1
|
Xu S, Chen J, Yue S, Zhang Y, Zhao S, Hu Y, Zhang C, Guan W, Zhang L, Zhang L, Liang C. Alcohol intake exacerbates experimental autoimmune prostatitis through activating PI3K/AKT/mTOR pathway-mediated Th1 differentiation. Front Immunol 2025; 15:1512456. [PMID: 39872540 PMCID: PMC11770681 DOI: 10.3389/fimmu.2024.1512456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025] Open
Abstract
Background Epidemiological investigations have revealed a significant association between alcohol consumption and chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS). Nevertheless, the potential mechanisms are still inadequately revealed. This research aimed to investigate the impact of alcohol on CP/CPPS using an animal model and to elucidate the underlying mechanisms. Methods We first established the widely used animal model for CP/CPPS, experimental autoimmune prostatitis (EAP). During the induction of EAP, mice were fed with alcohol or control diet. The HE staining, ELISA, and behavioral experiments were employed to assess the severity of inflammation in EAP mice and EAP-alcohol mice. Patients with a history of chronic alcohol consumption were also included to evaluate the effects of chronic alcohol consumption on CP/CPPS. Subsequently, proteomic analysis, flow cytometry, immunofluorescence, Western blotting, and immunohistochemistry were utilized to investigate the underlying mechanism involved both in vivo and in vitro. Results HE staining, ELISA, and behavioral experiments showed that alcohol exacerbated the severity of EAP in mice and patients. Proteomic and KEGG pathway analyses showed that abnormal Th1 differentiation and PI3K/AKT/mTOR pathway were significantly enriched. Subsequent mechanistic research showed that alcohol significantly activated PI3K/AKT/mTOR pathway and increased the Th1 cell differentiation both in vivo and in vitro. In contrast, PI3K inhibitor LY294002 and shRNA-PI3K plasmid inhibited PI3K/AKT/mTOR pathway activation, reduced Th1 cell differentiation, and alleviated EAP inflammation severity, respectively. Conclusion Our study is the first to demonstrate that alcohol intake promotes Th1 cell differentiation and exacerbates EAP by activating the PI3K/AKT/mTOR pathway. Additionally, the role of LY294002 in inhibiting PI3K/AKT/mTOR pathway to relieve EAP suggests that it can serve as a promising therapeutic target for CP/CPPS.
Collapse
Affiliation(s)
- Shun Xu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, China
| | - Jing Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, China
| | - Shaoyu Yue
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, China
| | - Yifan Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, China
| | - Shengyu Zhao
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, China
| | - Yongtao Hu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, China
| | - Cheng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, China
| | - Wenrui Guan
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, China
| | - Li Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, China
| | - Ligang Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, Anhui, China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, China
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
2
|
Roychowdhury S, Pant B, Cross E, Scheraga R, Vachharajani V. Effect of ethanol exposure on innate immune response in sepsis. J Leukoc Biol 2024; 115:1029-1041. [PMID: 38066660 PMCID: PMC11136611 DOI: 10.1093/jleuko/qiad156] [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: 05/10/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 01/06/2024] Open
Abstract
Alcohol use disorder, reported by 1 in 8 critically ill patients, is a risk factor for death in sepsis patients. Sepsis, the leading cause of death, kills over 270,000 patients in the United States alone and remains without targeted therapy. Immune response in sepsis transitions from an early hyperinflammation to persistent inflammation and immunosuppression and multiple organ dysfunction during late sepsis. Innate immunity is the first line of defense against pathogen invasion. Ethanol exposure is known to impair innate and adaptive immune response and bacterial clearance in sepsis patients. Specifically, ethanol exposure is known to modulate every aspect of innate immune response with and without sepsis. Multiple molecular mechanisms are implicated in causing dysregulated immune response in ethanol exposure with sepsis, but targeted treatments have remained elusive. In this article, we outline the effects of ethanol exposure on various innate immune cell types in general and during sepsis.
Collapse
Affiliation(s)
- Sanjoy Roychowdhury
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Bishnu Pant
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Emily Cross
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
| | - Rachel Scheraga
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
- Department of Pulmonary and Critical Care Medicine, Integrated Hospital-Care Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland OH 44195, United States
| | - Vidula Vachharajani
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, United States
- Department of Pulmonary and Critical Care Medicine, Integrated Hospital-Care Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland OH 44195, United States
| |
Collapse
|
3
|
Liu R, Zhao B, Zhao J, Zhang M. Ethanol causes non-communicable disease through activation of NLRP3 inflammasome: a review on mechanism of action and potential interventions. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2024; 50:139-149. [PMID: 38237017 DOI: 10.1080/00952990.2023.2297349] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/16/2023] [Indexed: 04/28/2024]
Abstract
Background: Ethanol exposure has been suggested to be implicated in the initiation and progression of several non-communicable diseases (NCD), including neurological disorders, diabetes mellitus, alcoholic liver disease, gastric injury, pancreatitis, and atherosclerosis. Recent findings show that the NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome is involved in the progression of ethanol-induced NCDs.Objective: The aim of this review was to summarize the research progress on NCDs associated with the action of the NLRP3 inflammasome by ethanol and potential interventions, with a specific focus on preclinical literature.Methods: A literature search was conducted on PubMed using the keywords "[ethanol] and [NLRP3]" up until January 2023. Articles describing cases of NCDs caused by ethanol and associated with the NLRP3 inflammasome were included.Results: After removing duplicates, 35 articles were included in this review. These studies, mostly conducted in animals or in vitro, provide evidence that ethanol can contribute to the development of NCDs, such as neurological disorders, alcoholic liver disease, gastric injury, pancreatitis, and atherosclerosis, by activating the NLRP3 inflammasome. Ethanol exposure primarily triggers NLRP3 inflammasome activation by influencing the TRL/NF-κB, ROS-TXNIP-NLRP3 and P2X7 receptor (P2X7R) signaling pathways. Several natural extracts and compounds have been found to alleviate NCDs caused by ethanol consumption by inhibiting the activation of the NLRP3 inflammasome.Conclusion: Preclinical research supports a role for ethanol-induced NLRP3 inflammasome in the development of NCDs. However, the clinical relevance remains uncertain in the relative absence of clinical studies.
