist, LY379268 or the H1 histamine receptor antagonist, meclizine decreases cisplatin's effects on neuronal Ca2+ activity and reduces pain hypersensitivity. Our results and experiments provide insights into cellular effects of cisplatin and drugs preventing CIPN pain.
Collapse
Affiliation(s)
| | - Matteo Bernabucci
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854
| | - Ruben Gomez
- Department of Oral & Maxillofacial Surgery, School of Dentistry
| | - Hyeonwi Son
- Department of Oral & Maxillofacial Surgery, School of Dentistry
| | - Yan Zhang
- Department of Oral & Maxillofacial Surgery, School of Dentistry
| | - Chih-Hsuan Ai
- Department of Oral & Maxillofacial Surgery, School of Dentistry
| | - Hirotake Ishida
- Department of Oral & Maxillofacial Surgery, School of Dentistry
| | - Yu Shin Kim
- Department of Oral & Maxillofacial Surgery, School of Dentistry
- Programs in Integrated Biomedical Sciences, Translational Sciences, Biomedical Engineering, Radiological Sciences, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| |
Collapse
|
9
|
Liu J, Zhang Y, Han L, Guo S, Wu S, Doud EH, Wang C, Chen H, Rubart-von der Lohe M, Wan J, Yang L. Genome-wide analyses reveal the detrimental impacts of SARS-CoV-2 viral gene Orf9c on human pluripotent stem cell-derived cardiomyocytes. Stem Cell Reports 2022; 17:522-537. [PMID: 35180394 PMCID: PMC8851680 DOI: 10.1016/j.stemcr.2022.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/17/2022] Open
Abstract
Patients with coronavirus disease 2019 (COVID-19) commonly have manifestations of heart disease. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome encodes 27 proteins. Currently, SARS-CoV-2 gene-induced abnormalities of human heart muscle cells remain elusive. Here, we comprehensively characterized the detrimental effects of a SARS-CoV-2 gene, Orf9c, on human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) by preforming multi-omic analyses. Transcriptomic analyses of hPSC-CMs infected by SARS-CoV-2 with Orf9c overexpression (Orf9cOE) identified concordantly up-regulated genes enriched into stress-related apoptosis and inflammation signaling pathways, and down-regulated CM functional genes. Proteomic analysis revealed enhanced expressions of apoptotic factors, whereas reduced protein factors for ATP synthesis by Orf9cOE. Orf9cOE significantly reduced cellular ATP level, induced apoptosis, and caused electrical dysfunctions of hPSC-CMs. Finally, drugs approved by the U.S. Food and Drug Administration, namely, ivermectin and meclizine, restored ATP levels and ameliorated CM death and functional abnormalities of Orf9cOE hPSC-CMs. Overall, we defined the molecular mechanisms underlying the detrimental impacts of Orf9c on hPSC-CMs and explored potentially therapeutic approaches to ameliorate Orf9c-induced cardiac injury and abnormalities.
Collapse
Affiliation(s)
- Juli Liu
- Department of Pediatrics, Indiana University School of Medicine, Herman B Wells Center for Pediatric Research, Indianapolis, IN 46202, USA
| | - Yucheng Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Lei Han
- Department of Pediatrics, Indiana University School of Medicine, Herman B Wells Center for Pediatric Research, Indianapolis, IN 46202, USA
| | - Shuai Guo
- Department of Pediatrics, Indiana University School of Medicine, Herman B Wells Center for Pediatric Research, Indianapolis, IN 46202, USA
| | - Shiyong Wu
- Department of Pediatrics, Indiana University School of Medicine, Herman B Wells Center for Pediatric Research, Indianapolis, IN 46202, USA
| | - Emma Helen Doud
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Cheng Wang
- Department of Pediatrics, Indiana University School of Medicine, Herman B Wells Center for Pediatric Research, Indianapolis, IN 46202, USA
| | - Hanying Chen
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Michael Rubart-von der Lohe
- Department of Pediatrics, Indiana University School of Medicine, Herman B Wells Center for Pediatric Research, Indianapolis, IN 46202, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Lei Yang
- Department of Pediatrics, Indiana University School of Medicine, Herman B Wells Center for Pediatric Research, Indianapolis, IN 46202, USA.
