1
|
Seplovich G, Bouchi Y, de Rivero Vaccari JP, Pareja JCM, Reisner A, Blackwell L, Mechref Y, Wang KK, Tyndall JA, Tharakan B, Kobeissy F. Inflammasome links traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Neural Regen Res 2025; 20:1644-1664. [PMID: 39104096 PMCID: PMC11688549 DOI: 10.4103/nrr.nrr-d-24-00107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/20/2024] [Accepted: 06/03/2024] [Indexed: 08/07/2024] Open
Abstract
Traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease are three distinct neurological disorders that share common pathophysiological mechanisms involving neuroinflammation. One sequela of neuroinflammation includes the pathologic hyperphosphorylation of tau protein, an endogenous microtubule-associated protein that protects the integrity of neuronal cytoskeletons. Tau hyperphosphorylation results in protein misfolding and subsequent accumulation of tau tangles forming neurotoxic aggregates. These misfolded proteins are characteristic of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease and can lead to downstream neuroinflammatory processes, including assembly and activation of the inflammasome complex. Inflammasomes refer to a family of multimeric protein units that, upon activation, release a cascade of signaling molecules resulting in caspase-induced cell death and inflammation mediated by the release of interleukin-1β cytokine. One specific inflammasome, the NOD-like receptor protein 3, has been proposed to be a key regulator of tau phosphorylation where it has been shown that prolonged NOD-like receptor protein 3 activation acts as a causal factor in pathological tau accumulation and spreading. This review begins by describing the epidemiology and pathophysiology of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Next, we highlight neuroinflammation as an overriding theme and discuss the role of the NOD-like receptor protein 3 inflammasome in the formation of tau deposits and how such tauopathic entities spread throughout the brain. We then propose a novel framework linking traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease as inflammasome-dependent pathologies that exist along a temporal continuum. Finally, we discuss potential therapeutic targets that may intercept this pathway and ultimately minimize long-term neurological decline.
Collapse
Affiliation(s)
| | - Yazan Bouchi
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jennifer C. Munoz Pareja
- Division of Pediatric Critical Care, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrew Reisner
- Department of Pediatrics, Emory University, Atlanta, GA, USA
- Department of Neurosurgery, Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Laura Blackwell
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Kevin K. Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
2
|
Komiya H, Takeuchi H, Ogasawara A, Ogawa Y, Kubota S, Hashiguchi S, Takahashi K, Kunii M, Tanaka K, Tada M, Doi H, Tanaka F. Siponimod inhibits microglial inflammasome activation. Neurosci Res 2025; 213:138-145. [PMID: 39921000 DOI: 10.1016/j.neures.2025.02.002] [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: 09/15/2024] [Revised: 01/16/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Siponimod is the first oral drug approved for active secondary progressive multiple sclerosis. It acts as a functional antagonist of sphingosine-1-phosphate (S1P) receptor 1 (S1P1) through S1P1 internalization, and also serves an agonist of S1P5; however, the detailed mechanisms of its therapeutic effects on glial cells have yet to be elucidated. In this study, we investigated the anti-inflammatory mechanism of siponimod in microglia. Pretreatment with either siponimod or the S1P1 antagonist W146 significantly suppressed the production of interleukin-1β in activated microglia stimulated with lipopolysaccharide plus nigericin, an inflammasome activator. Furthermore, siponimod treatment reduced the protein levels of cleaved caspase-1 and inhibited the formation of aggregates of apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC specks) in microglia. Our data indicate that siponimod achieves its anti-inflammatory effects by inhibiting inflammasome activation in microglia via S1P1 antagonism. This process is inferred to play a crucial role in mitigating the secondary progression of multiple sclerosis, where microglial activation in the gray matter is considered a key pathological factor.
Collapse
Affiliation(s)
- Hiroyasu Komiya
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hideyuki Takeuchi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan; Department of Neurology, Graduate School of Medicine, International University of Health and Welfare, Narita, Japan; Center for Intractable Neurological Diseases and Dementia, International University of Health and Welfare Atami Hospital, Atami, Japan.
| | - Akihiro Ogasawara
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuki Ogawa
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shun Kubota
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shunta Hashiguchi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Keita Takahashi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Misako Kunii
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kenichi Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Mikiko Tada
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroshi Doi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| |
Collapse
|
3
|
Roy U, Hadad R, Rodriguez AA, Saju A, Roy D, Gil M, Keane RW, Scott RT, Mao XW, de Rivero Vaccari JP. Effects of Space Flight on Inflammasome Activation in the Brain of Mice. Cells 2025; 14:417. [PMID: 40136666 PMCID: PMC11941215 DOI: 10.3390/cells14060417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Space flight exposes astronauts to stressors that alter the immune response, rendering them vulnerable to infections and diseases. In this study, we aimed to determine the levels of inflammasome activation in the brains of mice that were housed in the International Space Station (ISS) for 37 days. C57BL/6 mice were launched to the ISS as part of NASA's Rodent Research 1 Mission on SpaceX-4 CRS-4 Dragon cargo spacecraft from 21 September 2014 to 25 October 2014. Dissected mouse brains from that mission were analyzed by immunoblotting of inflammasome signaling proteins and Electrochemiluminescence Immunoassay (ECLIA) for inflammatory cytokine levels. Our data indicate decreased inflammasome activation in the brains of mice that were housed in the ISS for 37 days when compared to the brains of mice that were maintained on the ground, and in mice corresponding to the baseline group that were sacrificed at the time of launching of SpaceX-4. Moreover, we did not detect any significant changes in the expression levels of the pro-inflammatory cytokines TNF-α, IL-2, IFN-γ, IL-5, IL-6, IL-12p70 and IL-10 between the ground control and the flight groups. Together, these studies suggest that spaceflight results in a decrease in the levels of innate immune signaling molecules that govern inflammasome signaling in the brain of mice.
Collapse
Affiliation(s)
- Upal Roy
- Department of Health and Biomedical Science, University of Texas Rio Grande Valley, Brownsville, TX 78539, USA; (A.A.R.)
| | - Roey Hadad
- Department of Cellular Physiology and Molecular Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Angel A. Rodriguez
- Department of Health and Biomedical Science, University of Texas Rio Grande Valley, Brownsville, TX 78539, USA; (A.A.R.)
| | - Alen Saju
- Department of Health and Biomedical Science, University of Texas Rio Grande Valley, Brownsville, TX 78539, USA; (A.A.R.)
| | - Deepa Roy
- Department of Health and Biomedical Science, University of Texas Rio Grande Valley, Brownsville, TX 78539, USA; (A.A.R.)
| | - Mario Gil
- Department of Psychological Science and School of Medicine Institute of Neuroscience, University of Texas Rio Grande Valley, Brownsville, TX 78539, USA
| | - Robert W. Keane
- Department of Cellular Physiology and Molecular Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ryan T. Scott
- KBR, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA 94035, USA
| | - Xiao W. Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University, Loma Linda, CA 92354, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Cellular Physiology and Molecular Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
4
|
Gu HY, Liu N. Mechanism of effect and therapeutic potential of NLRP3 inflammasome in spinal cord injury. Exp Neurol 2025; 384:115059. [PMID: 39571746 DOI: 10.1016/j.expneurol.2024.115059] [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: 10/09/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
Spinal cord injury (SCI) is a serious and disabling central nervous system injury that can trigger various neuropathological conditions, resulting in neuronal damage and release of various pro-inflammatory mediators, leading to neurological dysfunction. Currently, surgical decompression, drugs and rehabilitation are primarily used to relieve symptoms and improve endogenous repair mechanisms; however, they cannot directly promote nerve regeneration and functional recovery. SCI can be divided into primary and secondary injuries. Secondary injury is key to determining the severity of injury, whereas inflammation and cell death are important pathological mechanisms in the process of secondary SCI. The activation of the inflammasome complex is thought to be a necessary step in neuro-inflammation and a key trigger for neuronal death. The NLRP3 inflammasome is a cytoplasmic multiprotein complex that is considered an important factor in the development of SCI. Once the NLRP3 inflammasome is activated after SCI, NLRP3 nucleates the assembly of an inflammasome, leading to caspase 1-mediated proteolytic activation of the interleukin-1β (IL-1β) family of cytokines, and induces an inflammatory, pyroptotic cell death. Inhibition of inflammasomes can effectively inhibit inflammation and cell death in the body and promote the recovery of nerve function after SCI. Therefore, inhibition of NLRP3 inflammasome activation may be a promising approach for the treatment of SCI. In this review, we describe the current understanding of NLRP3 inflammasome activation in SCI pathogenesis and its subsequent impact on SCI and summarize drugs and other potential inhibitors based on NLRP3 inflammasome regulation. The objective of this study was to emphasize the role of the NLRP3 inflammasome in SCI, and provide a new therapeutic strategy and theoretical basis for targeting the NLRP3 inflammasome as a therapy for SCI.
Collapse
Affiliation(s)
- Hou-Yun Gu
- Department of Spine Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China; Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital), Southern Medical University, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China.
| | - Ning Liu
- Department of Spine Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China; Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital), Southern Medical University, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China.
| |
Collapse
|
5
|
Gober R, Dallmeier J, Davis D, Brzostowicki D, de Rivero Vaccari JP, Cyr B, Barreda A, Sun X, Gultekin SH, Garamszegi S, Scott W, Vontell R. Increased inflammasome protein expression identified in microglia from postmortem brains with schizophrenia. J Neuropathol Exp Neurol 2024; 83:951-966. [PMID: 38904417 PMCID: PMC11487111 DOI: 10.1093/jnen/nlae066] [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] [Indexed: 06/22/2024] Open
Abstract
Schizophrenia (SCZ) is a complex psychiatric disorder that involves an inflammatory response thought to be characterized by microglial activation. The inflammasome complex may play critical roles in the pathomechanism of neuroinflammation but how this relates to SCZ remains unclear. In this study, we performed an immunohistochemical (IHC) analysis to compare the expression of inflammasome proteins in brain tissue from donors with SCZ (n = 16) and non-psychiatric donors (NP; n = 13) isolated from the superior frontal cortex (SFC), superior temporal cortex, and anterior cingulate cortex brain regions. To assess changes in the cell populations that express key inflammasome proteins, we performed IHC analyses of apoptosis-associated speck-like protein containing a CARD (ASC), nod-like receptor protein 3 (NLRP3), and interleukin (IL)-18 to determine if these proteins are expressed in microglia, astrocytes, oligodendrocytes, or neurons. Inflammasome proteins were expressed mainly in microglia from SCZ and NP brains. Increased numbers of microglia were present in the SFC of SCZ brains and exhibited higher inflammasome protein expression of ASC, NLRP3, and IL-18 compared to NPs. These findings suggest that increased inflammasome signaling may contribute to the pathology underlying SCZ.
Collapse
Affiliation(s)
- Ryan Gober
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Julian Dallmeier
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
| | - David Davis
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Daniel Brzostowicki
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami, Miami, FL, United States
| | - Brianna Cyr
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami, Miami, FL, United States
| | - Ayled Barreda
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Xiaoyan Sun
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sakir Humayun Gultekin
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Susanna Garamszegi
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - William Scott
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Regina Vontell
- Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
6
|
Castellanos-Molina A, Bretheau F, Boisvert A, Bélanger D, Lacroix S. Constitutive DAMPs in CNS injury: From preclinical insights to clinical perspectives. Brain Behav Immun 2024; 122:583-595. [PMID: 39222725 DOI: 10.1016/j.bbi.2024.07.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 09/04/2024] Open
Abstract
Damage-associated molecular patterns (DAMPs) are endogenous molecules released in tissues upon cellular damage and necrosis, acting to initiate sterile inflammation. Constitutive DAMPs (cDAMPs) have the particularity to be present within the intracellular compartments of healthy cells, where they exert diverse functions such as regulation of gene expression and cellular homeostasis. However, after injury to the central nervous system (CNS), cDAMPs are rapidly released by stressed, damaged or dying neuronal, glial and endothelial cells, and can trigger inflammation without undergoing structural modifications. Several cDAMPs have been described in the injured CNS, such as interleukin (IL)-1α, IL-33, nucleotides (e.g. ATP), and high-mobility group box protein 1. Once in the extracellular milieu, these molecules are recognized by the remaining surviving cells through specific DAMP-sensing receptors, thereby inducing a cascade of molecular events leading to the production and release of proinflammatory cytokines and chemokines, as well as cell adhesion molecules. The ensuing immune response is necessary to eliminate cellular debris caused by the injury, allowing for damage containment. However, seeing as some molecules associated with the inflammatory response are toxic to surviving resident CNS cells, secondary damage occurs, aggravating injury and exacerbating neurological and behavioral deficits. Thus, a better understanding of these cDAMPs, as well as their receptors and downstream signaling pathways, could lead to identification of novel therapeutic targets for treating CNS injuries such as SCI, TBI, and stroke. In this review, we summarize the recent literature on cDAMPs, their specific functions, and the therapeutic potential of interfering with cDAMPs or their signaling pathways.