Collapse
Affiliation(s)
- Ruizi Liu
- School of Clinical Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Bin Zhao
- Hospital of Shandong First Medical University, Jinan, Shandong Province, China
| | - Jie Zhao
- Hospital of Shandong First Medical University, Jinan, Shandong Province, China
| | - Meng Zhang
- School of Clinical Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| |
Collapse
|
4
|
Cao F, Wang Y, Song Y, Xu F, Xie Q, Jiang M, Liu X, Zhang D, Xu L. Celastrol Treatment Ameliorated Acute Ischemic Stroke-Induced Brain Injury by Microglial Injury Inhibition and Nrf2/HO-1 Pathway Activations. BIOMED RESEARCH INTERNATIONAL 2023; 2023:1076522. [PMID: 37082194 PMCID: PMC10113063 DOI: 10.1155/2023/1076522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 12/08/2022] [Accepted: 01/20/2023] [Indexed: 04/22/2023]
Abstract
Background Stroke is the third main reason of mortality, which is the leading reason for adult disability in the globe. Poststroke inflammation is well known to cause acute ischemic stroke- (AIS-) induced brain injury (BI) exacerbation. Celastrol (CL) has exhibited anti-inflammatory activities in various inflammatory traits though underlying mechanisms remain unknown. So, the present investigation is aimed at studying CL protective mechanism against AIS-induced BI. Methods A mouse model regarding middle cerebral artery occlusion and an oxygen-glucose deprivation (OGD) cell model with or not CL treatment were constructed to study CL protective effects. NF-E2-related factor 2 (Nrf2) was then silenced in BV2 microglia cells (BV2) to study Nrf2 role regarding CL-mediated neuroprotection. Results The results showed that CL treatment suppressed AIS-induced BI by inhibiting NLRP3/caspase-1 pathway activations and induction of apoptosis and pyroptosis in vivo and in vitro. NLRP3/caspase-1 pathway blocking activation suppressed OGD-induced cell pyroptosis and apoptosis. Also, CL treatment reversed OGD-induced microglial injury by promoting Nrf2/heme oxygenase-1 (HO-1) pathway activations. Nrf2 downregulation reversed CL protective effects against OGD-induced microglial injury, pyroptosis, and apoptosis. Conclusion The findings advise that CL treatment ameliorated AIS-induced BI by inhibiting microglial injury and activating the Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Fanfan Cao
- Sino-French Cooperative Central Lab, Gongli Hospital of Shanghai Pudong New Area, No. 207, Juye Rd., Pudong New District, Shanghai 200135, China
| | - Ying Wang
- Sino-French Cooperative Central Lab, Gongli Hospital of Shanghai Pudong New Area, No. 207, Juye Rd., Pudong New District, Shanghai 200135, China
| | - Yuting Song
- Sino-French Cooperative Central Lab, Gongli Hospital of Shanghai Pudong New Area, No. 207, Juye Rd., Pudong New District, Shanghai 200135, China
- Ningxia Medical University, Ningxia 750000, China
| | - Fengxia Xu
- Department of Clinical Laboratory, Gongli Hospital of Shanghai Pudong New Area, 207 Juye Road, Pudong New Area, Shanghai 200135, China
| | - Qiuhua Xie
- Department of Clinical Laboratory, Gongli Hospital of Shanghai Pudong New Area, 207 Juye Road, Pudong New Area, Shanghai 200135, China
| | - Mei Jiang
- Department of Neurology, Gongli Hospital of Shanghai Pudong New Area, Shanghai 200135, China
| | - Xinghui Liu
- Department of Clinical Laboratory, Gongli Hospital of Shanghai Pudong New Area, 207 Juye Road, Pudong New Area, Shanghai 200135, China
| | - Denghai Zhang
- Sino-French Cooperative Central Lab, Gongli Hospital of Shanghai Pudong New Area, No. 207, Juye Rd., Pudong New District, Shanghai 200135, China
- Ningxia Medical University, Ningxia 750000, China
| | - Limin Xu
- Department of Clinical Laboratory, Gongli Hospital of Shanghai Pudong New Area, 207 Juye Road, Pudong New Area, Shanghai 200135, China
| |
Collapse
|
5
|
Muneer PMA, Saikia BB, Bhowmick S. Synergistic effect of mild traumatic brain injury and alcohol aggravates neuroinflammation, amyloidogenesis, tau pathology, neurodegeneration, and blood-brain barrier alterations: Impact on psychological stress. Exp Neurol 2022; 358:114222. [PMID: 36089059 DOI: 10.1016/j.expneurol.2022.114222] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/12/2022] [Accepted: 09/03/2022] [Indexed: 11/24/2022]
Abstract
After a mild traumatic brain injury (mTBI), victims often experience emotional/psychological stress such as heightened irritability, anxiety, apathy, and depression. Severe mental health complications are common in military populations following a combat-acquired TBI and intensified unhealthy alcohol use. The high prevalence of alcohol abuse among TBI victims underscores how alcohol abuse exacerbates emotional/psychological symptoms such as depression and anxiety. The experimental mTBI was induced in vivo by fluid percussion injury (15 psi) in mice and ethanol diet feeding continued for 28 days. We analyzed different biomarkers of the biochemical mechanisms and pathophysiology of neurological damage, and functional outcome of psychological stress by sucrose preference, and light-dark tests. We demonstrated that the synergistic effect of TBI and alcohol leads to psychological stress such as depression and anxiety. The studies showed that oxidative stress, amyloidogenesis, tau pathology, neuroinflammation, and neurodegeneration markers were elevated, and glial activation and blood-brain barrier (BBB) damage were exacerbated during the synergistic effect of TBI and alcohol. Further, we studied the biochemical mechanisms of psychological stress that showed the significant reduction of 5-HT1AR, neuropeptide-Y, and norepinephrine, and an increase in monoamine oxidase-a in the combined effect of TBI and alcohol. This work suggested that the combined TBI and alcohol-induced effect leads to depression and anxiety, via sequential biochemical changes that cause neuroinflammation, amyloidogenesis, tau pathology, neurodegeneration, and BBB alterations. This clinically relevant study will contribute to developing a comprehensive therapeutic approach for patients suffering from TBI and alcohol-mediated neurological damage and psychological stress.
Collapse
Affiliation(s)
- P M Abdul Muneer
- Laboratory of CNS injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK University Medical Center, 65 James St, Edison, NJ 08820, United States; Department of Neurology, Hackensack Meridian School of Medicine, Nutley, NJ 07110, USA.
| | - Bibhuti Ballav Saikia
- Laboratory of CNS injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK University Medical Center, 65 James St, Edison, NJ 08820, United States
| | - Saurav Bhowmick
- Laboratory of CNS injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK University Medical Center, 65 James St, Edison, NJ 08820, United States
| |
Collapse
|
6
|
Zhu J, Chen H, Guo J, Zha C, Lu D. Sodium Tanshinone IIA Sulfonate Inhibits Vascular Endothelial Cell Pyroptosis via the AMPK Signaling Pathway in Atherosclerosis. J Inflamm Res 2022; 15:6293-6306. [PMID: 36408328 PMCID: PMC9673812 DOI: 10.2147/jir.s386470] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/03/2022] [Indexed: 11/15/2022] Open
Abstract
Introduction Atherosclerosis (AS) is the underlying cause of cardiovascular events. Endothelial cell mitochondrial damage and pyroptosis are important factors contributing to AS. Changes in internal mitochondrial conformation and increase in reactive oxygen species (ROS) lead to the disruption of mitochondrial energy metabolism, activation of the NLRP3 inflammasome and pyroptosis, which in turn affect atherogenesis by impairing endothelial function. AMPK is a core player in the regulation of cellular metabolism, not only by regulating mitochondrial homeostasis but also by regulating cellular inflammatory responses. Sodium tanshinone IIA sulfonate (STS), a water-soluble derivative of tanshinone IIA, has significant antioxidant and anti-inflammatory effects, and roles in cardiovascular protection. Purpose In this study, we investigated whether STS plays a protective role in AS by regulating endothelial cell mitochondrial function and pyroptosis through an AMPK-dependent mitochondrial pathway. Methods and Results Male ApoE−/− mice and HUVECs were used for the experiments. We found that STS treatment largely abrogated the upregulation of key proteins in aortic vessel wall plaques and typical pyroptosis signaling in ApoE−/− mice fed a western diet, consequently enhancing pAMPK expression, plaque stabilization, and anti-inflammatory responses. Consistently, STS pretreatment inhibited cholesterol crystallization (CC) -induced cell pyroptosis and activated pAMPK expression. In vitro, using HUVECs, we further found that STS treatment ameliorated mitochondrial ROS caused by CC, as evidenced by the finding that STS inhibited mitochondrial damage caused by CC. The improvement of endothelial cell mitochondrial function by STS is blocked by dorsomorphin (AMPK inhibitor). Consistently, the blockade of endothelial cell pyroptosis by STS is disrupted by dorsomorphin. Conclusion Our results suggest that STS enhances maintenance of mitochondrial homeostasis and inhibits mitochondrial ROS overproduction via AMPK, thereby improving endothelial cell pyroptosis during AS.