| |
Collapse
|
10
|
Solana-Manrique C, Sanz FJ, Ripollés E, Bañó MC, Torres J, Muñoz-Soriano V, Paricio N. Enhanced activity of glycolytic enzymes in Drosophila and human cell models of Parkinson's disease based on DJ-1 deficiency. Free Radic Biol Med 2020; 158:137-148. [PMID: 32726690 DOI: 10.1016/j.freeradbiomed.2020.06.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 01/07/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative debilitating disorder characterized by progressive disturbances in motor, autonomic and psychiatric functions. One of the genes involved in familial forms of the disease is DJ-1, whose mutations cause early-onset PD. Besides, it has been shown that an over-oxidized and inactive form of the DJ-1 protein is found in brains of sporadic PD patients. Interestingly, the DJ-1 protein plays an important role in cellular defense against oxidative stress and also participates in mitochondrial homeostasis. Valuable insights into potential PD pathogenic mechanisms involving DJ-1 have been obtained from studies in cell and animal PD models based on DJ-1 deficiency such as Drosophila. Flies mutant for the DJ-1β gene, the Drosophila ortholog of human DJ-1, exhibited disease-related phenotypes such as motor defects, increased reactive oxygen species production and high levels of protein carbonylation. In the present study, we demonstrate that DJ-1β mutants also show a significant increase in the activity of several regulatory glycolytic enzymes. Similar results were obtained in DJ-1-deficient SH-SY5Y neuroblastoma cells, thus suggesting that loss of DJ-1 function leads to an increase in the glycolytic rate. In such a scenario, an enhancement of the glycolytic pathway could be a protective mechanism to decrease ROS production by restoring ATP levels, which are decreased due to mitochondrial dysfunction. Our results also show that meclizine and dimethyl fumarate, two FDA-approved compounds with different clinical applications, are able to attenuate PD-related phenotypes in both models. Moreover, we found that they may exert their beneficial effect by increasing glycolysis through the activation of key glycolytic enzymes. Taken together, these results are consistent with the idea that increasing glycolysis could be a potential disease-modifying strategy for PD, as recently suggested. Besides, they also support further evaluation and potential repurposing of meclizine and dimethyl fumarate as modulators of energy metabolism for neuroprotection in PD.
Collapse
Affiliation(s)
- Cristina Solana-Manrique
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain; Estructura de Recerca Interdisciplinar en Biotecnologia I Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100, Burjassot, Spain
| | - Francisco José Sanz
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain; Estructura de Recerca Interdisciplinar en Biotecnologia I Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100, Burjassot, Spain
| | - Edna Ripollés
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain; Estructura de Recerca Interdisciplinar en Biotecnologia I Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100, Burjassot, Spain
| | - M Carmen Bañó
- Estructura de Recerca Interdisciplinar en Biotecnologia I Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100, Burjassot, Spain; Departamento de Bioquímica y Biología Molecular, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain
| | - Josema Torres
- Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, 46100, Burjassot, Spain
| | - Verónica Muñoz-Soriano
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain; Estructura de Recerca Interdisciplinar en Biotecnologia I Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100, Burjassot, Spain
| | - Nuria Paricio
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain; Estructura de Recerca Interdisciplinar en Biotecnologia I Biomedicina (ERI BIOTECMED), Universidad de Valencia, 46100, Burjassot, Spain.