Collapse
Affiliation(s)
- Adrian Castellanos-Molina
- Axe Neurosciences du Centre de recherche du Centre hospitalier universitaire (CHU) de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Floriane Bretheau
- Axe Neurosciences du Centre de recherche du Centre hospitalier universitaire (CHU) de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Ana Boisvert
- Axe Neurosciences du Centre de recherche du Centre hospitalier universitaire (CHU) de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Dominic Bélanger
- Axe Neurosciences du Centre de recherche du Centre hospitalier universitaire (CHU) de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Steve Lacroix
- Axe Neurosciences du Centre de recherche du Centre hospitalier universitaire (CHU) de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada.
| |
Collapse
|
7
|
Chikopela T, Mwesigwa N, Masenga SK, Kirabo A, Shibao CA. The Interplay of HIV and Long COVID in Sub-Saharan Africa: Mechanisms of Endothelial Dysfunction. Curr Cardiol Rep 2024; 26:859-871. [PMID: 38958890 DOI: 10.1007/s11886-024-02087-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE OF REVIEW Long COVID affects approximately 5 million people in Africa. This disease is characterized by persistent symptoms or new onset of symptoms after an acute SARS-CoV-2 infection. Specifically, the most common symptoms include a range of cardiovascular problems such as chest pain, orthostatic intolerance, tachycardia, syncope, and uncontrolled hypertension. Importantly, these conditions appear to have endothelial dysfunction as the common denominator, which is often due to impaired nitric oxide (NO) mechanisms. This review discusses the role of mechanisms contributing to endothelial dysfunction in Long COVID, particularly in people living with HIV. RECENT FINDINGS Recent studies have reported that increased inflammation and oxidative stress, frequently observed in Long COVID, may contribute to NO dysfunction, ultimately leading to decreased vascular reactivity. These mechanisms have also been reported in people living with HIV. In regions like Africa, where HIV infection is still a major public health challenge with a prevalence of approximately 26 million people in 2022. Specifically, endothelial dysfunction has been reported as a major mechanism that appears to contribute to cardiovascular diseases and the intersection with Long COVID mechanisms is of particular concern. Further, it is well established that this population is more likely to develop Long COVID following infection with SARS-CoV-2. Therefore, concomitant infection with SARS-CoV-2 may lead to accelerated cardiovascular disease. We outline the details of the worsening health problems caused by Long COVID, which exacerbate pre-existing conditions such as endothelial dysfunction. The overlapping mechanisms of HIV and SARS-CoV-2, particularly the prolonged inflammatory response and chronic hypoxia, may increase susceptibility to Long COVID. Addressing these overlapping health issues is critical as it provides clinical entry points for interventions that could improve and enhance outcomes and quality of life for those affected by both HIV and Long COVID in the region.
Collapse
Affiliation(s)
- Theresa Chikopela
- Department of Human Physiology, Faculty of Medicine, Lusaka Apex Medical University, Lusaka, Zambia
| | - Naome Mwesigwa
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37332-0615, USA
| | - Sepiso K Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone, Zambia
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37332-0615, USA
| | - Cyndya A Shibao
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37332-0615, USA.
| |
Collapse
|
8
|
Yang H, Li D, Gao G. Kaempferol Alleviates Hepatic Injury in Nonalcoholic Steatohepatitis (NASH) by Suppressing Neutrophil-Mediated NLRP3-ASC/TMS1-Caspase 3 Signaling. Molecules 2024; 29:2630. [PMID: 38893506 PMCID: PMC11173805 DOI: 10.3390/molecules29112630] [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: 03/31/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a significant hepatic condition that has gained worldwide attention. Kaempferol (Kae), renowned for its diverse biological activities, including anti-inflammatory, antioxidant, anti-aging, and cardio-protective properties, has emerged as a potential therapeutic candidate for non-alcoholic steatohepatitis (NASH). Despite its promising therapeutic potential, the precise underlying mechanism of Kae's beneficial effects in NASH remains unclear. Therefore, this study aims to clarify the mechanism by conducting comprehensive in vivo and in vitro experiments. RESULTS In this study, a murine model of non-alcoholic steatohepatitis (NASH) was established by feeding C57BL/6 female mice a high-fat diet for 12 weeks. Kaempferol (Kae) was investigated for its ability to modulate systemic inflammatory responses and lipid metabolism in this model (20 mg/kg per day). Notably, Kae significantly reduced the expression of NLRP3-ASC/TMS1-Caspase 3, a crucial mediator of liver tissue inflammation. Additionally, in a HepG2 cell model induced with palmitic acid/oleic acid (PA/OA) to mimic NASH conditions, Kae demonstrated the capacity to decrease lipid droplet accumulation and downregulate the expression of NLRP3-ASC/TMS1-Caspase 3 (20 µM and the final concentration to 20 nM). These findings suggest that Kae may hold therapeutic potential in the treatment of NASH by targeting inflammatory and metabolic pathways. CONCLUSIONS These findings suggest that kaempferol holds potential as a promising therapeutic intervention for ameliorating non-alcoholic fatty liver disease (NAFLD).
Collapse
Affiliation(s)
- He Yang
- Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | | | - Guolan Gao
- Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
9
|
Cyr B, Cabrera Ranaldi EDLRM, Hadad R, Dietrich WD, Keane RW, de Rivero Vaccari JP. Extracellular vesicles mediate inflammasome signaling in the brain and heart of Alzheimer's disease mice. Front Mol Neurosci 2024; 17:1369781. [PMID: 38660388 PMCID: PMC11039928 DOI: 10.3389/fnmol.2024.1369781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/28/2024] [Indexed: 04/26/2024] Open
Abstract
Introduction Alzheimer's disease (AD) is an inflammatory neurodegenerative disease characterized by memory loss and cognitive impairment that worsens over time. AD is associated with many comorbidities, including cardiovascular disease that are associated with poorer outcomes. Comorbidities, especially heart disease and stroke, play a significant role in the demise of AD patients. Thus, it is important to understand how comorbidities are linked to AD. We have previously shown that extracellular vesicle (EV)-mediated inflammasome signaling plays an important role in the pathogenesis of brain injury and acute lung injury after traumatic brain injury. Methods We analyzed the cortical, hippocampal, ventricular, and atrial protein lysates from APP/PS1 mice and their respective controls for inflammasome signaling activation. Additionally, we analyzed serum-derived EV for size, concentration, and content of inflammasome proteins as well as the EV marker CD63. Finally, we performed conditioned media experiments of EV from AD patients and healthy age-matched controls delivered to cardiovascular cells in culture to assess EV-induced inflammation. Results We show a significant increase in Pyrin, NLRP1, caspase-1, and ASC in the brain cortex whereas caspase-8, ASC, and IL-1β were significantly elevated in the heart ventricles of AD mice when compared to controls. We did not find significant differences in the size or concentration of EV between groups, but there was a significant increase of caspase-1 and IL-1β in EV from AD mice compared to controls. In addition, conditioned media experiments of serum-derived EV from AD patients and age-matched controls delivered to cardiovascular cells in culture resulted in inflammasome activation, and significant increases in TNF-α and IL-2. Conclusion These results indicate that EV-mediated inflammasome signaling in the heart may play a role in the development of cardiovascular diseases in AD patients.
Collapse
Affiliation(s)
- Brianna Cyr
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Erika D. L. R. M. Cabrera Ranaldi
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Roey Hadad
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - W. Dalton Dietrich
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Robert W. Keane
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
10
|
Li W, Wang J, Tang C, Lv X, Zhu S. A Prospective Cohort Study of Elevated Serum NLRP1 Levels to Prognosticate Neurological Outcome After Acute Intracerebral Hemorrhage at a Single Academic Institution. Neuropsychiatr Dis Treat 2024; 20:737-753. [PMID: 38566883 PMCID: PMC10986417 DOI: 10.2147/ndt.s455049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/26/2024] [Indexed: 04/04/2024] Open
Abstract
Background Nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 1 (NLRP1) participates in neuroinflammation. This study aimed to identify serum NLRP as a potential prognostic biomarker of acute intracerebral hemorrhage (ICH). Methods This prospective cohort study enrolled 145 patients with supratentorial ICH and 51 healthy controls. Serum NLRP1 levels were quantified on admission of all 145 patients, on days 1, 3, 5, 7, and 10 after stroke in 51 of 145 patients and at entry into the study of controls. Poststroke 6-month modified Rankin Scale (mRS) scores of 3-6 signified a poor prognosis. Results Compared to controls, patients had prominently increased serum NLRP1 levels until day 10 after ICH, with the highest levels at days 1 and 3. Serum NLRP1 levels were independently correlated with National Institutes of Health Stroke Scale (NIHSS) scores, hematoma volume and six-month mRS scores, and independently predicted six-month bad prognosis. A linear relationship was observed between serum NLRP1 levels and the risk of poor prognosis in a restricted cubic spline. Under the receiver operating characteristic (ROC) curve, serum NLRP levels efficiently discriminated poor prognosis. Serum NLRP1, NIHSS, and hematoma volume were merged into a prognosis prediction model, which was portrayed using a nomogram. Good performance of the model was verified using calibration curve, decision curve, and ROC curve. Conclusion Serum NLRP1 levels are elevated during the early period following ICH and are independently related to hemorrhagic severity and poor prognosis, suggesting that serum NLRP1 may represent a promising prognostic biomarker of ICH.