Collapse
Affiliation(s)
- Ji Zhu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou, People’s Republic of China
| | - Hang Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Jianan Guo
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Chen Zha
- The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou, People’s Republic of China
| | - Dezhao Lu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Correspondence: Dezhao Lu, Email
| |
Collapse
|
7
|
Sang W, Chen S, Lin L, Wang N, Kong X, Ye J. Antioxidant mitoquinone ameliorates EtOH-LPS induced lung injury by inhibiting mitophagy and NLRP3 inflammasome activation. Front Immunol 2022; 13:973108. [PMID: 36059543 PMCID: PMC9436256 DOI: 10.3389/fimmu.2022.973108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 07/29/2022] [Indexed: 12/02/2022] Open
Abstract
Chronic ethanol abuse is a systemic disorder and a risk factor for acute respiratory distress syndrome (ARDS) and chronic obstructive pulmonary disease (COPD). However, the mechanisms involved are unknown. One explanation is that ethanol produces damaging reactive oxygen species (ROS) and disturbs the balance of mitochondria within the lungs to promote a pro-injury environment. We hypothesized that targeting an antioxidant to the mitochondria would prevent oxidative damage and attenuate EtOH-LPS-induced lung injury. To test this, we investigated the effects of mitochondria-targeted ubiquinone, Mitoquinone (MitoQ) on ethanol-sensitized lung injury induced by LPS. Lung inflammation, ROS, mitochondria function, and mitophagy were assessed. We demonstrated that chronic ethanol feeding sensitized the lung to LPS-induced lung injury with significantly increased reactive oxygen species ROS level and mitochondrial injury as well as lung cellular NLRP3 inflammasome activation. These deleterious effects were attenuated by MitoQ administration in mice. The protective effects of MitoQ are associated with decreased cellular mitophagy and NLRP3 inflammasome activation in vivo and in vitro. Taken together, our results demonstrated that ethanol aggravated LPS-induced lung injury, and antioxidant MitoQ protects from EtOH-LPS-induced lung injury, probably through reducing mitophagy and protecting mitochondria, followed by NLRP3 inflammasome activation. These results will provide the prevention and treatment of ethanol intake effects with new ideas.
Collapse
Affiliation(s)
- Wenhua Sang
- School of Basic Medical Sciences, Institute of Hypoxia Research, Cixi Biomedical Institute, Wenzhou Medical University, Wenzhou, China
- School of Basic Medical Sciences, Zhejiang University, Hangzhou, China
| | - Sha Chen
- School of Basic Medical Sciences, Institute of Hypoxia Research, Cixi Biomedical Institute, Wenzhou Medical University, Wenzhou, China
| | - Lidan Lin
- School of Basic Medical Sciences, Institute of Hypoxia Research, Cixi Biomedical Institute, Wenzhou Medical University, Wenzhou, China
| | - Nan Wang
- School of Basic Medical Sciences, Institute of Hypoxia Research, Cixi Biomedical Institute, Wenzhou Medical University, Wenzhou, China
| | - Xiaoxia Kong
- School of Basic Medical Sciences, Institute of Hypoxia Research, Cixi Biomedical Institute, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Xiaoxia Kong, ; Jinyan Ye,
| | - Jinyan Ye
- Department of Respiratory Medicine and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Xiaoxia Kong, ; Jinyan Ye,
| |
Collapse
|
8
|
Bhowmick S, Alikunju S, Muneer PMA. NADPH oxidase-induced activation of transforming growth factor-beta-1 causes neuropathy by suppressing antioxidant signaling pathways in alcohol use disorder. Neuropharmacology 2022; 213:109136. [PMID: 35584723 DOI: 10.1016/j.neuropharm.2022.109136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 04/20/2022] [Accepted: 05/10/2022] [Indexed: 11/22/2022]
Abstract
Oxidative signaling and inflammatory cascades are the central mechanism in alcohol-induced brain injury, which result in glial activation, neuronal and myelin loss, neuronal apoptosis, and ultimately long-term neurological deficits. While transforming growth factor-beta1 (TGF-β1) has a significant role in inflammation and apoptosis in myriads of other pathophysiological conditions, the precise function of increased TGF-β1 in alcohol use disorder (AUD)-induced brain damage is unknown. In this study, our objective is to study ethanol-induced activation of TGF-β1 and associated mechanisms of neuroinflammation and apoptosis. Using a mouse model feeding with ethanol diet and an in vitro model in mouse cortical neuronal cultures, we explored the significance of TGF-β1 activation in the pathophysiology of AUD. Our study demonstrated that the activation of TGF-β1 in ethanol ingestion correlated with the induction of free radical generating enzyme NADPH oxidase (NOX). Further, using TGF-β type I receptor (TGF-βRI) inhibitor SB431542 and TGF-β antagonist Smad7, we established that the alcohol-induced activation of TGF-β1 impairs antioxidant signaling pathways and leads to neuroinflammation and apoptosis. Blocking of TGF-βRI or inhibition of TGF-β1 diminished TGF-β1-induced inflammation and apoptosis. Further, TGF-β1 activation increased the phosphorylation of R-Smads including Smad2 and Smad3 proteins. Using various biochemical analyses and genetic approaches, we demonstrated the up-regulation of pro-inflammatory cytokines IL-1β and TNF-α and apoptotic cell death in neurons. In conclusion, this study significantly extends our understanding of the pathophysiology of AUD and provides a unique insight for developing various therapeutic interventions by activating antioxidant signaling pathways for the treatment of AUD-induced neurological complications.
Collapse
Affiliation(s)
- Saurav Bhowmick
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK University Medical Center, 65 James St, Edison, NJ, 08820, United States
| | - Saleena Alikunju
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Ave, Newark, NJ, 07103, United States
| | - P M Abdul Muneer
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK University Medical Center, 65 James St, Edison, NJ, 08820, United States; Department of Neurology, Hackensack Meridian School of Medicine, Nutley, NJ, 07110, USA.
| |
Collapse
|
9
|
Alcohol-Induced Oxidative Stress and the Role of Antioxidants in Alcohol Use Disorder: A Systematic Review. Antioxidants (Basel) 2022; 11:antiox11071374. [PMID: 35883865 PMCID: PMC9311529 DOI: 10.3390/antiox11071374] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 12/12/2022] Open
Abstract
Alcohol use disorder (AUD) is a highly prevalent, comorbid, and disabling disorder. The underlying mechanism of ethanol neurotoxicity and the involvement of oxidative stress is still not fully elucidated. However, ethanol metabolism has been associated with increased oxidative stress through alcohol dehydrogenase, the microsomal ethanol oxidation system, and catalase metabolic pathways. We searched the PubMed and genome-wide association studies (GWAS) catalog databases to review the literature systematically and summarized the findings focusing on AUD and alcohol abstinence in relation to oxidative stress. In addition, we reviewed the ClinicalTrials.gov resource of the US National Library of Medicine to identify all ongoing and completed clinical trials that include therapeutic interventions based on antioxidants. The retrieved clinical and preclinical studies show that oxidative stress impacts AUD through genetics, alcohol metabolism, inflammation, and neurodegeneration.