| |
Collapse
|
11
|
Salama AH, Elmotasem H, Salama AAA. Nanotechnology based blended chitosan-pectin hybrid for safe and efficient consolidative antiemetic and neuro-protective effect of meclizine hydrochloride in chemotherapy induced emesis. Int J Pharm 2020; 584:119411. [PMID: 32423876 DOI: 10.1016/j.ijpharm.2020.119411] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 12/23/2022]
Abstract
The aim of this study was to formulate an easily-administered, safe and effective dosage form loaded with meclizine for treatment of chemotherapy-induced nausea and vomiting (CINV) through the buccal route. CINV comprises bothersome side effects accompanying cytotoxic drugs administration in cancer patients. Meclizine was loaded in chitosan-pectin nanoparticles which were further incorporated within a buccal film. Different formulations were prepared based on a 21.31 full factorial study using Design Expert®8. The optimum formulation possessed favorable characters regarding its particle size (129 nm), entrapment efficiency (90%) and release profile. Moreover, its permeation efficiency through sheep buccal mucosa was assessed via Franz cell diffusion and confocal laser microscopy methods. Enhanced permeation was achieved compared with the free drug form. In-vivo performance was assessed using cyclophosphamide induced emesis. The proposed formulation exerted significant relief of the measured responses (reduced body weight and motor coordination, elevated emesis, anorexia, proinflammatory mediators and neurotransmitters that were also associated with scattered degenerated neurons and glial cells). The developed formulation ameliorated all behavioral, biochemical and histopathological changes induced by cyclophosphamide. The obtained data were promising suggesting that our bioadhesive formulation can offer an auspicious medication for treating distressing symptoms associated with chemotherapy for cancer patients.
Collapse
Affiliation(s)
- Alaa H Salama
- Pharmaceutical Technology Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Dokki, Cairo 12622, Egypt; Department of Pharmaceutics, Faculty of Pharmacy, Ahram Canadian University, 6(th) of October City, Cairo, Egypt.
| | - Heba Elmotasem
- Pharmaceutical Technology Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Abeer A A Salama
- Pharmacology Department, Medical Research Division, National Research Centre, Dokki, Cairo 12622, Egypt
| |
Collapse
|
12
|
Zhuo M, Gorgun FM, Tyler DS, Englander EW. Hypoxia potentiates the capacity of melanoma cells to evade cisplatin and doxorubicin cytotoxicity via glycolytic shift. FEBS Open Bio 2020; 10:789-801. [PMID: 32134564 PMCID: PMC7193165 DOI: 10.1002/2211-5463.12830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/27/2020] [Accepted: 03/02/2020] [Indexed: 12/11/2022] Open
Abstract
The hypoxic environment within solid tumors impedes the efficacy of chemotherapeutic treatments. Here, we demonstrate that hypoxia augments the capacity of melanoma cells to withstand cisplatin and doxorubicin cytotoxicity. We show that B16F10 cells derived from spontaneously formed melanoma and YUMM1.7 cells, engineered to recapitulate human‐relevant melanoma driver mutations, profoundly differ in their vulnerabilities to cisplatin and doxorubicin. The differences are manifested in magnitude of proliferative arrest and cell death rates, extent of mtDNA depletion, and impairment of mitochondrial respiration. In both models, cytotoxicity is mitigated by hypoxia, which augments glycolytic metabolism. Collectively, the findings implicate metabolic reprogramming in drug evasion and suggest that melanoma tumors with distinct genetic makeup may have differential drug vulnerabilities, highlighting the importance of precision anticancer treatments.