Collapse
Affiliation(s)
- Wei Li
- Department of Neurosurgery, First People’s Hospital of Linping District, Hangzhou, People’s Republic of China
- Department of Neurosurgery, Linping Campus, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Jun Wang
- Department of Neurosurgery, First People’s Hospital of Linping District, Hangzhou, People’s Republic of China
- Department of Neurosurgery, Linping Campus, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Chao Tang
- Department of Neurosurgery, First People’s Hospital of Linping District, Hangzhou, People’s Republic of China
- Department of Neurosurgery, Linping Campus, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Xuan Lv
- Department of Neurosurgery, First People’s Hospital of Linping District, Hangzhou, People’s Republic of China
- Department of Neurosurgery, Linping Campus, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Suijun Zhu
- Department of Neurosurgery, First People’s Hospital of Linping District, Hangzhou, People’s Republic of China
- Department of Neurosurgery, Linping Campus, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| |
Collapse
|
11
|
Salemi M, Ravo M, Lanza G, Schillaci FA, Ventola GM, Marchese G, Salluzzo MG, Cappelletti G, Ferri R. Gene Expression Profiling of Post Mortem Midbrain of Parkinson's Disease Patients and Healthy Controls. Int J Mol Sci 2024; 25:707. [PMID: 38255780 PMCID: PMC10815072 DOI: 10.3390/ijms25020707] [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: 11/13/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 01/24/2024] Open
Abstract
Parkinson's disease (PD) stands as the most prevalent degenerative movement disorder, marked by the degeneration of dopaminergic neurons in the substantia nigra of the midbrain. In this study, we conducted a transcriptome analysis utilizing post mortem mRNA extracted from the substantia nigra of both PD patients and healthy control (CTRL) individuals. Specifically, we acquired eight samples from individuals with PD and six samples from CTRL individuals, with no discernible pathology detected in the latter group. RNA sequencing was conducted using the TapeStation 4200 system from Agilent Technologies. A total of 16,148 transcripts were identified, with 92 mRNAs displaying differential expression between the PD and control groups. Specifically, 33 mRNAs were significantly up-regulated, while 59 mRNAs were down-regulated in PD compared to the controls. The identification of statistically significant signaling pathways, with an adjusted p-value threshold of 0.05, unveiled noteworthy insights. Specifically, the enriched categories included cardiac muscle contraction (involving genes such as ATPase Na+/K+ transporting subunit beta 2 (ATP1B2), solute carrier family 8 member A1 (SLC8A1), and cytochrome c oxidase subunit II (COX2)), GABAergic synapse (involving GABA type A receptor-associated protein-like 1 (GABARAPL1), G protein subunit beta 5 (GNB5), and solute carrier family 38 member 2 (SLC38A2), autophagy (involving GABARAPL1 and tumor protein p53-inducible nuclear protein 2 (TP53INP2)), and Fc gamma receptor (FcγR) mediated phagocytosis (involving amphiphysin (AMPH)). These findings uncover new pathophysiological dimensions underlying PD, implicating genes associated with heart muscle contraction. This knowledge enhances diagnostic accuracy and contributes to the advancement of targeted therapies.
Collapse
Affiliation(s)
- Michele Salemi
- Oasi Research Institute–IRCCS, 94018 Troin, Italy; (G.L.); (F.A.S.); (M.G.S.); (R.F.)
| | - Maria Ravo
- Genomix4Life Srl, 94081 Baroniss, Italy; (M.R.); (G.M.V.); (G.M.)
- Genome Research Center for Health–CRGS, 94081 Baronissi, Italy
| | - Giuseppe Lanza
- Oasi Research Institute–IRCCS, 94018 Troin, Italy; (G.L.); (F.A.S.); (M.G.S.); (R.F.)
- Department of Surgery and Medical–Surgical Specialties, University of Catania, 95100 Catania, Italy
| | | | - Giovanna Maria Ventola
- Genomix4Life Srl, 94081 Baroniss, Italy; (M.R.); (G.M.V.); (G.M.)
- Genome Research Center for Health–CRGS, 94081 Baronissi, Italy
| | - Giovanna Marchese
- Genomix4Life Srl, 94081 Baroniss, Italy; (M.R.); (G.M.V.); (G.M.)
- Genome Research Center for Health–CRGS, 94081 Baronissi, Italy
| | - Maria Grazia Salluzzo
- Oasi Research Institute–IRCCS, 94018 Troin, Italy; (G.L.); (F.A.S.); (M.G.S.); (R.F.)
| | | | - Raffaele Ferri
- Oasi Research Institute–IRCCS, 94018 Troin, Italy; (G.L.); (F.A.S.); (M.G.S.); (R.F.)
| |
Collapse
|
12
|
Chen TC, Chen HL, Tseng WC, Chou TY, Tu JH, Parcell AC, Nosaka K. Contralateral versus ipsilateral protective effect against muscle damage of the elbow flexors and knee extensors induced by maximal eccentric exercise. Scand J Med Sci Sports 2023; 33:2548-2560. [PMID: 37642310 DOI: 10.1111/sms.14482] [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: 05/13/2023] [Revised: 07/30/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
The present study compared the ipsilateral repeated bout effect (IL-RBE) and contralateral repeated bout effect (CL-RBE) of the elbow flexors (EF) and knee flexors (KF) for the same interval between bouts to shed light on their mechanisms. Fifty-two healthy sedentary young (20-28 years) men were randomly assigned to the IL-EF, IL-KF, CL-EF, and CL-KF groups (n = 13/group). Thirty maximal eccentric contractions of the EF were performed in IL-EF and CL-EF, and 60 maximal eccentric contractions of the KF were performed in IL-KF and CL-KF, with a 2-week interval between bouts. Changes in muscle damage markers such as maximal voluntary contraction (MVC) torque, muscle soreness, and plasma creatine kinase activity, and proprioception measures before to 5 days post-exercise were compared between groups. Changes in all variables were greater (p < 0.05) after the first than second bout for all groups, and the changes were greater (p < 0.05) for the EF than KF. The changes in all variables after the second bout were greater (p < 0.05) for the CL than IL condition for both EF and KF. The magnitude of the average protective effect was similar between CL-EF (33%) and CL-KF (32%), but slightly greater (p < 0.05) for IL-EF (67%) than IL-KF (61%). These demonstrate that the magnitude of CL-RBE relative to IL-RBE was similar between the EF and KF (approximately 50%), regardless of the greater muscle damage for the EF than KF. It appears that the CL-RBE is more associated with neural adaptations at cerebrum, cerebellum, interhemispheric inhibition, and coricospinal tract, but the IL-RBE is induced by additional adaptations at muscles.
Collapse
Affiliation(s)
- Trevor C Chen
- Department of Physical Education and Sport Sciences, National Taiwan Normal University, Taipei City, Taiwan
| | - Hsin-Lian Chen
- Department of Physical Education, Health and Recreation, National Chiayi University, Chiayi County, Taiwan
| | - Wei-Chin Tseng
- Department of Physical Education, University of Taipei, Taipei City, Taiwan
| | - Tai-Ying Chou
- Department of Physical Education and Sport Sciences, National Taiwan Normal University, Taipei City, Taiwan
- Department of Athletic Performance, National Taiwan Normal University, Taipei City, Taiwan
| | - Jui-Hung Tu
- Department of Physical Education, National Pingtung University, Pingtung City, Taiwan
| | - Allen C Parcell
- Department of Exercise Sciences, Brigham Young University, Provo, Utah, USA
| | - Kazunori Nosaka
- Centre for Human Performance, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| |
Collapse
|
13
|
Cho K. Neutrophil-Mediated Progression of Mild Cognitive Impairment to Dementia. Int J Mol Sci 2023; 24:14795. [PMID: 37834242 PMCID: PMC10572848 DOI: 10.3390/ijms241914795] [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: 09/05/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/15/2023] Open
Abstract
Cognitive impairment is a serious condition that begins with amnesia and progresses to cognitive decline, behavioral dysfunction, and neuropsychiatric impairment. In the final stage, dysphagia and incontinence occur. There are numerous studies and developed drugs for cognitive dysfunction in neurodegenerative diseases, such as Alzheimer's disease (AD); however, their clinical effectiveness remains equivocal. To date, attempts have been made to overcome cognitive dysfunction and understand and delay the aging processes that lead to degenerative and chronic diseases. Cognitive dysfunction is involved in aging and the disruption of inflammation and innate immunity. Recent reports have indicated that the innate immune system is prevalent in patients with AD, and that peripheral neutrophil markers can predict a decline in executive function in patients with mild cognitive impairment (MCI). Furthermore, altered levels of pro-inflammatory interleukins have been reported in MCI, which have been suggested to play a role in the peripheral immune system during the process from early MCI to dementia. Neutrophils are the first responders of the innate immune system. Neutrophils eliminate harmful cellular debris via phagocytosis, secrete inflammatory factors to activate host defense systems, stimulate cytokine production, kill pathogens, and regulate extracellular proteases and inhibitors. This review investigated and summarized the regulation of neutrophil function during cognitive impairment caused by various degenerative diseases. In addition, this work elucidates the cellular mechanism of neutrophils in cognitive impairment and what is currently known about the effects of activated neutrophils on cognitive decline.
Collapse
Affiliation(s)
- KyoungJoo Cho
- Department of Life Science, Kyonggi University, Suwon 16227, Republic of Korea
| |
Collapse
|
14
|
Keane RW, Hadad R, Scott XO, Cabrera Ranaldi EDLRM, Pérez-Bárcena J, de Rivero Vaccari JP. Neural-Cardiac Inflammasome Axis after Traumatic Brain Injury. Pharmaceuticals (Basel) 2023; 16:1382. [PMID: 37895853 PMCID: PMC10610322 DOI: 10.3390/ph16101382] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/04/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Traumatic brain injury (TBI) affects not only the brain but also peripheral organs like the heart and the lungs, which influences long-term outcomes. A heightened systemic inflammatory response is often induced after TBI, but the underlying pathomechanisms that contribute to co-morbidities remain poorly understood. Here, we investigated whether extracellular vehicles (EVs) containing inflammasome proteins are released after severe controlled cortical impact (CCI) in C57BL/6 mice and cause activation of inflammasomes in the heart that result in tissue damage. The atrium of injured mice at 3 days after TBI showed a significant increase in the levels of the inflammasome proteins AIM2, ASC, caspases-1, -8 and -11, whereas IL-1β was increased in the ventricles. Additionally, the injured cortex showed a significant increase in IL-1β, ASC, caspases-1, -8 and -11 and pyrin at 3 days after injury when compared to the sham. Serum-derived extracellular vesicles (EVs) from injured patients were characterized with nanoparticle tracking analysis and Ella Simple Plex and showed elevated levels of the inflammasome proteins caspase-1, ASC and IL-18. Mass spectrometry of serum-derived EVs from mice after TBI revealed a variety of complement- and cardiovascular-related signaling proteins. Moreover, adoptive transfer of serum-derived EVs from TBI patients resulted in inflammasome activation in cardiac cells in culture. Thus, TBI elicits inflammasome activation, primarily in the atrium, that is mediated, in part, by EVs that contain inflammasome- and complement-related signaling proteins that are released into serum and contribute to peripheral organ systemic inflammation, which increases inflammasome activation in the heart.
Collapse
Affiliation(s)
- Robert W. Keane
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (R.W.K.); (E.d.l.R.M.C.R.)
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Roey Hadad
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Xavier O. Scott
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Erika d. l. R. M. Cabrera Ranaldi
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (R.W.K.); (E.d.l.R.M.C.R.)
| | - Jon Pérez-Bárcena
- Intensive Care Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (R.W.K.); (E.d.l.R.M.C.R.)
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
15
|
Cabrera Ranaldi EDLRM, Nuytemans K, Martinez A, Luca CC, Keane RW, de Rivero Vaccari JP. Proof-of-Principle Study of Inflammasome Signaling Proteins as Diagnostic Biomarkers of the Inflammatory Response in Parkinson's Disease. Pharmaceuticals (Basel) 2023; 16:883. [PMID: 37375830 DOI: 10.3390/ph16060883] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/07/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder marked by the death of dopaminergic neurons in the midbrain, the accumulation of α-synuclein aggregates, and motor deficits. A major contributor to dopaminergic neuronal loss is neuroinflammation. The inflammasome is a multiprotein complex that perpetuates neuroinflammation in neurodegenerative disorders including PD. Increases in inflammasome proteins are associated with worsened pathology. Thus, the inhibition of inflammatory mediators has the potential to aid in PD treatment. Here, we investigated inflammasome signaling proteins as potential biomarkers of the inflammatory response in PD. Plasma from PD subjects and healthy age-matched controls were evaluated for levels of the inflammasome protein apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), caspase-1, and interleukin (IL)-18. This was carried out using Simple Plex technology to identify changes in inflammasome proteins in the blood of PD subjects. The area under the curve (AUC) was obtained through calculation of the receiver operating characteristics (ROC) to obtain information on biomarker reliability and traits. Additionally, we completed a stepwise regression selected from the lowest Akaike information criterion (AIC) to assess how the inflammasome proteins caspase-1 and ASC contribute to IL-18 levels in people with PD. PD subjects demonstrated elevated caspase-1, ASC, and IL-18 levels when compared to controls; each of these proteins were found to be promising biomarkers of inflammation in PD. Furthermore, inflammasome proteins were determined to significantly contribute to and predict IL-18 levels in subjects with PD. Thus, we demonstrated that inflammasome proteins serve as reliable biomarkers of inflammation in PD and that inflammasome proteins provide significant contributions to IL-18 levels in PD.