Collapse
|
10
|
Ilješ AP, Plesničar BK, Dolžan V. Associations of NLRP3 and CARD8 gene polymorphisms with alcohol dependence and commonly related psychiatric disorders: a preliminary study. Arh Hig Rada Toksikol 2021; 72:191-197. [PMID: 34587665 PMCID: PMC8576752 DOI: 10.2478/aiht-2021-72-3432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/01/2020] [Accepted: 09/01/2021] [Indexed: 12/11/2022] Open
Abstract
We investigated two functional polymorphisms in NLRP3 inflammasome genes (NLRP3 rs35829419 and CARD8 rs2043211) and their association with alcohol dependence and related anxiety, depression, obsession-compulsion, or aggression in 88 hospitalised alcohol-dependent patients, 99 abstinent alcohol-dependent participants, and 94 controls, all male Caucasian. Alcohol dependence-related psychiatric disorders were assessed with the Zung Depression and Anxiety scale, Buss-Durkee Hostility Inventory, Alcohol Use Disorders Identification Test, Brief Social Phobia Scale, Obsessive Compulsive Drinking Scale, and Yale-Brown Obsessive-Compulsive Scale. For genotyping we used the allele-specific quantitative polymerase chain reaction-based methods. The three groups differed significantly in CARD8 rs2043211 distribution (P=0.049; chi-squared=9.557; df=4). The NLPR3 rs35829419 polymorphism was not significantly associated with alcohol dependence. In hospitalised alcohol-dependent patients the investigated polymorphisms were not associated with scores indicating alcohol consumption or comorbid symptoms. In abstinent alcohol-dependent subjects homozygotes for the polymorphic CARD8 allele presented with the highest scores on the Zung Anxiety Scale (p=0.048; df=2; F=3.140). Among controls, CARD8 genotype was associated with high scores on the Obsessive Compulsive Drinking Scale (P=0.027; df=2; F=3.744). In conclusion, our results reveal that CARD8 rs2043211 may play some role in susceptibility to alcohol dependence, expression of anxiety symptoms in abstinent alcohol-dependent subjects, and in obsessive compulsive drinking in healthy controls. However, further studies with larger cohorts are required to confirm these preliminary findings.
Collapse
Affiliation(s)
| | - Blanka Kores Plesničar
- University Psychiatric Clinic, Ljubljana, Slovenia
- University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Vita Dolžan
- University of Ljubljana, Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, Pharmacogenetics Laboratory, Ljubljana, Slovenia
| |
Collapse
|
11
|
Bhowmick S, Malat A, Caruso D, Ponery N, D'Mello V, Finn C, Muneer PMA. Intercellular Adhesion Molecule-1-Induced Posttraumatic Brain Injury Neuropathology in the Prefrontal Cortex and Hippocampus Leads to Sensorimotor Function Deficits and Psychological Stress. eNeuro 2021; 8:ENEURO.0242-21.2021. [PMID: 34135004 PMCID: PMC8287878 DOI: 10.1523/eneuro.0242-21.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 06/04/2021] [Indexed: 02/07/2023] Open
Abstract
Intercellular adhesion molecule-1 (ICAM-1) promotes adhesion and transmigration of circulating leukocytes across the blood-brain barrier (BBB). Traumatic brain injury (TBI) causes transmigrated immunocompetent cells to release mediators [function-associated antigen (LFA)-1 and macrophage-1 antigen (Mac-1)] that stimulate glial and endothelial cells to express ICAM-1 and release cytokines, sustaining neuroinflammation and neurodegeneration. Although a strong correlation exists between TBI-mediated inflammation and impairment in functional outcome following brain trauma, the role of ICAM-1 in impairing functional outcome by inducing neuroinflammation and neurodegeneration after TBI remains inconclusive. The experimental TBI was induced in vivo by fluid percussion injury (FPI; 10 and 20 psi) in wild-type (WT) and ICAM-1-/- mice and in vitro by stretch injury (3 psi) in brain endothelial cells. We manipulate ICAM-1 pharmacologically and genetically and conducted several biochemical analyses to gain insight into the mechanisms underlying ICAM-1-mediated neuroinflammation and performed rotarod, grid-walk, sucrose preference, and light-dark tests to assess functional outcome. TBI-induced ICAM-1-mediated neuroinflammation and cell death occur via LFA-1 or Mac-1 signaling pathways that rely on oxidative stress, matrix metalloproteinase (MMP), and vascular endothelial growth factor (VEGF) pathways. The deletion or blocking of ICAM-1 resulted in a better outcome in attenuating neuroinflammation and cell death as marked by the markers such as NF-kB, IL-1β, TNF-α, cleaved-caspase-3 (cl-caspase-3), Annexin V, and by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and Trypan blue staining. ICAM-1 deletion in TBI improves sensorimotor, depression, and anxiety-like behavior with significant upregulation of norepinephrine (NE), dopamine (DA) D1 receptor (DAD1R), serotonin (5-HT)1AR, and neuropeptide Y (NPY). This study could establish the significance of ICAM-1 as a novel therapeutic target against the pathophysiology to establish functional recovery after TBI.
Collapse
Affiliation(s)
- Saurav Bhowmick
- Laboratory of CNS injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK University Medical Center, Edison, NJ 08820
| | - Anitha Malat
- Laboratory of CNS injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK University Medical Center, Edison, NJ 08820
| | - Danielle Caruso
- Laboratory of CNS injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK University Medical Center, Edison, NJ 08820
| | - Nizmi Ponery
- Laboratory of CNS injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK University Medical Center, Edison, NJ 08820
| | - Veera D'Mello
- Laboratory of CNS injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK University Medical Center, Edison, NJ 08820
| | - Christina Finn
- Laboratory of CNS injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK University Medical Center, Edison, NJ 08820
| | - P M Abdul Muneer
- Laboratory of CNS injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK University Medical Center, Edison, NJ 08820
- Department of Neurology, Hackensack Meridian School of Medicine, Nutley, NJ 07110
| |
Collapse
|
12
|
Bai R, Lang Y, Shao J, Deng Y, Refuhati R, Cui L. The Role of NLRP3 Inflammasome in Cerebrovascular Diseases Pathology and Possible Therapeutic Targets. ASN Neuro 2021; 13:17590914211018100. [PMID: 34053242 PMCID: PMC8168029 DOI: 10.1177/17590914211018100] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cerebrovascular diseases are pathological conditions involving impaired blood flow in the brain, primarily including ischaemic stroke, intracranial haemorrhage, and subarachnoid haemorrhage. The nucleotide-binding and oligomerisation (NOD) domain-like receptor (NLR) family pyrin domain (PYD)-containing 3 (NLRP3) inflammasome is a protein complex and a vital component of the immune system. Emerging evidence has indicated that the NLRP3 inflammasome plays an important role in cerebrovascular diseases. The function of the NLRP3 inflammasome in the pathogenesis of cerebrovascular diseases remains an interesting field of research. In this review, we first summarised the pathological mechanism of cerebrovascular diseases and the pathological mechanism of the NLRP3 inflammasome in aggravating atherosclerosis and cerebrovascular diseases. Second, we outlined signalling pathways through which the NLRP3 inflammasome participates in aggravating or mitigating cerebrovascular diseases. Reactive oxygen species (ROS)/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), ROS/thioredoxin-interacting protein (TXNIP) and purinergic receptor-7 (P2X7R) signalling pathways can activate the NLRP3 inflammasome; activation of the NLRP3 inflammasome can aggravate cerebrovascular diseases by mediating apoptosis and pyroptosis. Autophagy/mitochondrial autophagy, nuclear factor E2-related factor-2 (Nrf2), interferon (IFN)-β, sirtuin (SIRT), and phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) reportedly alleviate cerebrovascular diseases by inhibiting NLRP3 inflammasome activation. Finally, we explored specific inhibitors of the NLRP3 inflammasome based on the two-step activation of the NLRP3 inflammasome, which can be developed as new drugs to treat cerebrovascular diseases.