Collapse
Affiliation(s)
- Ming Zhuo
- Department of SurgeryUniversity of Texas Medical BranchGalvestonTXUSA
| | - Falih M. Gorgun
- Department of SurgeryUniversity of Texas Medical BranchGalvestonTXUSA
| | - Douglas S. Tyler
- Department of SurgeryUniversity of Texas Medical BranchGalvestonTXUSA
| | - Ella W. Englander
- Department of SurgeryUniversity of Texas Medical BranchGalvestonTXUSA
| |
Collapse
|
13
|
Liu B, Bai W, Ou G, Zhang J. Cdh1-Mediated Metabolic Switch from Pentose Phosphate Pathway to Glycolysis Contributes to Sevoflurane-Induced Neuronal Apoptosis in Developing Brain. ACS Chem Neurosci 2019; 10:2332-2344. [PMID: 30741526 DOI: 10.1021/acschemneuro.8b00644] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cdh1 is a regulatory subunit of the anaphase promoting complex/cyclosome (APC/C), known to be involved in regulating neuronal survival. The role of Cdh1 in volatile anesthetics-induced neuronal apoptosis in the developing brain is unknown. In this study, we used postnatal day 7 (P7) and day 21 (P21) mice exposed to 2.3% sevoflurane for 6 h to investigate at which age and duration of exposure sevoflurane affects the expression of Cdh1 and glycolytic enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) and that of the pentose phosphate pathway (PPP) enzyme, glucose-6-phosphate dehydrogenase (G6PD). Furthermore, we tested whether the cyclin-dependent kinases (cdks) inhibitor roscovatine could counteract the effects caused by exposure to sevoflurane. Finally, we applied the glycolysis inhibitor 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3-PO), G6PD inhibitor dehydroepiandrosterone (DHEA), and exogenous reduced glutathione to examine the contribution of the glycolysis pathway and PPP to sevoflurane-induced neuroapoptosis. We found that prolonged sevoflurane anesthesia significantly reduces the Cdh1 level in P7 mice compared to in the P21 ones; moreover, the decrease in Cdh1 level results in a switch in glucose metabolism from the PPP to neuronal glycolysis. This leads to an imbalance between reactive oxygen species production and reduced glutathione level in the developing brain, which is more susceptible to oxidative stress. As a result, sevoflurane induces neuroapoptosis through Cdh1-mediated glucose metabolism reprogramming. Our study demonstrates a critical role of Cdh1 in sevoflurane-induced neuroapoptosis by shifting PPP to the glycolytic pathway in the developing brain. These findings suggest that Cdh1 may be a novel target for preventing volatile anesthetics-induced neurotoxicity and memory impairment.
Collapse
Affiliation(s)
- Bin Liu
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China 200040
| | - Wenjie Bai
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China 200040
| | - Guoyao Ou
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China 200040
| | - Jun Zhang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China 200040
| |
Collapse
|
14
|
Zhuo M, Gorgun MF, Englander EW. Neurotoxicity of cytarabine (Ara-C) in dorsal root ganglion neurons originates from impediment of mtDNA synthesis and compromise of mitochondrial function. Free Radic Biol Med 2018; 121:9-19. [PMID: 29698743 PMCID: PMC5971160 DOI: 10.1016/j.freeradbiomed.2018.04.570] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/12/2018] [Accepted: 04/21/2018] [Indexed: 12/18/2022]
Abstract
Peripheral Nervous System (PNS) neurotoxicity caused by cancer drugs hinders attainment of chemotherapy goals. Due to leakiness of the blood nerve barrier, circulating chemotherapeutic drugs reach PNS neurons and adversely affect their function. Chemotherapeutic drugs are designed to target dividing cancer cells and mechanisms underlying their toxicity in postmitotic neurons remain to be fully clarified. The objective of this work was to elucidate progression of events triggered by antimitotic drugs in postmitotic neurons. For proof of mechanism study, we chose cytarabine (ara-C), an antimetabolite used in treatment of hematological cancers. Ara-C is a cytosine analog that terminates DNA synthesis. To investigate how ara-C affects postmitotic neurons, which replicate mitochondrial but not genomic DNA, we adapted a model of Dorsal Root Ganglion (DRG) neurons. We showed that DNA polymerase γ, which is responsible for mtDNA synthesis, is inhibited by ara-C and that sublethal ara-C exposure of DRG neurons leads to reduction in mtDNA content, ROS generation, oxidative mtDNA damage formation, compromised mitochondrial respiration and diminution of NADPH and GSH stores, as well as, activation of the DNA damage response. Hence, it is plausible that in ara-C exposed DRG neurons, ROS amplified by the high mitochondrial content shifts from physiologic to pathologic levels signaling stress to the nucleus. Combined, the findings suggest that ara-C neurotoxicity in DRG neurons originates in mitochondria and that continuous mtDNA synthesis and reliance on oxidative phosphorylation for energy needs sensitize the highly metabolic neurons to injury by mtDNA synthesis terminating cancer drugs.