Collapse
Affiliation(s)
- Erika D L R M Cabrera Ranaldi
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Karen Nuytemans
- The Dr. John T. Macdonald Foundation, Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Anisley Martinez
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Corneliu C Luca
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Robert W Keane
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
16
|
Yuan YG, Wang JL, Zhang YX, Li L, Reza AMMT, Gurunathan S. Biogenesis, Composition and Potential Therapeutic Applications of Mesenchymal Stem Cells Derived Exosomes in Various Diseases. Int J Nanomedicine 2023; 18:3177-3210. [PMID: 37337578 PMCID: PMC10276992 DOI: 10.2147/ijn.s407029] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/31/2023] [Indexed: 06/21/2023] Open
Abstract
Exosomes are nanovesicles with a wide range of chemical compositions used in many different applications. Mesenchymal stem cell-derived exosomes (MSCs-EXOs) are spherical vesicles that have been shown to mediate tissue regeneration in a variety of diseases, including neurological, autoimmune and inflammatory, cancer, ischemic heart disease, lung injury, and liver fibrosis. They can modulate the immune response by interacting with immune effector cells due to the presence of anti-inflammatory compounds and are involved in intercellular communication through various types of cargo. MSCs-EXOs exhibit cytokine storm-mitigating properties in response to COVID-19. This review discussed the potential function of MSCs-EXOs in a variety of diseases including neurological, notably epileptic encephalopathy and Parkinson's disease, cancer, angiogenesis, autoimmune and inflammatory diseases. We provided an overview of exosome biogenesis and factors that regulate exosome biogenesis. Additionally, we highlight the functions and potential use of MSCs-EXOs in the treatment of the inflammatory disease COVID-19. Finally, we covered a strategies and challenges of MSCs-EXOs. Finally, we discuss conclusion and future perspectives of MSCs-EXOs.
Collapse
Affiliation(s)
- Yu-Guo Yuan
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Jiangsu Co-Innovation Center of Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Jia-Lin Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Jiangsu Co-Innovation Center of Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Ya-Xin Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Jiangsu Co-Innovation Center of Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Ling Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Jiangsu Co-Innovation Center of Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Abu Musa Md Talimur Reza
- Department of Molecular Biology and Genetics, Faculty of Science, Gebze Technical University, Gebze, Kocaeli, Türkiye
| | | |
Collapse
|
17
|
Franklin ME, Bennett C, Arboite M, Alvarez-Ciara A, Corrales N, Verdelus J, Dietrich WD, Keane RW, de Rivero Vaccari JP, Prasad A. Activation of inflammasomes and their effects on neuroinflammation at the microelectrode-tissue interface in intracortical implants. Biomaterials 2023; 297:122102. [PMID: 37015177 PMCID: PMC10614166 DOI: 10.1016/j.biomaterials.2023.122102] [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: 10/11/2022] [Revised: 03/16/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023]
Abstract
Invasive neuroprosthetics rely on microelectrodes (MEs) to record or stimulate the activity of large neuron assemblies. However, MEs are subjected to tissue reactivity in the central nervous system (CNS) due to the foreign body response (FBR) that contribute to chronic neuroinflammation and ultimately result in ME failure. An endogenous, acute set of mechanisms responsible for the recognition and targeting of foreign objects, called the innate immune response, immediately follows the ME implant-induced trauma. Inflammasomes are multiprotein structures that play a critical role in the initiation of an innate immune response following CNS injuries. The activation of inflammasomes facilitates a range of innate immune response cascades and results in neuroinflammation and programmed cell death. Despite our current understanding of inflammasomes, their roles in the context of neural device implantation remain unknown. In this study, we implanted a non-functional Utah electrode array (UEA) into the rat somatosensory cortex and studied the inflammasome signaling and the corresponding downstream effects on inflammatory cytokine expression and the inflammasome-mediated cell death mechanism of pyroptosis. Our results not only demonstrate the continuous activation of inflammasomes and their contribution to neuroinflammation at the electrode-tissue interface but also reveal the therapeutic potential of targeting inflammasomes to attenuate the FBR in invasive neuroprosthetics.
Collapse
Affiliation(s)
- Melissa E Franklin
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Cassie Bennett
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Maelle Arboite
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | | | - Natalie Corrales
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Jennifer Verdelus
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - W Dalton Dietrich
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA
| | - Robert W Keane
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, USA
| | - Abhishek Prasad
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA.
| |
Collapse
|
18
|
Cyr B, de Rivero Vaccari JP. Sex Differences in the Inflammatory Profile in the Brain of Young and Aged Mice. Cells 2023; 12:1372. [PMID: 37408205 DOI: 10.3390/cells12101372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/05/2023] [Accepted: 05/10/2023] [Indexed: 07/07/2023] Open
Abstract
Neurodegenerative diseases are a leading cause of death worldwide with no cures identified. Thus, there is a critical need for preventative measures and treatments as the number of patients is expected to increase. Many neurodegenerative diseases have sex-biased prevalence, indicating a need to examine sex differences when investigating prevention and treatment strategies. Inflammation is a key contributor to many neurodegenerative diseases and is a promising target for prevention since inflammation increases with age, which is known as inflammaging. Here, we analyzed the protein expression levels of cytokines, chemokines, and inflammasome signaling proteins in the cortex of young and aged male and female mice. Our results show an increase in caspase-1, interleukin (IL)-1β, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and ASC specks in females compared to males. Additionally, there was an increase in IL-1α, VEGF-A, CCL3, CXCL1, CCL4, CCL17, and CCL22 in aging females and an increase in IL-8, IL-17a, IL-7, LT-α, and CCL22 in aging males. IL-12/IL-23p40, CCL13, and IL-10 were increased in females compared to males but not with age. These results indicate that there are sex differences in cortical inflammaging and provide potential targets to attenuate inflammation to prevent the development of neurodegenerative disease.
Collapse
Affiliation(s)
- Brianna Cyr
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Center for Cognitive Neuroscience and Aging, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
19
|
Chen J, Shen Y, Shao X, Wu W. An emerging role of inflammasomes in spinal cord injury and spinal cord tumor. Front Immunol 2023; 14:1119591. [PMID: 36969234 PMCID: PMC10033975 DOI: 10.3389/fimmu.2023.1119591] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Spinal cord injury (SCI) and spinal cord tumor are devastating events causing structural and functional impairment of the spinal cord and resulting in high morbidity and mortality; these lead to a psychological burden and financial pressure on the patient. These spinal cord damages likely disrupt sensory, motor, and autonomic functions. Unfortunately, the optimal treatment of and spinal cord tumors is limited, and the molecular mechanisms underlying these disorders are unclear. The role of the inflammasome in neuroinflammation in diverse diseases is becoming increasingly important. The inflammasome is an intracellular multiprotein complex and participates in the activation of caspase-1 and the secretion of pro-inflammatory cytokines such as interleukin (IL)-1β and IL-18. The inflammasome in the spinal cord is involved in the stimulation of immune-inflammatory responses through the release of pro-inflammatory cytokines, thereby mediating further spinal cord damage. In this review, we highlight the role of inflammasomes in SCI and spinal cord tumors. Targeting inflammasomes is a promising therapeutic strategy for the treatment of SCI and spinal cord tumors.
Collapse
|
20
|
Mandell JT, de Rivero Vaccari JP, Sabater AL, Galor A. The inflammasome pathway: A key player in ocular surface and anterior segment diseases. Surv Ophthalmol 2023; 68:280-289. [PMID: 35798189 DOI: 10.1016/j.survophthal.2022.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 01/06/2023]
Abstract
Inflammasomes are multicomplex molecular regulators with an emerging importance in regulating ocular surface and anterior segment health and disease. Key components found in the eye include NF-κB, NLRP3, NLRC4, NLRP6, ASC, IL-1β, IL-18, and caspase-1. The role of NLRP1, NLRC4, AIM2, and NLRP3 inflammasomes in the pathogenesis of infectious ulcers, DED, uveitis, glaucoma, corneal edema, and other diseases is being studied with many developments. Attenuation of these diseases has been explored by blocking various molecules along the inflammasome pathway with agents like NAC, polydatin, calcitriol, glyburide, YVAD, and disulfiram. We provide a background on the inflammasome pathway as it relates to the ocular surface and anterior segment of the eye, discuss the role of inflammasomes in the above diseases in animals and humans, investigate new therapeutic targets, and explore the efficacy of new anti-inflammasome therapies.
Collapse
Affiliation(s)
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, FL, USA
| | | | - Anat Galor
- Bascom Palmer Eye Institute, University of Miami, Miami, FL, USA; Ophthalmology, Miami Veterans Affairs (VA) Medical Center, Miami, FL, USA.
| |
Collapse
|
21
|
de Rivero Vaccari JP, Mim C, Hadad R, Cyr B, Stefansdottir TA, Keane RW. Mechanism of action of IC 100, a humanized IgG4 monoclonal antibody targeting apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC). Transl Res 2023; 251:27-40. [PMID: 35793783 PMCID: PMC10615563 DOI: 10.1016/j.trsl.2022.06.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 02/09/2023]
Abstract
Inflammasomes are multiprotein complexes of the innate immune response that recognize a diverse range of intracellular sensors of infection or cell damage and recruit the adaptor protein apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) into an inflammasome signaling complex. The recruitment, polymerization and cross-linking of ASC is upstream of caspase-1 activation and interleukin-1β release. Here we provide evidence that IC 100, a humanized IgG4κ monoclonal antibody against ASC, is internalized into the cell and localizes with endosomes, while another part is recycled and redistributed out of the cell. IC 100 binds intracellular ASC and blocks interleukin-1β release in a human whole blood cell inflammasome assay. In vitro studies demonstrate that IC 100 interferes with ASC polymerization and assembly of ASC specks. In vivo bioluminescence imaging showed that IC 100 has broad tissue distribution, crosses the blood brain barrier, and readily penetrates the brain and spinal cord parenchyma. Confocal microscopy of fluorescent-labeled IC 100 revealed that IC 100 is rapidly taken up by macrophages via a mechanism utilizing the Fc region of IC 100. Coimmunoprecipitation experiments and confocal immunohistochemistry showed that IC 100 binds to ASC and to the atypical antibody receptor Tripartite motif-containing protein-21 (TRIM21). In A549 WT and TRIM21 KO cells treated with either IC 100 or IgG4κ isotype control, the levels of intracellular IC 100 were higher than in the IgG4κ-treated controls at 2 hours, 1 day and 3 days after administration, indicating that IC 100 escapes degradation by the proteasome. Lastly, electron microscopy studies demonstrate that IC 100 binds to ASC filaments and alters the architecture of ASC filaments. Thus, IC 100 readily penetrates a variety of cell types, and it binds to intracellular ASC, but it is not degraded by the TRIM21 antibody-dependent intracellular neutralization pathway.