Collapse
Affiliation(s)
- Rongrong Bai
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yue Lang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jie Shao
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yu Deng
- Department of Hepatopancreatobiliary Surgery, The First Hospital of Jilin University, Changchun, China
| | - Reyisha Refuhati
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Li Cui
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Li G, Dong Y, Liu D, Zou Z, Hao G, Gao X, Pan P, Liang G. NEK7 Coordinates Rapid Neuroinflammation After Subarachnoid Hemorrhage in Mice. Front Neurol 2020; 11:551. [PMID: 32733353 PMCID: PMC7360676 DOI: 10.3389/fneur.2020.00551] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 05/15/2020] [Indexed: 01/02/2023] Open
Abstract
Background: Subarachnoid hemorrhage (SAH) is a devastating disease which leads to high morbidity and mortality. Recent studies have indicated that, never in mitosis gene A-related expressed kinase 7 (NEK7), is involved in NLRP3 (NLR family, pyrin domain containing 3) associated inflammation, which may result in subsequent cellular and vascular damage. The aim of this study was to investigate whether NEK7 is involved in the pathophysiology of subarachnoid hemorrhage. Methods: 455 adult male C57B6J mice, weighing 22 to 30 g, were used to investigate the time course of NEK7 expression in the ipsilateral cortex after SAH, and to investigate the intrinsic function and mechanism of NEK7. A vascular puncture model was used to create the mouse SAH model, and intracerebroventricular injection was used to deliver NEK7 recombinant protein, NEK7 small interfering RNA, nigericin, and MCC950. Neurological score, brain water content, Evans blue extravasation, immunofluorescence, and western blot were evaluated for neurological outcome, neuronal apoptosis, blood-brain barrier damage, microglia accumulation, and the mechanism of NEK7 and NLRP3 activation. Results: Our results exhibited that intrinsic NEK7 was elevated after SAH in the cortex of the left/ipsilateral hemisphere and was colocalized with microglia, endothelial cells, neuron, astrocyte, and oligodendrocyte, and highly expressed in microglia and endothelial cells after SAH. NEK7 recombinant protein aggravated neurological deficits, brain edema, neuronal apoptosis, BBB permeability, microglial accumulation, and activated caspase-1 and IL-1β maturation, while NEK7 small interfering RNA injection reversed those effects. Nigericin administration enhanced ASC oligomerization, caspase-1 and IL-1β maturation without increasing the protein level of NLRP3, and ASC oligomerization and caspase-1 IL-1β maturation reduced when combined with NEK7 knockdown or MCC950 delivery. We found the level of NEK7 expression increased after SAH and could activate the downstream NLRP3 pathway to induce caspase-1, IL-1β expression and then increased the BBB opening, microglia accumulation and neuronal apoptosis after SAH. Conclusions: This study demonstrated for the first time that NEK7 mediated the harmful effects of neuronal apoptosis and BBB disruption after SAH, which may potentially be mediated by the NEK7/NLRP3 signal. NEK7 served as a co-component for NLRP3 inflammasome activation after SAH. NEK7 may be a promising target on the management of SAH patients.
Collapse
Affiliation(s)
- Gen Li
- Department of Neurosurgery, General Hospital of Northern Theater Command (General Hospital of Shenyang Military Command), Shenyang, China.,Dalian Medical University, Dalian, China
| | - Yushu Dong
- Department of Neurosurgery, General Hospital of Northern Theater Command (General Hospital of Shenyang Military Command), Shenyang, China
| | - Dongdong Liu
- Department of Neurosurgery, General Hospital of Northern Theater Command (General Hospital of Shenyang Military Command), Shenyang, China.,Dalian Medical University, Dalian, China
| | - Zheng Zou
- Department of Neurosurgery, General Hospital of Northern Theater Command (General Hospital of Shenyang Military Command), Shenyang, China
| | - Guangzhi Hao
- Department of Neurosurgery, General Hospital of Northern Theater Command (General Hospital of Shenyang Military Command), Shenyang, China
| | - Xu Gao
- Department of Neurosurgery, General Hospital of Northern Theater Command (General Hospital of Shenyang Military Command), Shenyang, China
| | - Pengyu Pan
- Department of Neurosurgery, General Hospital of Northern Theater Command (General Hospital of Shenyang Military Command), Shenyang, China
| | - Guobiao Liang
- Department of Neurosurgery, General Hospital of Northern Theater Command (General Hospital of Shenyang Military Command), Shenyang, China
| |
Collapse
|
14
|
Bhowmick S, D'Mello V, Caruso D, Abdul-Muneer PM. Traumatic brain injury-induced downregulation of Nrf2 activates inflammatory response and apoptotic cell death. J Mol Med (Berl) 2019; 97:1627-1641. [PMID: 31758217 DOI: 10.1007/s00109-019-01851-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/11/2019] [Accepted: 11/05/2019] [Indexed: 12/17/2022]
Abstract
Recent studies from our group and others have demonstrated that oxidative stress, Ca2+ signaling, and neuroinflammation are major mechanisms contributing to post-traumatic neurodegeneration. The present study investigated the mechanisms of regulation of nuclear factor E2-related factor 2 (Nrf2) and its role in regulating antioxidant genes and oxidative stress-induced neuroinflammation and neurodegeneration following TBI. Nrf2 transcriptional system is the major regulator of endogenous defense mechanisms operating within the cells. Wild-type (Nrf2+/+) and Nrf2-deficient mice (Nrf2-/-) were subjected to 15 psi fluid percussion injury and demonstrated the regulatory role of Nrf2 in the expression antioxidant genes and oxidative stress, neuroinflammation, and cell death. Immunohistochemistry, q-RT-PCR, and western blotting techniques detected downregulation of Nrf2 and antioxidant proteins such as HO-1, GPx1, GSTm1, and NQO1 in mouse brain samples. Further, our study demonstrated that the downregulation of Nrf2 and antioxidant genes in TBI correlated with the induction of free radical-generating enzyme NADPH oxidase 1 and inducible nitric oxide synthase and their corresponding oxidative/nitrosative stress markers 4-hydroxynonenal and 3-nitrotyrosine. The decrease in Nrf2 with subsequent increase in oxidative stress markers led to the activation of MMP3/9, TGF-β1, and NF-kB that further led to neuroinflammation and apoptosis. The absence of Nrf2 function in mice resulted in exacerbated brain injury as shown by the increased oxidative stress markers, pro-inflammatory cytokines, and apoptosis markers at 24 h after TBI. In conclusion, this study could establish the significance of Nrf2 in transforming into a novel preventive approach against the pathophysiology of TBI. KEY MESSAGES: • Traumatic brain injury impairs Nrf2 signaling in mouse. • Nrf2-mediated activation of antioxidant genes are altered after TBI. • Impairment of Nrf2 signaling leads to oxidative stress. • TBI-induced downregulation of Nrf2 activates MMPs, TGF-β1, and NF-kB. • Nrf2 regulates neuroinflammation and apoptotic cell death in TB.
Collapse
Affiliation(s)
- Saurav Bhowmick
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St, Edison, NJ, 08820, USA
| | - Veera D'Mello
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St, Edison, NJ, 08820, USA
| | - Danielle Caruso
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St, Edison, NJ, 08820, USA
| | - P M Abdul-Muneer
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St, Edison, NJ, 08820, USA.