Collapse
Affiliation(s)
- Ming Zhuo
- Division of Neurosurgery, Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | - Murat F Gorgun
- Division of Neurosurgery, Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | - Ella W Englander
- Division of Neurosurgery, Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
15
|
Gorgun MF, Zhuo M, Cortez I, Dineley KT, Englander EW. Acute inhalation of combustion smoke triggers neuroinflammation and persistent anxiety-like behavior in the mouse. Inhal Toxicol 2018; 29:598-610. [PMID: 29405081 DOI: 10.1080/08958378.2018.1432728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
CONTEXT Acute inhalation of combustion smoke triggers neurologic sequelae in survivors. Due to the challenges posed by heterogeneity of smoke exposures in humans, mechanistic links between acute smoke inhalation and neuropathologic sequelae have not been systematically investigated. METHODS Here, using mouse model of acute inhalation of combustion smoke, we studied longitudinal neurobehavioral manifestations of smoke exposures and molecular/cellular changes in the mouse brain. RESULTS Immunohistochemical analyses at eight months post-smoke, revealed hippocampal astrogliosis and microgliosis accompanied by reduced myelination. Elevated expression of proinflammatory cytokines was also detected. Longitudinal testing in different neurobehavioral paradigms in the course of post-smoke recovery, revealed lasting anxiety-like behavior. The examined paradigms included the open field exploration/anxiety testing at two, four and six months post-smoke, which detected decreases in total distance traveled and time spent in the central arena in the smoke-exposed compared to sham-control mice, suggestive of dampened exploratory activity and increased anxiety-like behavior. In agreement with reduced open field activity, cued fear conditioning test revealed increased freezing in response to conditioned auditory stimulus in mice after acute smoke inhalation. Similarly, elevated plus maze testing demonstrated lesser presence in open arms of the maze, consistent with anxiety-like behavior, for the post-smoke exposure mice. CONCLUSIONS Taken together, our data demonstrate for the first time persistent neurobehavioral manifestations of acute inhalation of combustion smoke and provide new insights into long-term progression of events initiated by disrupted brain oxygenation that might contribute to lasting adverse sequelae in survivors of smoke inhalation injuries.
Collapse
Affiliation(s)
- Murat F Gorgun
- a Department of Surgery , University of Texas Medical Branch , Galveston , TX , USA
| | - Ming Zhuo
- a Department of Surgery , University of Texas Medical Branch , Galveston , TX , USA
| | - IbDanelo Cortez
- b Department of Neurology , University of Texas Medical Branch , Galveston , TX , USA.,c Mitchell Center for Neurodegenerative Diseases , University of Texas Medical Branch , Galveston , TX , USA.,d Center for Addiction Research , University of Texas Medical Branch , Galveston , TX , USA
| | - Kelly T Dineley
- b Department of Neurology , University of Texas Medical Branch , Galveston , TX , USA.,c Mitchell Center for Neurodegenerative Diseases , University of Texas Medical Branch , Galveston , TX , USA.,d Center for Addiction Research , University of Texas Medical Branch , Galveston , TX , USA
| | - Ella W Englander
- a Department of Surgery , University of Texas Medical Branch , Galveston , TX , USA.,e Shriners Hospitals for Children and University of Texas Medical Branch , Galveston , TX , USA
| |
Collapse
|
16
|
Translesion Synthesis DNA Polymerase Kappa Is Indispensable for DNA Repair Synthesis in Cisplatin Exposed Dorsal Root Ganglion Neurons. Mol Neurobiol 2017; 55:2506-2515. [PMID: 28391554 DOI: 10.1007/s12035-017-0507-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 03/21/2017] [Indexed: 10/19/2022]
Abstract