Collapse
Affiliation(s)
- Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL
| | - Carsten Mim
- Department of Biomedical Engineering and Health Systems, Kungliga Tekniska Högscholan (Royal Institute of Technology), Sweden
| | - Roey Hadad
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL
| | - Brianna Cyr
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL
| | - Thorunn Anna Stefansdottir
- Department of Biomedical Engineering and Health Systems, Kungliga Tekniska Högscholan (Royal Institute of Technology), Sweden
| | - Robert W Keane
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL; Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL.
| |
Collapse
|
22
|
Kattan D, Barsa C, Mekhijian S, Shakkour Z, Jammoul M, Doumit M, Zabala MCP, Darwiche N, Eid AH, Mechref Y, Wang KK, de Rivero Vaccari JP, Munoz Pareja JC, Kobeissy F. Inflammasomes as biomarkers and therapeutic targets in traumatic brain injury and related-neurodegenerative diseases: A comprehensive overview. Neurosci Biobehav Rev 2023; 144:104969. [PMID: 36423707 PMCID: PMC9805531 DOI: 10.1016/j.neubiorev.2022.104969] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/18/2022] [Accepted: 11/20/2022] [Indexed: 11/23/2022]
Abstract
Given the ambiguity surrounding traumatic brain injury (TBI) pathophysiology and the lack of any Food and Drug Administration (FDA)-approved neurotherapeutic drugs, there is an increasing need to better understand the mechanisms of TBI. Recently, the roles of inflammasomes have been highlighted as both potential therapeutic targets and diagnostic markers in different neurodegenerative disorders. Indeed, inflammasome activation plays a pivotal function in the central nervous system (CNS) response to many neurological conditions, as well as to several neurodegenerative disorders, specifically, TBI. This comprehensive review summarizes and critically discusses the mechanisms that govern the activation and assembly of inflammasome complexes and the major methods used to study inflammasome activation in TBI and its implication for other neurodegenerative disorders. Also, we will review how inflammasome activation is critical in CNS homeostasis and pathogenesis, and how it can impact chronic TBI sequalae and increase the risk of developing neurodegenerative diseases. Additionally, we discuss the recent updates on inflammasome-related biomarkers and the potential to utilize inflammasomes as putative therapeutic targets that hold the potential to better diagnose and treat subjects with TBI.
Collapse
Affiliation(s)
- Dania Kattan
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Chloe Barsa
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Sarin Mekhijian
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Zaynab Shakkour
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon; Program for Interdisciplinary Neuroscience, Department of Child Health, School of Medicine, University of Missouri, USA
| | - Maya Jammoul
- Department of Anatomy, Cell Biology, and Physiology, American University of Beirut, Beirut, Lebanon
| | - Mark Doumit
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Maria Camila Pareja Zabala
- Division of Pediatric Critical Care, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nadine Darwiche
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Kevin K Wang
- Morehouse School of Medicine, Department of Neurobiology, Atlanta, GA, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Jennifer C Munoz Pareja
- Division of Pediatric Critical Care, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon; Morehouse School of Medicine, Department of Neurobiology, Atlanta, GA, USA.
| |
Collapse
|
23
|
Cyr B, de Rivero Vaccari JP. Methods to Study Inflammasome Activation in the Central Nervous System: Immunoblotting and Immunohistochemistry. Methods Mol Biol 2023; 2696:223-238. [PMID: 37578726 DOI: 10.1007/978-1-0716-3350-2_15] [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: 08/15/2023]
Abstract
The inflammasome is a multiprotein complex that is responsible for mounting an innate immune response through the activation of caspase-1 and the cleavage of interleukin-1β. This multiprotein complex plays an important role in a variety of central nervous system (CNS) diseases and conditions such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, stroke, and traumatic brain injury, among others. Here we describe methodological procedures to carry out immunoblotting and immunohistochemical techniques used to study inflammasome signaling in CNS tissues (brain and spinal cord).
Collapse
Affiliation(s)
- Brianna Cyr
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
24
|
Vontell RT, de Rivero Vaccari JP, Sun X, Gultekin SH, Bramlett HM, Dietrich WD, Keane RW. Identification of inflammasome signaling proteins in neurons and microglia in early and intermediate stages of Alzheimer's disease. Brain Pathol 2022:e13142. [PMID: 36579934 DOI: 10.1111/bpa.13142] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/08/2022] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that destroys memory and cognitive function. Inflammasome activation has been suggested to play a critical role in the neuroinflammatory response in AD progression, but the cell-type expression of inflammasome proteins in the brain has not been fully characterized. In this study, we used samples from the hippocampus formation, the subiculum, and the entorhinal cortex brain from 17 donors with low-level AD pathology and 17 intermediate AD donors to assess the expression of inflammasome proteins. We performed analysis of hippocampal thickness, β-amyloid plaques, and hyperphosphorylated tau to ascertain the cellular pathological changes that occur between low and intermediate AD pathology. Next, we determined changes in the cells that express the inflammasome sensor proteins NOD-like receptor proteins (NLRP) 1 and 3, and caspase-1. In addition, we stained section with IC100, a humanized monoclonal antibody directed against the inflammasome adaptor protein apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and a commercially available anti-ASC antibody. Our results indicate that hippocampal cortical thickness did not significantly change between low and intermediate AD pathology, but there was an increase in pTau and β-amyloid clusters in intermediate AD cases. NLRP3 was identified mainly in microglial populations, whereas NLRP1 was seen in neuronal cytoplasmic regions. There was a significant increase of ASC in neurons labeled by IC100, whereas microglia in the hippocampus and subiculum were labeled with the commercial anti-ASC antibody. Caspase-1 was present in the parenchyma in the CA regions where amyloid and pTau were identified. Together, our results indicate increased inflammasome protein expression in the early pathological stages of AD, that IC100 identifies neurons in early stages of AD and that ASC expression correlates with Aβ and pTau in postmortem AD brains.
Collapse
Affiliation(s)
- Regina T Vontell
- Department of Neurology, University of Miami Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, Florida, USA.,Evelyn F. McKnight Brain Institute, Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida, USA.,Center for Cognitive Neuroscience and Aging, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Xiaoyan Sun
- Department of Neurology, University of Miami Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, Florida, USA.,Evelyn F. McKnight Brain Institute, Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sakir Humayun Gultekin
- Department of Neurology, University of Miami Brain Endowment Bank, University of Miami Miller School of Medicine, Miami, Florida, USA.,Evelyn F. McKnight Brain Institute, Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA.,Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Helen M Bramlett
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA.,Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida, USA
| | - W Dalton Dietrich
- Evelyn F. McKnight Brain Institute, Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA.,Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA.,Center for Cognitive Neuroscience and Aging, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Robert W Keane
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, USA.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
25
|
Yin J, Gong G, Wan W, Liu X. Pyroptosis in spinal cord injury. Front Cell Neurosci 2022; 16:949939. [PMID: 36467606 PMCID: PMC9715394 DOI: 10.3389/fncel.2022.949939] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 11/03/2022] [Indexed: 10/21/2023] Open
Abstract
Spinal cord injury (SCI) often brings devastating consequences to patients and their families. Pathophysiologically, the primary insult causes irreversible damage to neurons and glial cells and initiates the secondary damage cascade, further leading to inflammation, ischemia, and cells death. In SCI, the release of various inflammatory mediators aggravates nerve injury. Pyroptosis is a new pro-inflammatory pattern of regulated cell death (RCD), mainly mediated by caspase-1 or caspase-11/4/5. Gasdermins family are pore-forming proteins known as the executor of pyroptosis and the gasdermin D (GSDMD) is best characterized. Pyroptosis occurs in multiple central nervous system (CNS) cell types, especially plays a vital role in the development of SCI. We review here the evidence for pyroptosis in SCI, and focus on the pyroptosis of different cells and the crosstalk between them. In addition, we discuss the interaction between pyroptosis and other forms of RCD in SCI. We also summarize the therapeutic strategies for pyroptosis inhibition, so as to provide novel ideas for improving outcomes following SCI.
Collapse
Affiliation(s)
- Jian Yin
- Department of Orthopedics, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, China
- Department of Orthopedics, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Ge Gong
- Department of Geriatrics, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenhui Wan
- Department of Geriatrics, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xinhui Liu
- Department of Orthopedics, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, China
| |
Collapse
|
26
|
Hu X, Zhang Y, Wang L, Ding J, Li M, Li H, Wu L, Zeng Z, Xia H. Microglial activation in the motor cortex mediated NLRP3-related neuroinflammation and neuronal damage following spinal cord injury. Front Cell Neurosci 2022; 16:956079. [PMID: 36339822 PMCID: PMC9630363 DOI: 10.3389/fncel.2022.956079] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/27/2022] [Indexed: 12/30/2023] Open
Abstract
Spinal cord injury (SCI) is a traumatic event that can lead to neurodegeneration. Neuronal damage in the primary motor cortex (M1) can hinder motor function recovery after SCI. However, the exact mechanisms involved in neuronal damage after SCI remain incompletely understood. In this study, we found that microglia were activated in M1 after SCI, which triggered Nod-like receptor protein 3 (NLRP3) related chronic neuroinflammation and neuronal damage in vivo. Meanwhile, treatment with the microglia inhibitor minocycline reduced inflammation-induced neuronal damage in M1, protected the integrity of the motor conduction pathway, and promoted motor function recovery. Furthermore, we simulated chronic inflammation in M1 after SCI by culturing the primary neurons in primary microglia-conditioned medium, and observed that the injury to the primary neurons also occurred in vitro; however, as observed in vivo, these effects could be mitigated by minocycline treatment. Our results indicated that microglial activation in M1 mediates NLRP3-related neuroinflammation and causes the injury to M1 neurons, thereby impairing the integrity of the motor conduction pathway and inhibiting motor function recovery. These findings might contribute to the identification of novel therapeutic strategies for SCI.
Collapse
Affiliation(s)
- Xvlei Hu
- Department of Neurosurgery, Shanxi Provincial People's Hospital, Taiyuan, China
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yinchuan, China
| | - Yifan Zhang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Ningxia Human Stem Cell Research Institute, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Lei Wang
- Department of Neurosurgery, Henan Provincial People's Hospital, Zhengzhou, China
| | - Jiangwei Ding
- Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yinchuan, China
| | - Mei Li
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yinchuan, China
| | - Hailiang Li
- Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yinchuan, China
| | - Liang Wu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zhong Zeng
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Craniocerebral Diseases, Ningxia Medical University, Yinchuan, China
| | - Hechun Xia
- Ningxia Human Stem Cell Research Institute, General Hospital of Ningxia Medical University, Yinchuan, China
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
27
|
Serna-Rodríguez MF, Bernal-Vega S, de la Barquera JAOS, Camacho-Morales A, Pérez-Maya AA. The role of damage associated molecular pattern molecules (DAMPs) and permeability of the blood-brain barrier in depression and neuroinflammation. J Neuroimmunol 2022; 371:577951. [PMID: 35994946 DOI: 10.1016/j.jneuroim.2022.577951] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 08/03/2022] [Accepted: 08/13/2022] [Indexed: 10/15/2022]
Abstract
Depression is a heterogeneous mental disorder characterized by feelings of sadness and loss of interest that render the subject unable to handle basic daily activities such as sleeping, eating, or working. Neurobiological traits leading to depression include genetic background, early life abuse, life stressors, and systemic and central inflammatory profiles. Several clinical and preclinical reports documented that depression shows an increase in pro-inflammatory markers such as interleukin (IL-)1β, IL-6, IL-12, tumor necrosis factor (TNF), and interferon (IFN)-γ; and a decrease in anti-inflammatory IL-4, IL-10, and transforming growth factor (TGF)-β species. Inflammatory activation may trigger and maintain depression. Dynamic crosstalk between the peripheral immune system and the central nervous system (CNS) such as activated endothelial cells, monocytes, monocyte-derived dendritic cells, macrophages, T cells, and microglia has been proposed as a leading cause of neuroinflammation. Notably, pro-inflammatory cytokines disrupt the hypothalamic-pituitary-adrenal (HPA) axis and serotonergic, noradrenergic, dopaminergic, and glutamatergic neurotransmission. While still under investigation, peripheral cytokines can engage brain pathways and affect the central synthesis of HPA hormones and neurotransmitters through several mechanisms such as activation of the vagus nerve, increasing the permeability of the blood-brain barrier (BBB), altered cytokines transport systems, and engaging toll-like receptors (TLRs) by pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). However, physiological mechanisms that favor time-dependent central inflammation before or during illness are not totally understood. This review will provide preclinical and clinical evidence of DAMPs and the BBB permeability as contributors to depression and neuroinflammation. We will also discuss pharmacologic approaches that could potentially modulate DAMPs and BBB permeability for future interventions against major depression.