| |
Collapse
|
15
|
Gaydos J, McNally A, Burnham EL. The impact of alcohol use disorders on pulmonary immune cell inflammatory responses to Streptococcus pneumoniae. Alcohol 2019; 80:119-130. [PMID: 30195043 DOI: 10.1016/j.alcohol.2018.08.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/29/2018] [Accepted: 08/31/2018] [Indexed: 12/11/2022]
Abstract
Community-acquired pneumonia due to Streptococcus pneumoniae occurs commonly in alcohol use disorders (AUDs). Pneumonia in the AUD patient is associated with poorer outcomes, and specific therapies to mitigate disease severity in these patients do not exist. Numerous investigations have attributed increased severity of pneumonia in AUDs to aberrant function of the alveolar macrophage (AM), a lung immune cell critical in host defense initiation. No studies have examined the response of human AMs to S. pneumoniae in AUDs. We hypothesized that the inflammatory mediators released by AMs after S. pneumoniae stimulation would differ quantitatively in individuals with AUDs compared to non-AUD participants. We further postulated that AM inflammatory mediators would be diminished after exposure to the antioxidant, N-acetylcysteine (NAC). For comparison, responses of peripheral blood mononuclear cells (PBMCs) to pneumococcal protein were also examined. Otherwise healthy participants with AUDs and smoking-matched controls underwent bronchoalveolar lavage and peripheral blood sampling to obtain AMs and PBMCs, respectively. Freshly collected cells were cultured with increasing doses of heat-killed S. pneumoniae protein, with and without exposure to N-acetylcysteine. Cell culture supernatants were collected, and inflammatory mediators were measured, including interferon (IFN)-γ, interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α. IFN-γ and IL-6 were significantly higher in unstimulated AM cell culture supernatants from subjects with AUDs. After stimulation with pneumococcal protein, a dose-response and time-dependent increase in pro-inflammatory cytokine production by both AMs and PBMCs was also observed; differences were not observed between AUD and non-AUD subjects. Addition of NAC to pneumococcal-stimulated AMs and PBMCs was generally associated with diminished cytokine production, with the exception of IL-1β that was elevated in AM culture supernatants from subjects with AUDs. Our observations suggest that AUDs contribute to basal alterations in AM pro-inflammatory cytokine elaboration, but did not support consistent differences in pneumococcal-stimulated AM or PBMC inflammatory mediator secretion that were referable to AUDs.
Collapse
|
16
|
Loftis JM, Taylor J, Hudson R, Firsick EJ. Neuroinvasion and cognitive impairment in comorbid alcohol dependence and chronic viral infection: An initial investigation. J Neuroimmunol 2019; 335:577006. [PMID: 31325774 DOI: 10.1016/j.jneuroim.2019.577006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 12/16/2022]
Abstract
Viruses that invade the central nervous system (CNS) can cause neuropsychiatric impairments. Similarly, chronic alcohol exposure can induce inflammatory responses that alter brain function. However, the effects of a chronic viral infection and comorbid alcohol use on neuroinflammation and behavior are not well-defined. We investigated the role of heavy alcohol intake in regulating inflammatory responses and behavioral signs of cognitive impairments in mice infected with lymphocytic choriomeningitis virus (LCMV) clone 13. LCMV-infected mice exposed to alcohol had increased peripheral inflammation and impaired cognitive function (as indicated by performance on the novel object recognition test). Initial findings suggest that brain region-specific dysregulation of microglial response to viral infection may contribute to cognitive impairments in the context of heavy alcohol use.
Collapse
Affiliation(s)
- Jennifer M Loftis
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA; Methamphetamine Abuse Research Center, Veterans Affairs Portland Health Care System, Oregon Health & Science University, Portland, OR, USA.
| | - Jonathan Taylor
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA
| | - Rebekah Hudson
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA; Department of Public Health and Preventive Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Evan J Firsick
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA
| |
Collapse
|
17
|
ER Stress Activates the NLRP3 Inflammasome: A Novel Mechanism of Atherosclerosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3462530. [PMID: 31687078 PMCID: PMC6800950 DOI: 10.1155/2019/3462530] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/21/2019] [Accepted: 08/31/2019] [Indexed: 02/06/2023]
Abstract
The endoplasmic reticulum (ER) is an important organelle that regulates several fundamental cellular processes, and ER dysfunction has implications for many intracellular events. The nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome is an intracellularly produced macromolecular complex that can trigger pyroptosis and inflammation, and its activation is induced by a variety of signals. ER stress has been found to affect NLRP3 inflammasome activation through multiple effects including the unfolded protein response (UPR), calcium or lipid metabolism, and reactive oxygen species (ROS) generation. Intriguingly, the role of ER stress in inflammasome activation has not attracted a great deal of attention. In addition, increasing evidence highlights that both ER stress and NLRP3 inflammasome activation contribute to atherosclerosis (AS). AS is a common cardiovascular disease with complex pathogenesis, and the precise mechanisms behind its pathogenesis remain to be determined. Both ER stress and the NLRP3 inflammasome have emerged as critical individual contributors of AS, and owing to the multiple associations between these two events, we speculate that they contribute to the mechanisms of pathogenesis in AS. In this review, we aim to summarize the molecular mechanisms of ER stress, NLRP3 inflammasome activation, and the cross talk between these two pathways in AS in the hopes of providing new pharmacological targets for AS treatment.
Collapse
|
18
|
Zhang LG, Chen J, Meng JL, Zhang Y, Liu Y, Zhan CS, Chen XG, Zhang L, Liang CZ. Effect of alcohol on chronic pelvic pain and prostatic inflammation in a mouse model of experimental autoimmune prostatitis. Prostate 2019; 79:1439-1449. [PMID: 31233226 DOI: 10.1002/pros.23866] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/28/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) is a prevalent disease of the urogenital system. Alcohol has been reported to be closely related to CP/CPPS. Thus, we intended to verify the role of alcohol in CP/CPPS and determine the underlying mechanism. METHODS We induced experimental autoimmune prostatitis (EAP) mouse model by intradermally injecting a mixture of prostate antigens (PAgs) and complete Freund's adjuvant on days 0 and 28. Mice were treated with alcohol (control-alcohol and EAP-alcohol groups) or vehicle (control-vehicle, and EAP-vehicle groups) from day 32 to 42. Forty-two days after PAg injection, the pathological appearance of the prostate tissues was evaluated, and histological analyses of the prostate were performed. Chronic pelvic pain was assessed by applying von Frey filaments to the lower abdomen. Proinflammatory cytokines were detected by enzyme-linked immunosorbent assay tests. Then, we explored the effects of the NLRP3 inhibitor MCC950 on chronic pelvic pain and prostatic inflammation in this model. RESULTS Histological analyses showed diffuse inflammation in the stromal tissues that were characterized by severe infiltration of neutrophils and mononuclear cells in mice in the EAP-alcohol group compared with EAP-vehicle group. Chronic pain tests showed that the response frequency was significantly increased using a von Frey filament at forces of 0.4, 1.0, and 4.0 g in EAP-alcohol group compared with EAP-vehicle (P < .05). The levels of proinflammatory cytokines, including interferon (IFN)-γ, tumor necrosis factor (TNF)-α, IL-17, and IL-1β were all significantly elevated in EAP-alcohol group compared with the EAP-vehicle group (P < .05). However, between the control-alcohol and control-vehicle groups, chronic pain tests, histological assays, and cytokine determinations showed no differences. Furthermore, our results demonstrated that MCC950 could decrease the expression level of NLRP3 inflammasome-related proteins including NLRP3, ASC, and caspase-1. The chronic pain tests, histological assays, and cytokine determinations showed that MCC950 could attenuate the chronic pain and prostatic inflammation through the inhibition of the NLRP3 inflammasome. CONCLUSIONS This study indicated that alcohol could aggravate the severity of prostatic inflammation in EAP model though activating the NLRP3 inflammasome. Furthermore, the role of MCC950 in inhibiting NLRP3 inflammasome and decreasing IL-1β secretion to alleviate EAP severity may show that it is a promising therapeutic agent for CP/CPPS.