In the peripheral nervous system (PNS) in the absence of tight blood barrier, neurons are at increased risk of DNA damage, yet the question of how effectively PNS neurons manage DNA damage remains largely unanswered. Genotoxins in systemic circulation include chemotherapeutic drugs that reach peripheral neurons and damage their DNA. Because neurotoxicity of platinum-based class of chemotherapeutic drugs has been implicated in PNS neuropathies, we utilized an in vitro model of Dorsal Root Ganglia (DRGs) to investigate how peripheral neurons respond to cisplatin that forms intra- and interstrand crosslinks with their DNA. Our data revealed strong transcriptional upregulation of the translesion synthesis DNA polymerase kappa (Pol κ), while expression of other DNA polymerases remained unchanged. DNA Pol κ is involved in bypass synthesis of diverse DNA lesions and considered a vital player in cellular survival under injurious conditions. To assess the impact of Pol κ deficiency on cisplatin-exposed DRG neurons, Pol κ levels were reduced using siRNA. Pol κ targeting siRNA diminished the cisplatin-induced nuclear Pol κ immunoreactivity in DRG neurons and decreased the extent of cisplatin-induced DNA repair synthesis, as reflected in reduced incorporation of thymidine analog into nuclear DNA. Moreover, Pol κ depletion exacerbated global transcriptional suppression induced by cisplatin in DRG neurons. Collectively, these findings provide the first evidence for critical role of Pol κ in DNA damage response in the nervous system and call attention to implications of polymorphisms that modify Pol κ activity, on maintenance of genomic integrity and neuronal function in exogenously challenged PNS.
Collapse
|
17
|
Cisplatin Toxicity in Dorsal Root Ganglion Neurons Is Relieved by Meclizine via Diminution of Mitochondrial Compromise and Improved Clearance of DNA Damage. Mol Neurobiol 2016; 54:7883-7895. [PMID: 27858292 DOI: 10.1007/s12035-016-0273-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/30/2016] [Indexed: 01/09/2023]
Abstract
Chemotherapy-induced neurotoxicity of peripheral nervous system (PNS) hinders efficacy of cancer treatments. Mechanisms initiating PNS injury by anticancer drugs are incompletely understood delaying development of effective management strategies. To understand events triggered in PNS by cancer drugs, we exposed dorsal root ganglion (DRG) neurons to cisplatin, a drug from platinum-based class of chemotherapeutics frequently implicated in peripheral neuropathies. While cisplatin enters cancer cells and forms cisplatin/DNA crosslinks that block cell proliferation, circulating cisplatin can also reach the PNS and produce crosslinks that impede critical DNA transactions in postmitotic neurons. Cisplatin forms crosslinks with both, nuclear and mitochondrial DNA (mtDNA). Crosslinks are repairable primarily via the nucleotide excision repair (NER) pathway, which is present in nuclei but absent from mitochondrial compartment. Hence, high mitochondrial content and limited shielding by blood nerve barrier make DRG neurons particularly vulnerable to mitochondrial injury by cisplatin. We report that in DRG neurons, cisplatin elevates reactive oxygen species, depletes mtDNA, and impairs mitochondrial respiration, whereas concomitant meclizine supplementation preserves redox balance, attenuates mitochondrial compromise, and augments DNA repair. Meclizine is an antihistamine drug recently implicated in neuroprotection via modulation of energy metabolism. Our data demonstrate that in the mitochondria-rich DRG neurons, meclizine mitigates cisplatin-induced mitochondrial compromise via enhancement of pentose phosphate pathway and repletion of nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione stores. The findings suggest that meclizine-mediated preservation of redox balance sustains mitochondrial respiration and supports execution of cellular processes, including timely removal of cisplatin crosslinks from nuclear DNA, thereby attenuating cisplatin toxicity in DRG neurons. Collectively, the findings reveal potential for pharmacologic modulation of dorsal root ganglion neurons metabolism for protection against toxicity of chemotherapeutic drugs.
Collapse
|