Collapse
Affiliation(s)
- María Fernanda Serna-Rodríguez
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Departamento de Bioquímica y Medicina Molecular. Monterrey CP. 64460, Nuevo Leon, Mexico
| | - Sofía Bernal-Vega
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Departamento de Bioquímica y Medicina Molecular. Monterrey CP. 64460, Nuevo Leon, Mexico
| | | | - Alberto Camacho-Morales
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Departamento de Bioquímica y Medicina Molecular. Monterrey CP. 64460, Nuevo Leon, Mexico.
| | - Antonio Alí Pérez-Maya
- Universidad Autónoma de Nuevo León, Facultad de Medicina, Departamento de Bioquímica y Medicina Molecular. Monterrey CP. 64460, Nuevo Leon, Mexico.
| |
Collapse
|
28
|
Arik E, Heinisch O, Bienert M, Gubeljak L, Slowik A, Reich A, Schulz JB, Wilhelm T, Huber M, Habib P. Erythropoietin Enhances Post-ischemic Migration and Phagocytosis and Alleviates the Activation of Inflammasomes in Human Microglial Cells. Front Cell Neurosci 2022; 16:915348. [PMID: 35813499 PMCID: PMC9263298 DOI: 10.3389/fncel.2022.915348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/08/2022] [Indexed: 11/19/2022] Open
Abstract
Recombinant human erythropoietin (rhEPO) has been shown to exert anti-apoptotic and anti-inflammatory effects after cerebral ischemia. Inflammatory cytokines interleukin-1β and -18 (IL-1β and IL-18) are crucial mediators of apoptosis and are maturated by multiprotein complexes termed inflammasomes. Microglia are the first responders to post-ischemic brain damage and are a main source of inflammasomes. However, the impact of rhEPO on microglial activation and the subsequent induction of inflammasomes after ischemia remains elusive. To address this, we subjected human microglial clone 3 (HMC-3) cells to various durations of oxygen-glucose-deprivation/reperfusion (OGD/R) to assess the impact of rhEPO on cell viability, metabolic activity, oxidative stress, phagocytosis, migration, as well as on the regulation and activation of the NLRP1, NLRP3, NLRC4, and AIM2 inflammasomes. Administration of rhEPO mitigated OGD/R-induced oxidative stress and cell death. Additionally, it enhanced metabolic activity, migration and phagocytosis of HMC-3. Moreover, rhEPO attenuated post-ischemic activation and regulation of the NLRP1, NLRP3, NLRC4, and AIM2 inflammasomes as well as their downstream effectors CASPASE1 and IL-1β. Pharmacological inhibition of NLRP3 via MCC950 had no effect on the activation of CASPASE1 and maturation of IL-1β after OGD/R, but increased protein levels of NLRP1, NLRC4, and AIM2, suggesting compensatory activities among inflammasomes. We provide evidence that EPO-conveyed anti-inflammatory actions might be mediated via the regulation of the inflammasomes.
Collapse
Affiliation(s)
- Eren Arik
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Ole Heinisch
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Michaela Bienert
- Institute of Molecular and Cellular Anatomy, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Lara Gubeljak
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Alexander Slowik
- Department of Anatomy and Cell Biology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Arno Reich
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Jörg B. Schulz
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
- JARA-BRAIN Institute of Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
| | - Thomas Wilhelm
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Pardes Habib
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
- JARA-BRAIN Institute of Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
- *Correspondence: Pardes Habib, ; orcid.org/0000-0002-5771-216X
| |
Collapse
|
29
|
Mi L, Min X, Chai Y, Zhang J, Chen X. NLRP1 Inflammasomes: A Potential Target for the Treatment of Several Types of Brain Injury. Front Immunol 2022; 13:863774. [PMID: 35707533 PMCID: PMC9189285 DOI: 10.3389/fimmu.2022.863774] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/28/2022] [Indexed: 12/28/2022] Open
Abstract
NOD-like receptor (NLR) family pyrin domain-containing 1 (NLRP1) is a member of the NLR family. The NLRP1 inflammasome consists of the NLRP1 protein, the adaptor protein apoptosis-associated speck-like protein containing a CARD domain, and the effector molecule pro-caspase-1. When stimulated, the inflammasome initiates the cleavage of pro-caspase-1 and converts it into its active form, caspase-1; then, caspase-1 facilitates the cleavage of the proinflammatory cytokines interleukin-1β and interleukin-18 into their active and secreted forms. In addition, caspase-1 also mediates the cleavage of gasdermin D, which leads to pyroptosis, an inflammatory form of cell death. Pathological events that damage the brain and result in neuropathological conditions can generally be described as brain injury. Neuroinflammation, especially that driven by NLRP1, plays a considerable role in the pathophysiology of brain injury, such as early brain injury (EBI) of subarachnoid hemorrhage, ischemic brain injury during stroke, and traumatic brain injury (TBI). In this article, a thorough overview of NLRP1 is presented, including its structure, mechanism of activation, and role in neuroinflammation. We also present recent studies on NLRP1 as a target for the treatment of EBI, ischemic brain injury, TBI, and other types of brain injury, thus highlighting the perspective of NLRP1 as an effective mediator of catastrophic brain injury.
Collapse
Affiliation(s)
- Liang Mi
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Posttrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Xiaobin Min
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Baodi Clinical College, Tianjin Medical University, Tianjin, China
| | - Yan Chai
- Tianjin Neurological Institute, Key Laboratory of Posttrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Posttrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Xin Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Neurological Institute, Key Laboratory of Posttrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
- *Correspondence: Xin Chen,
| |
Collapse
|
30
|
Johnson NH, Hadad R, Taylor RR, Rodríguez Pilar J, Salazar O, Llompart-Pou JA, Dietrich WD, Keane RW, Pérez-Bárcena J, de Rivero Vaccari JP. Inflammatory Biomarkers of Traumatic Brain Injury. Pharmaceuticals (Basel) 2022; 15:ph15060660. [PMID: 35745576 PMCID: PMC9227014 DOI: 10.3390/ph15060660] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 12/26/2022] Open
Abstract
Traumatic brain injury (TBI) has a complex pathology in which the initial injury releases damage associated proteins that exacerbate the neuroinflammatory response during the chronic secondary injury period. One of the major pathological players in the inflammatory response after TBI is the inflammasome. Increased levels of inflammasome proteins during the acute phase after TBI are associated with worse functional outcomes. Previous studies reveal that the level of inflammasome proteins in biological fluids may be used as promising new biomarkers for the determination of TBI functional outcomes. In this study, we provide further evidence that inflammatory cytokines and inflammasome proteins in serum may be used to determine injury severity and predict pathological outcomes. In this study, we analyzed blood serum from TBI patients and respective controls utilizing Simple Plex inflammasome and V-PLEX inflammatory cytokine assays. We performed statistical analyses to determine which proteins were significantly elevated in TBI individuals. The receiver operating characteristics (ROC) were determined to obtain the area under the curve (AUC) to establish the potential fit as a biomarker. Potential biomarkers were then compared to documented patient Glasgow coma scale scores via a correlation matrix and a multivariate linear regression to determine how respective biomarkers are related to the injury severity and pathological outcome. Inflammasome proteins and inflammatory cytokines were elevated after TBI, and the apoptosis-associated speck like protein containing a caspase recruitment domain (ASC), interleukin (IL)-18, tumor necrosis factor (TNF)-α, IL-4 and IL-6 were the most reliable biomarkers. Additionally, levels of these proteins were correlated with known clinical indicators of pathological outcome, such as the Glasgow coma scale (GCS). Our results show that inflammatory cytokines and inflammasome proteins are promising biomarkers for determining pathological outcomes after TBI. Additionally, levels of biomarkers could potentially be utilized to determine a patient’s injury severity and subsequent pathological outcome. These findings show that inflammation-associated proteins in the blood are reliable biomarkers of injury severity that can also be used to assess the functional outcomes of TBI patients.
Collapse
Affiliation(s)
- Nathan H. Johnson
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (N.H.J.); (R.H.); (R.W.K.)
| | - Roey Hadad
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (N.H.J.); (R.H.); (R.W.K.)
| | - Ruby Rose Taylor
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (R.R.T.); (W.D.D.)
| | - Javier Rodríguez Pilar
- Intensive Care Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain; (J.R.P.); (O.S.); (J.A.L.-P.); (J.P.-B.)
| | - Osman Salazar
- Intensive Care Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain; (J.R.P.); (O.S.); (J.A.L.-P.); (J.P.-B.)
| | - Juan Antonio Llompart-Pou
- Intensive Care Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain; (J.R.P.); (O.S.); (J.A.L.-P.); (J.P.-B.)
| | - W. Dalton Dietrich
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (R.R.T.); (W.D.D.)
| | - Robert W. Keane
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (N.H.J.); (R.H.); (R.W.K.)
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (R.R.T.); (W.D.D.)
| | - Jon Pérez-Bárcena
- Intensive Care Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain; (J.R.P.); (O.S.); (J.A.L.-P.); (J.P.-B.)
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (R.R.T.); (W.D.D.)
- Correspondence:
| |
Collapse
|
31
|
Cyr B, Hadad R, Keane RW, de Rivero Vaccari JP. The Role of Non-canonical and Canonical Inflammasomes in Inflammaging. Front Mol Neurosci 2022; 15:774014. [PMID: 35221912 PMCID: PMC8864077 DOI: 10.3389/fnmol.2022.774014] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 01/17/2022] [Indexed: 01/11/2023] Open
Abstract
Neurodegenerative diseases currently affect millions of people worldwide and continues to increase in the expanding elderly population. Neurodegenerative diseases usually involve cognitive decline and are among the top causes of death. Thus, there is a critical need for the development of treatments and preventive strategies for neurodegenerative diseases. One of the risk factors of neurodegeneration is inflammaging, a low level of chronic inflammation due to old age. We have previously shown that the inflammasome contributes to inflammaging in the central nervous system (CNS). The inflammasome is a multiprotein complex of the innate immune response consisting of a sensor protein, apoptosis speck-like protein containing a CARD (ASC), and caspase-1. Our lab has developed a humanized monoclonal antibody against ASC (anti-ASC). Here, we analyzed cortical lysates from young (3 months old), aged (18 months old), and aged anti-ASC treated mice for the expression of canonical and non-canonical inflammasome proteins. We show that the protein levels of NLRP1, ASC, caspase-1, and caspase-8 were elevated in the cortex of aged mice, and that anti-ASC decreased the expression of these proteins, consistent with lower levels of the pro-inflammatory cytokine interleukin (IL)-1β. Additionally, we show that these proteins form a novel NLRP1-caspase-8 non-canonical inflammasome comprised of NLRP1, caspase-8 and ASC. Moreover, these inflammasome proteins were present in neurons in young and aged mice. Together, these results indicate that a novel NLRP1-caspase-8 non-canonical inflammasome is present in the cortex of mice and that anti-ASC is a potential therapeutic to decrease inflammasome-mediated inflammaging in the CNS.
Collapse
Affiliation(s)
- Brianna Cyr
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Roey Hadad
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Robert W. Keane
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
- Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, United States
- *Correspondence: Juan Pablo de Rivero Vaccari,
| |
Collapse
|
32
|
Mechanism of Caspase-1 Inhibition by Four Anti-inflammatory Drugs Used in COVID-19 Treatment. Int J Mol Sci 2022; 23:ijms23031849. [PMID: 35163769 PMCID: PMC8837144 DOI: 10.3390/ijms23031849] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 01/27/2023] Open
Abstract
The inflammatory protease caspase-1 is associated with the release of cytokines. An excessive number of cytokines (a “cytokine storm”) is a dangerous consequence of COVID-19 infection and has been indicated as being among the causes of death by COVID-19. The anti-inflammatory drug colchicine (which is reported in the literature to be a caspase-1 inhibitor) and the corticosteroid drugs, dexamethasone and methylprednisolone, are among the most effective active compounds for COVID-19 treatment. The SERM raloxifene has also been used as a repurposed drug in COVID-19 therapy. In this study, inhibition of caspase-1 by these four compounds was analyzed using computational methods. Our aim was to see if the inhibition of caspase-1, an important biomolecule in the inflammatory response that triggers cytokine release, could shed light on how these drugs help to alleviate excessive cytokine production. We also measured the antioxidant activities of dexamethasone and colchicine when scavenging the superoxide radical using cyclic voltammetry methods. The experimental findings are associated with caspase-1 active site affinity towards these compounds. In evaluating our computational and experimental results, we here formulate a mechanism for caspase-1 inhibition by these drugs, which involves the active site amino acid Cys285 residue and is mediated by a transfer of protons, involving His237 and Ser339. It is proposed that the molecular moiety targeted by all of these drugs is a carbonyl group which establishes a S(Cys285)–C(carbonyl) covalent bond.