Collapse
Affiliation(s)
- Li-Gang Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Jing Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Jia-Lin Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Yong Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Yi Liu
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, P.R. China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Chang-Sheng Zhan
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, P.R. China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Xian-Guo Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, P.R. China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Li Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, P.R. China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Chao-Zhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, P.R. China
- Institute of Urology, Anhui Medical University, Hefei, Anhui, P.R. China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
19
|
Bhowmick S, D'Mello V, Caruso D, Wallerstein A, Muneer PMA. Impairment of pericyte-endothelium crosstalk leads to blood-brain barrier dysfunction following traumatic brain injury. Exp Neurol 2019; 317:260-270. [PMID: 30926390 DOI: 10.1016/j.expneurol.2019.03.014] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/25/2019] [Accepted: 03/25/2019] [Indexed: 01/17/2023]
Abstract
The blood-brain barrier (BBB) constitutes a neurovascular unit formed by microvascular endothelial cells, pericytes, and astrocytes. Brain pericytes are important regulators of BBB integrity, permeability, and blood flow. Pericyte loss has been implicated in injury; however, how the crosstalk among pericytes, endothelial cells, and astrocytes ultimately leads to BBB dysfunction in traumatic brain injury (TBI) remains elusive. In this study, we demonstrate the importance of pericyte-endothelium interaction in maintaining the BBB function. TBI causes the platelet-derived growth factor-B (PDGF-B)/PDGF receptor-β signaling impairment that results in loss of interaction with endothelium and leads to neurovascular dysfunction. Using in vivo mild (7 psi) and moderate (15 psi) fluid percussion injury (FPI) in mice, we demonstrate the expression of various pericyte markers including PDGFR-β, NG2 and CD13 that were significantly reduced with a subsequent reduction in the expression of various integrins; adherent junction protein, N-cadherin; gap junction protein, connexin-43; and tight junction proteins such as occludin, claudin-5, ZO-1, and JAM-a. Impairment of pericyte-endothelium interaction increases the BBB permeability to water that is marked by a significant increase in aquaporin4 expression in injured animals. Similarly, pericyte-endothelium integrity impairment in FPI animals greatly increases the permeability of small-molecular-weight sodium fluorescein and high-molecular-weight-tracer Evans blue across the BBB. In addition, the injury-inflicted animals show significantly higher levels of S100β and NSE in the blood samples compared with controls. In conclusion, our data provide an insight that brain trauma causes an early impairment of pericyte-endothelium integrity and results in BBB dysregulation that initiates pathological consequences associated with TBI.
Collapse
Affiliation(s)
- Saurav Bhowmick
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St, Edison, New Jersey 08820, United States
| | - Veera D'Mello
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St, Edison, New Jersey 08820, United States
| | - Danielle Caruso
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St, Edison, New Jersey 08820, United States
| | - Alex Wallerstein
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St, Edison, New Jersey 08820, United States
| | - P M Abdul Muneer
- Laboratory of CNS Injury and Molecular Therapy, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St, Edison, New Jersey 08820, United States.
| |
Collapse
|
20
|
Qi S, Wang Q, Xie B, Chen Y, Zhang Z, Xu Y. P38 MAPK signaling pathway mediates COM crystal-induced crystal adhesion change in rat renal tubular epithelial cells. Urolithiasis 2019; 48:9-18. [PMID: 31183507 PMCID: PMC6989645 DOI: 10.1007/s00240-019-01143-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 06/03/2019] [Indexed: 02/07/2023]
Abstract
The objective of the study is to clarify the mechanism of p38 mitogen-activated protein kinase (p38 MAPK) signaling pathway in the change of crystal adhesion in rat renal tubular epithelial cells (NRK-52E) induced by calcium oxalate monohydrate (COM) crystals. NRK-52E cells were divided into COM crystal-treated group and control group according to whether the cell culture medium contains different concentrations of COM crystals. The concentrations of lactate dehydrogenase in the both group medium were determined after being cultured for 24 h. Protein and RNA were extracted from both cell groups after being cultured at different time points. SB239063, an inhibitor of the activation of p38 MAPK, was pretreated for 2 h before incubation with COM crystals. Western blotting and RT-qPCR were performed to confirm the expression levels of relative genes. All the experimental results were summarized and analyzed by SPSS 20.0 statistical analysis software. COM crystals (146 µg/cm2) could induce the expression levels of NLRP3, caspase-1 and interleukin-1β (IL-1β) significantly increased in NRK-52E cells. Compared with the control group cells, the transcription and translation levels of p38 MAPK-related molecule (such as p-p38) and adhesion molecules (such as osteopontin, hyaluronic acid and CD44) were significantly increased in COM crystal-treated cells and can be inhibited by SB239063 and NLRP3 gene silencing. This study demonstrated that the p38 MAPK signaling pathway mediated the COM crystal-induced crystal adhesion change in NRK-52E cells and required the involvement of NLRP3 inflammasome.
Collapse
Affiliation(s)
- Shiyong Qi
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Hexi District, Tianjin, 300211, China
| | - Qi Wang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Hexi District, Tianjin, 300211, China
| | - Bin Xie
- Department of Surgery, Linyi People's Hospital, Linyi, Shandong, China
| | - Yue Chen
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Hexi District, Tianjin, 300211, China
| | - Zhihong Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Hexi District, Tianjin, 300211, China
| | - Yong Xu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, 23 Pingjiang Road, Hexi District, Tianjin, 300211, China.
| |
Collapse
|
21
|
Bhowmick S, D'Mello V, Abdul-Muneer PM. Synergistic Inhibition of ERK1/2 and JNK, Not p38, Phosphorylation Ameliorates Neuronal Damages After Traumatic Brain Injury. Mol Neurobiol 2019; 56:1124-1136. [PMID: 29873042 DOI: 10.1007/s12035-018-1132-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 05/15/2018] [Indexed: 01/14/2023]
Abstract
Mitogen-activated protein (MAP) kinases are serine/threonine protein kinases that play a critical role in signal transduction and are activated by phosphorylation in response to a variety of pathophysiology stimuli. While MAP kinase signaling has a significant role in the pathophysiology of several neurodegenerative diseases, the precise function of activation of MAP kinase in traumatic brain injury (TBI) is unknown. Therefore, it is important to study the role of MAP kinase signaling in TBI-associated neurological ailments. In this study, using an in vitro stretch injury model in rat embryo neuronal cultures and the in vivo fluid percussion injury (FPI) model in rats, we explored the role of MAP kinase signaling in the mechanisms of cell death in TBI. Our study demonstrated that the stretch injury in vitro and FPI in vivo upregulated the phosphorylation of MAP kinase proteins ERK1/2 and JNK, but not p38. Using ERK1/2 inhibitor U0126, JNK inhibitor SP600125, and p38 inhibitor SB203580, we validated the role of MAP kinase proteins in the activation of NF-kB and caspase-3. By immunofluorescence and western blotting, further, we demonstrated the role of ERK1/2 and JNK phosphorylation in neurodegeneration by analyzing cell death proteins annexin V and Poly-ADP-Ribose-Polymerase p85. Interestingly, combined use of ERK1/2 and JNK inhibitors further attenuated the cell death in stretch-injured neurons. In conclusion, this study could establish the significance of MAP kinase signaling in the pathophysiology of TBI and may have significant implications for developing therapeutic strategies using ERK1/2 and JNK inhibitors for TBI-associated neurological complications.