Collapse
|
33
|
Ulgheri F, Spanu P, Deligia F, Loriga G, Fuggetta MP, de Haan I, Chandgudge A, Groves M, Domling A. Design, synthesis and biological evaluation of 1,5-disubstituted α-amino tetrazole derivatives as non-covalent inflammasome-caspase-1 complex inhibitors with potential application against immune and inflammatory disorders. Eur J Med Chem 2022; 229:114002. [PMID: 34823899 PMCID: PMC8598261 DOI: 10.1016/j.ejmech.2021.114002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/05/2021] [Accepted: 11/14/2021] [Indexed: 02/08/2023]
Abstract
Compounds targeting the inflammasome-caspase-1 pathway could be of use for the treatment of inflammation and inflammatory diseases. Previous caspase-1 inhibitors were in great majority covalent inhibitors and failed in clinical trials. Using a mixed modelling, computational screening, synthesis and in vitro testing approach, we identified a novel class of non-covalent caspase-1 non cytotoxic inhibitors which are able to inhibit IL-1β release in activated macrophages in the low μM range, in line with the best activities observed for the known covalent inhibitors. Our compounds could form the basis of further optimization towards potent drugs for the treatment of inflammation and inflammatory disorders including also dysregulated inflammation in Covid 19.
Collapse
Affiliation(s)
- Fausta Ulgheri
- Institute of Biomolecular Chemistry, National Research Council (CNR), Trav. La Crucca 3, 07100, Sassari, Italy,Corresponding author
| | - Pietro Spanu
- Institute of Biomolecular Chemistry, National Research Council (CNR), Trav. La Crucca 3, 07100, Sassari, Italy,Corresponding author
| | - Francesco Deligia
- Institute of Biomolecular Chemistry, National Research Council (CNR), Trav. La Crucca 3, 07100, Sassari, Italy
| | - Giovanni Loriga
- Institute of Biomolecular Chemistry, National Research Council (CNR), Trav. La Crucca 3, 07100, Sassari, Italy
| | - Maria Pia Fuggetta
- Institute of Traslational Pharmacology, National Research Council (CNR), Via Fosso Del Cavaliere 100, 00133, Roma, Italy
| | - Iris de Haan
- Department of Drug Design, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Ajay Chandgudge
- Department of Drug Design, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Matthew Groves
- Department of Drug Design, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Alexander Domling
- Department of Drug Design, University of Groningen, 9713 AV Groningen, the Netherlands,Corresponding author
| |
Collapse
|
34
|
Baazm M, Behrens V, Beyer C, Nikoubashman O, Zendedel A. Regulation of Inflammasomes by Application of Omega-3 Polyunsaturated Fatty Acids in a Spinal Cord Injury Model. Cells 2021; 10:3147. [PMID: 34831370 PMCID: PMC8618254 DOI: 10.3390/cells10113147] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 11/17/2022] Open
Abstract
Omega-3 polyunsaturated fatty acids (PUFA n3) ameliorate inflammation in different diseases and potentially improve neurological function after neuronal injury. Following spinal cord injury (SCI), inflammatory events result in caspase-1 mediated activation of interleukin-1 beta (IL-1b) and 18. We aim to evaluate the neuroprotective potency of PUFA n3 in suppressing the formation and activation of inflammasomes following SCI. Male Wistar rats were divided into four groups: control, SCI, SCI+PUFA n3, and SCI+Lipofundin MCT (medium-chain triglyceride; vehicle). PUFA n3 or vehicle was intravenously administered immediately after SCI and every 24 h for the next three days. We analyzed the expression of NLRP3, NLRP1, ASC, caspase-1, IL-1b, and 18 in the spinal cord. The distribution of microglia, oligodendrocytes, and astrocytes was assessed by immunohistochemistry analysis. Behavioral testing showed significantly improved locomotor recovery in PUFA n3-treated animals and the SCI-induced upregulation of inflammasome components was reduced. Histopathological evaluation confirmed the suppression of microgliosis, increased numbers of oligodendrocytes, and the prevention of demyelination by PUFA n3. Our data support the neuroprotective role of PUFA n3 by targeting the NLRP3 inflammasome. These findings provide evidence that PUFA n3 has therapeutic effects which potentially attenuate neuronal damage in SCI and possibly also in other neuronal injuries.
Collapse
Affiliation(s)
- Maryam Baazm
- Department of Anatomy, School of Medicine, Arak University of Medical Sciences, Arak 3819693345, Iran;
| | - Victoria Behrens
- Institute of Neuroanatomy, Uniklinik RWTH Aachen, 52074 Aachen, Germany; (V.B.); (C.B.)
| | - Cordian Beyer
- Institute of Neuroanatomy, Uniklinik RWTH Aachen, 52074 Aachen, Germany; (V.B.); (C.B.)
| | - Omid Nikoubashman
- Department of Neuroradiology, University Hospital RWTH, 52074 Aachen, Germany;
| | - Adib Zendedel
- Institute of Neuroanatomy, Uniklinik RWTH Aachen, 52074 Aachen, Germany; (V.B.); (C.B.)
| |
Collapse
|
35
|
Erythropoietin Abrogates Post-Ischemic Activation of the NLRP3, NLRC4, and AIM2 Inflammasomes in Microglia/Macrophages in a TAK1-Dependent Manner. Transl Stroke Res 2021; 13:462-482. [PMID: 34628598 PMCID: PMC9046144 DOI: 10.1007/s12975-021-00948-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/18/2021] [Accepted: 09/18/2021] [Indexed: 12/18/2022]
Abstract
Inflammasomes are known to contribute to brain damage after acute ischemic stroke (AIS). TAK1 is predominantly expressed in microglial cells and can regulate the NLRP3 inflammasome, but its impact on other inflammasomes including NLRC4 and AIM2 after AIS remains elusive. EPO has been shown to reduce NLRP3 protein levels in different disease models. Whether EPO-mediated neuroprotection after AIS is conveyed via an EPO/TAK1/inflammasome axis in microglia remains to be clarified. Subjecting mice deficient for TAK1 in microglia/macrophages (Mi/MΦ) to AIS revealed a significant reduction in infarct sizes and neurological impairments compared to the corresponding controls. Post-ischemic increased activation of TAK1, NLRP3, NLRC4, and AIM2 inflammasomes including their associated downstream cascades were markedly reduced upon deletion of Mi/MΦ TAK1. EPO administration improved clinical outcomes and dampened stroke-induced activation of TAK1 and inflammasome cascades, which was not evident after the deletion of Mi/MΦ TAK1. Pharmacological inhibition of NLRP3 in microglial BV-2 cells did not influence post-OGD IL-1β levels, but increased NLRC4 and AIM2 protein levels, suggesting compensatory activities among inflammasomes. Overall, we provide evidence that Mi/MΦ TAK1 regulates the expression and activation of the NLRP3, NLRC4, AIM2 inflammasomes. Furthermore, EPO mitigated stroke-induced activation of TAK1 and inflammasomes, indicating that EPO conveyed neuroprotection might be mediated via an EPO/TAK1/inflammasome axis.
Collapse
|
36
|
Pérez-Bárcena J, Rodríguez Pilar J, Salazar O, Crespí C, Frontera G, Novo MA, Guardiola MB, Llompart-Pou JA, Ibáñez J, de Rivero Vaccari JP. Serum Caspase-1 as an Independent Prognostic Factor in Traumatic Brain Injured Patients. Neurocrit Care 2021; 36:527-535. [PMID: 34498205 DOI: 10.1007/s12028-021-01340-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 08/23/2021] [Indexed: 01/27/2023]
Abstract
BACKGROUND The objectives of this study were to assess the association between serum caspase 1 levels and known clinical and radiological prognostic factors and determine whether caspase 1was a more powerful predictor of outcome after traumatic brain injury (TBI) than clinical indices alone, to determine the association between the serum levels of caspase 1 and the 6-month outcome, and to evaluate if there is any association between caspase 1 with clinical and radiological variables. METHODS This prospective and observational study was conducted in a university hospital and included patients with TBI who required hospital admission. Serum samples were collected at hospital admission and 24 h after TBI. Caspase 1 levels were determined by enzyme-linked immunosorbent assay. Receiver operating characteristic curves were obtained to test the potential of caspase 1 to predict mortality (Glasgow Outcome Scale Extended score of 1) and unfavorable outcome (Glasgow Outcome Scale Extended scores of 1-4). Multivariate logistic regression was used to assess the effect of serum caspase 1 levels, adjusted by known clinical and radiological prognostic indices, on the outcome. RESULTS One hundred thirty-two patients and 33 healthy controls were included. We obtained 6-month outcome in 118 patients. On admission, the mean serum levels of caspase 1 were higher in patients with TBI compared with controls (157.9 vs. 108.5 pg/mL; p < 0.05) but not at 24 h after TBI. Serum caspase 1 levels on admission were higher in patients with unfavorable outcomes (189.5 vs. 144.1 pg/mL; p = 0.009). Similarly, serum caspase 1 levels on admission were higher in patients who died vs. patients who survived (213.6 vs. 146.8 pg/mL; p = 0.03). A logistic regression model showed that the serum caspase 1 level on admission was an independent predictor of 6-month unfavorable outcomes (odds ratio 1.05; 95% confidence interval 1-1.11; p = 0.05). Caspase 1 levels were higher in patients with severe TBI compared with those with moderate TBI, those with mild TBI, and healthy controls (p < 0.001). We did not find any correlation between caspase 1 and the radiological variables studied. CONCLUSIONS In this cohort of patients with TBI, we show that serum caspase 1 protein levels on admission are an independent prognostic factor after TBI. Serum caspase 1 levels on admission are higher in patients who will present unfavorable outcomes 6 months after TBI. Caspase 1 levels on admission are associated with the injury severity determined by the Glasgow Coma Scale.