Collapse
Affiliation(s)
- Saurav Bhowmick
- Laboratory of CNS Injury and Repair, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St., Edison, NJ, 08820, USA
| | - Veera D'Mello
- Laboratory of CNS Injury and Repair, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St., Edison, NJ, 08820, USA
| | - P M Abdul-Muneer
- Laboratory of CNS Injury and Repair, JFK Neuroscience Institute, Hackensack Meridian Health JFK Medical Center, 65 James St., Edison, NJ, 08820, USA.
| |
Collapse
|
22
|
Bian F, Yang XY, Xu G, Zheng T, Jin S. CRP-Induced NLRP3 Inflammasome Activation Increases LDL Transcytosis Across Endothelial Cells. Front Pharmacol 2019; 10:40. [PMID: 30761006 PMCID: PMC6363700 DOI: 10.3389/fphar.2019.00040] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/14/2019] [Indexed: 01/01/2023] Open
Abstract
The NLRP3 inflammasome, a multiprotein cytosolic complex that activates the IL-1 family of cytokines, plays an important role in atherosclerosis (AS). High-sensitivity c-reactive protein (hs-CRP) is widely recognized as a major cardiovascular risk predictor and recent studies name NLRP3 as a predictor of CRP levels. Mounting evidence has indicated that subendothelial retention of apolipoprotein B100-containing lipoproteins, such as low-density lipoprotein (LDL), is the initial step of atherogenesis, and is usually termed the “response to retention hypothesis.” We previously reported that CRP promotes AS by directly increasing LDL transcytosis across endothelial cells (ECs). The present study aims to investigate the effects of CRP on NLRP3 inflammasome activation and the role of the NLRP3 inflammasome in CRP-induced LDL transcytosis. We found that CRP upregulated NF-κB activity, the NF-κB inhibitor (BAY-11-7082) and Fcγ receptors (FcγRs) inhibitor (CD32/64Ab) blocked CRP-induced NF-κB activation. CRP also induced expression of pro-IL-1β and NLRP3, while BAY and CD32/64 Ab suppressed CRP-mediated expression of NLRP3 and pro-IL-1β. Moreover, CRP activated the NLRP3 inflammasome in ECs. NADPH oxidase inhibitor, diphenylene iodonium (DPI) and dithiothreitol (DTT), a broad-spectrum P2 receptor inhibitor, oxidized ATP (oATP), and a broad inhibitor of cysteine proteases, E-64d, inhibited CRP-induced NLRP3 inflammasome activation. Furthermore, NLRP3 siRNA and caspase-1 inhibitor blocked CRP-mediated LDL transcytosis across ECs. In conclusion, NLRP3 inflammasome activation was shown to be involved in CRP-mediated LDL transcytosis across ECs. CRP not only increased the expression of pro-IL-1β and NLRP3 via the FcγRs/NF-κB pathway, but also promoted NLRP3 inflammasome activation and IL-1β maturation by upregulation of reactive oxygen species (ROS) levels, purinergic receptor signaling, and activation of cysteine proteases.
Collapse
Affiliation(s)
- Fang Bian
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Pharmacy, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Xiao-Yan Yang
- Department of Pharmacology, Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gao Xu
- Department of Pharmacy, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Zheng
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Pharmacy, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Nebivolol prevents vascular oxidative stress and hypertension in rats chronically treated with ethanol. Atherosclerosis 2018; 274:67-76. [DOI: 10.1016/j.atherosclerosis.2018.04.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 04/16/2018] [Accepted: 04/27/2018] [Indexed: 01/15/2023]
|
24
|
Ismael S, Zhao L, Nasoohi S, Ishrat T. Inhibition of the NLRP3-inflammasome as a potential approach for neuroprotection after stroke. Sci Rep 2018; 8:5971. [PMID: 29654318 PMCID: PMC5899150 DOI: 10.1038/s41598-018-24350-x] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 03/23/2018] [Indexed: 11/10/2022] Open
Abstract
Activation of the NOD-like receptor protein (NLRP3)-inflammasome has been postulated to mediate inflammatory responses to brain damage during ischemic/reperfusion (I/R) injury. We therefore hypothesized that MCC950, a selective NLRP3-inflammasome inhibitor provides protection in mouse model of transient middle cerebral artery occlusion (tMCAO). Focal cerebral ischemia was induced by 60 min tMCAO followed by intraperitoneal administration of MCC950 (50 mg/kg) or saline at 1 h and 3 h post-occlusion. After 24 h of I/R, mice were tested for neurological outcome and were sacrificed for the analysis of infarct size and estimating NLRP3-inflammasome and apoptotic markers as well. Spectrophotometric method was used to determine hemoglobin (Hb) content as a marker of intracerebral hemorrhage. MCC950-treated mice showed a substantial reduction in infarction, edema and Hb content compared to saline controls in parallel with improved neurological deficits. MCC950 reduced expression of NLRP3-inflammasome cleavage products Caspase-1 and interlukin-1β (IL-1β) in penumbral region. These protective effects of MCC950 were associated with decreased TNF-α levels as well as poly (ADP-ribose) polymerase (PARP) and Caspase-3 cleavage and paralleled less phosphrylated NFκBp65 and IκBα levels. Taken together, these data indicate that inhibition of NLRP3-inflammasome with MCC950 has therapeutic potential in ischemic stroke models. Further investigations into the therapeutic efficacy and protocols are needed to confirm whether MCC950 treatment could be a promising candidate for clinical trials.
Collapse
Affiliation(s)
- Saifudeen Ismael
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Liang Zhao
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sanaz Nasoohi
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA.,Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, USA. .,Neuroscience Institute, The University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
25
|
|
26
|
Chen ML, Zhu XH, Ran L, Lang HD, Yi L, Mi MT. Trimethylamine-N-Oxide Induces Vascular Inflammation by Activating the NLRP3 Inflammasome Through the SIRT3-SOD2-mtROS Signaling Pathway. J Am Heart Assoc 2017; 6:JAHA.117.006347. [PMID: 28871042 PMCID: PMC5634285 DOI: 10.1161/jaha.117.006347] [Citation(s) in RCA: 389] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Trimethylamine‐N‐oxide (TMAO) has recently been identified as a novel and independent risk factor for promoting atherosclerosis through inducing vascular inflammation. However, the exact mechanism is currently unclear. Studies have established a central role of nucleotide‐binding oligomerization domain–like receptor family pyrin domain–containing 3 (NLRP3) inflammasome in the pathogenesis of vascular inflammation. Here, we examined the potential role of the NLRP3 inflammasome in TMAO‐induced vascular inflammation in vitro and in vivo and the underlying mechanisms. Methods and Results Experiments using liquid chromatography‐tandem mass spectrometry, Western blot, and fluorescent probes showed that TMAO‐induced inflammation in human umbilical vein endothelial cells (HUVECs) and aortas from ApoE−/− mice. Moreover, TMAO promoted NLRP3 and activated caspase‐1 p20 expression and caspase‐1 activity in vitro and in vivo. Notably, a caspase‐1 inhibitor (YVAD), an NLRP3 inhibitor (MCC950), as well as NLRP3 short interfering RNA attenuated TMAO‐induced activation of the NLRP3 inflammasome, subsequently leading to suppression of inflammation in HUVECs. TMAO additionally stimulated reactive oxygen species (ROS) generation, in particular, mitochondrial ROS, while inhibiting manganese superoxide dismutase 2 (SOD2) activation and sirtuin 3 (SIRT3) expression in HUVECs and aortas from ApoE−/− mice. TMAO‐induced endothelial NLRP3 inflammasome activation was ameliorated by the mitochondrial ROS scavenger Mito‐TEMPO, or SIRT3 overexpression in HUVECs. Conversely, TMAO failed to further inhibit magnesium SOD2 and activate the NLRP3 inflammasome or induce inflammation in SIRT3 short interfering RNA–treated HUVECs and aortas from SIRT3−/− mice. Conclusions TMAO promoted vascular inflammation by activating the NLRP3 inflammasome, and the NLRP3 inflammasome activation in part was mediated through inhibition of the SIRT3‐SOD2–mitochondrial ROS signaling pathway.
Collapse
Affiliation(s)
- Ming-Liang Chen
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, China.,Institute of Toxicology, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Xiao-Hui Zhu
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Li Ran
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, China
| | - He-Dong Lang
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Long Yi
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Man-Tian Mi
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing, China
| |
Collapse
|
27
|
Kalavakunta JK, Mittal MK, Janoudi A, Abela OG, Alreefi F, Abela GS. Role of Cholesterol Crystals During Acute Myocardial Infarction and Cerebrovascular Accident. CARDIOVASCULAR INNOVATIONS AND APPLICATIONS 2017. [DOI: 10.15212/cvia.2017.0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|