Collapse
Affiliation(s)
- Jon Pérez-Bárcena
- Intensive Care Department, Son Espases University Hospital, Carretera de Valldemossa, 79, 07120, Palma de Mallorca, Islas Baleares, Spain.
| | - Javier Rodríguez Pilar
- Intensive Care Department, Son Espases University Hospital, Carretera de Valldemossa, 79, 07120, Palma de Mallorca, Islas Baleares, Spain
| | - Osman Salazar
- Department of Neurological Surgery, Son Espases University Hospital, Palma de Mallorca, Spain
| | - Catalina Crespí
- Fundación Instituto de Investigación Sanitaria Islas Baleares (IdISBa), Son Espases University Hospital, Palma de Mallorca, Spain
| | - Guillem Frontera
- Fundación Instituto de Investigación Sanitaria Islas Baleares (IdISBa), Son Espases University Hospital, Palma de Mallorca, Spain
| | - Mariana Andrea Novo
- Intensive Care Department, Son Espases University Hospital, Carretera de Valldemossa, 79, 07120, Palma de Mallorca, Islas Baleares, Spain
| | - María Begoña Guardiola
- Intensive Care Department, Son Espases University Hospital, Carretera de Valldemossa, 79, 07120, Palma de Mallorca, Islas Baleares, Spain
| | - Juan Antonio Llompart-Pou
- Intensive Care Department, Son Espases University Hospital, Carretera de Valldemossa, 79, 07120, Palma de Mallorca, Islas Baleares, Spain
| | - Javier Ibáñez
- Department of Neurological Surgery, Son Espases University Hospital, Palma de Mallorca, Spain
| | - Juan Pablo de Rivero Vaccari
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, USA
| |
Collapse
|
37
|
Zhang Y, Ding S, Chen Y, Sun Z, Zhang J, Han Y, Dong X, Fang Z, Li W. Ginsenoside Rg1 alleviates lipopolysaccharide-induced neuronal damage by inhibiting NLRP1 inflammasomes in HT22 cells. Exp Ther Med 2021; 22:782. [PMID: 34055081 PMCID: PMC8145787 DOI: 10.3892/etm.2021.10214] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
Lipopolysaccharide (LPS) is a toxic component of cell walls of Gram-negative bacteria that are widely present in gastrointestinal tracts. Increasing evidence showed that LPS plays important roles in the pathogeneses of neurodegenerative disorders, such as Alzheimer's disease (AD). NADPH oxidase s2 (NOX2) is a complex membrane protein that contributes to the production of reactive oxygen species (ROS) in several neurological diseases. The NLRP1 inflammasome can be activated in response to an accumulation of ROS in neurons. However, it is still unknown whether LPS exposure can deteriorate neuronal damage by activating NOX2-NLRP1 inflammasomes. Ginsenoside Rg1 (Rg1) has protective effects on neurons, although whether Rg1 alleviates LPS-induced neuronal damage by inhibiting NOX2-NLRP1 inflammasomes remains unclear. In the present study, the effect of concentration gradients and different times of LPS exposure on neuronal damage was investigated in HT22 cells, and further observed the effect of Rg1 treatment on NOX2-NLPR1 inflammasome activation, ROS production and neuronal damage in LPS-treated HT22 cells. The results demonstrated that LPS exposure significantly induced NOX2-NLRP1 inflammasome activation, excessive production of ROS, and neuronal damage in HT22 cells. It was also shown that Rg1 treatment significantly decreased NOX2-NLRP1 inflammasome activation and ROS production and alleviated neuronal damage in LPS-induced HT22 cells. The present data suggested that Rg1 has protective effects on LPS-induced neuronal damage by inhibiting NOX2-NLRP1 inflammasomes in HT22 cells, and Rg1 may be a potential therapeutic approach for delaying neuronal damage in AD.
Collapse
Affiliation(s)
- Yaodong Zhang
- Department of Pharmacy, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang 311200, P.R. China
| | - Shixin Ding
- Department of Pharmacology, Basic Medicine College, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yali Chen
- Department of Pharmacology, Basic Medicine College, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Zhenghao Sun
- Department of Pharmacology, Basic Medicine College, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Junyan Zhang
- Department of Pharmacology, Basic Medicine College, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yuli Han
- Department of Pharmacology, Basic Medicine College, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Xianan Dong
- Department of Pharmacology, Basic Medicine College, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Zhirui Fang
- Department of Pharmacology, Basic Medicine College, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Weizu Li
- Department of Pharmacology, Basic Medicine College, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
38
|
Pérez-Bárcena J, Crespí C, Frontera G, Llompart-Pou JA, Salazar O, Goliney V, Ibáñez J, Bullock MR, de Rivero Vaccari JP. Levels of caspase-1 in cerebrospinal fluid of patients with traumatic brain injury: correlation with intracranial pressure and outcome. J Neurosurg 2021; 134:1644-1649. [PMID: 32357337 DOI: 10.3171/2020.2.jns193079] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/24/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The objectives of this study were to evaluate levels of inflammasome-signaling proteins in serum and CSF of patients with traumatic brain injury (TBI), and to correlate these protein levels with intracranial pressure (ICP) and clinical outcomes at 6 months after injury. METHODS This is a prospective and observational study in patients with moderate and severe TBI who required an external ventricular drain as part of their treatment. Serum and CSF samples were collected 3 times a day for the first 5 days after TBI. The authors have determined the protein concentration of caspase-1 in the CSF and serum of patients with TBI by using commercially available enzyme-linked immunosorbent assays. The ICP value was recorded hourly. The 6-month outcome was assessed using the Glasgow Outcome Scale-Extended. RESULTS A total of 21 patients were included in this study, and a total of 234 paired serum-CSF samples were analyzed. The area under the curve (AUC) value of caspase-1 in CSF during the 5-day period was 2452.9 pg/mL·hr in the group of patients with high ICP vs 617.6 pg/mL·hr in the patients with low ICP. The differences were mainly on day 2 (19.7 pg/mL vs 1.8 pg/mL; p = 0.06) and day 3 (13.9 pg/mL vs 1 pg/mL; p = 0.05). The AUC value of caspase in CSF during the 5-day period was 1918.9 pg/mL·hr in the group of patients with poor outcome versus 924.5 pg/mL·hr in the patients with good outcome. The protein levels of caspase-1 in CSF were higher in patients with unfavorable outcomes during the first 96 hours after TBI. CONCLUSIONS In this cohort of patients with TBI who were admitted to the neurosurgical ICU, the inflammasome protein caspase-1 is increased in the CSF of patients with high ICP, especially on days 2 and 3 after TBI. Also the protein levels of caspase-1 in CSF were higher in patients with poor outcome during the first 96 hours after TBI. Moreover, not only the absolute value of caspase-1 in CSF but also its trend is associated with poor outcomes.
Collapse
Affiliation(s)
| | - Catalina Crespí
- 2Fundación Instituto de Investigación Sanitaria Islas Baleares (IdISBa), and
| | - Guillem Frontera
- 2Fundación Instituto de Investigación Sanitaria Islas Baleares (IdISBa), and
| | | | - Osman Salazar
- 3Department of Neurological Surgery, Son Espases University Hospital, Palma de Mallorca, Spain; and
| | - Victor Goliney
- 3Department of Neurological Surgery, Son Espases University Hospital, Palma de Mallorca, Spain; and
| | - Javier Ibáñez
- 3Department of Neurological Surgery, Son Espases University Hospital, Palma de Mallorca, Spain; and
| | - M Ross Bullock
- 4The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Juan Pablo de Rivero Vaccari
- 4The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
39
|
Cyr B, de Rivero Vaccari JP. Age-Dependent Microglial Response to Systemic Infection. Cells 2021; 10:cells10051037. [PMID: 33924771 PMCID: PMC8145069 DOI: 10.3390/cells10051037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammation is part of the aging process, and the inflammatory innate immune response is more exacerbated in older individuals when compared to younger individuals. Similarly, there is a difference in the response to systemic infection that varies with age. In a recent article by Hoogland et al., the authors studied the microglial response to systemic infection in young (2 months) and middle-aged mice (13–14 months) that were challenged with live Escherichia coli to investigate whether the pro- and anti-inflammatory responses mounted by microglia after systemic infection varies with age. Here, we comment on this study and its implications on how inflammation in the brain varies with age.
Collapse
Affiliation(s)
- Brianna Cyr
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- Center for Cognitive Neuroscience and Aging, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Correspondence:
| |
Collapse
|
40
|
Gomez A, Serrano A, Salero E, Tovar A, Amescua G, Galor A, Keane RW, de Rivero Vaccari JP, Sabater AL. Tumor necrosis factor-alpha and interferon-gamma induce inflammasome-mediated corneal endothelial cell death. Exp Eye Res 2021; 207:108574. [PMID: 33848524 DOI: 10.1016/j.exer.2021.108574] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/19/2021] [Accepted: 04/06/2021] [Indexed: 12/27/2022]
Abstract
PURPOSE Chronic corneal endothelial cell (CEC) loss results in corneal edema and vision loss in conditions such as pseudophakic bullous keratopathy (PBK), Fuchs' dystrophy, and corneal graft failure. Low CEC density has been associated with an elevation of intraocular pro-inflammatory cytokines such as tumor necrosis factor (TNF)-α and interferon (INF)-γ. These cytokines are capable of triggering pyroptosis, a programmed cell death mechanism mediated by the inflammasome, prompting the activation of the pro-inflammatory cytokine interleukin (IL)-1β, the perpetuation of inflammation, and subsequent damage of corneal endothelial tissue. Therefore, the purpose of this study was to determine the deleterious contribution of the inflammasome and pyroptosis to CEC loss. METHODS CECs from human donor corneas were treated ex vivo with TNF-α and IFN-γ for 48 h. Levels of caspase-1 and IL-1β were then assayed by ELISA, and the expression of caspase-1 and gasdermin-D (GSDM-D) were confirmed by immunofluorescence. Endothelial cell damage was analyzed by a lactate dehydrogenase (LDH) release assay, and oxidative stress was determined by measuring the levels of reactive oxygen species (ROS) in the culture media. RESULTS Inflammasome activation and oxidative stress were elevated in CECs following exposure to TNF-α and IFN-γ, which resulted in cell death by pyroptosis as determined by LDH release which was inhibited by the caspase-1 inhibitor Ac-YVAD-cmk. CONCLUSION CEC death is induced by the pro-inflammatory cytokines TNF-α and IFN-γ, which contribute to inflammasome activation. Moreover, the inflammasome is a promising therapeutic target for the treatment of chronic CEC loss.
Collapse
Affiliation(s)
- Angela Gomez
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA; Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andres Serrano
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA; Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Enrique Salero
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA; Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Arianna Tovar
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA; Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Guillermo Amescua
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Anat Galor
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Robert W Keane
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, FL, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, FL, USA
| | - Alfonso L Sabater
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA; Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
41
|
Kerr NA, de Rivero Vaccari JP, Weaver C, Dietrich WD, Ahmed T, Keane RW. Enoxaparin Attenuates Acute Lung Injury and Inflammasome Activation after Traumatic Brain Injury. J Neurotrauma 2021; 38:646-654. [PMID: 32669032 PMCID: PMC7898405 DOI: 10.1089/neu.2020.7257] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Traumatic brain injury (TBI) patients frequently develop cardiopulmonary system complications such as acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). However, the mechanism by which TBI causes ALI/ARDS is not fully understood. Here, we used a severe TBI model to examine the effects of a low-molecular-weight heparin, enoxaparin, on inflammasome activation and lung injury damage. We investigated whether enoxaparin inhibits ALI and inflammasome signaling protein expression in the brain and lungs after TBI in mice. C57/BL6 mice were subjected to severe TBI and were treated with vehicle or 1 mg/kg of enoxaparin 30 min after injury. Lung and brain tissue were collected 24 h post-TBI and were analyzed by immunoblotting for expression of the inflammasome proteins, caspase-1 and interleukin (IL)-1β. In addition, lung tissue was collected for histological analysis to determine ALI scoring and neutrophil and macrophage infiltration post-injury. Our data show that severe TBI induces increased expression of inflammasome proteins caspase-1 and IL-1β in the brain and lungs of mice after injury. Treatment with enoxaparin attenuated inflammasome expression in the brain and lungs 24 h after injury. Enoxaparin significantly decreased ALI score as well as neutrophil and macrophage infiltration in lungs at 24 h after injury. This study demonstrates that enoxaparin attenuates ALI and inhibits inflammasome expression in the brain and lungs after TBI. These findings support the hypothesis that inhibition of the neural-respiratory inflammasome axis that is activated after TBI may have therapeutic potential.
Collapse
Affiliation(s)
- Nadine A. Kerr
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Cailey Weaver
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - W. Dalton Dietrich
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Tahir Ahmed
- Pulmonary Division, Mount Sinai Medical Center, Miami Beach, Florida, USA
| | - Robert W. Keane
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
42
|
Chen SH, Scott XO, Ferrer Marcelo Y, Almeida VW, Blackwelder PL, Yavagal DR, Peterson EC, Starke RM, Dietrich WD, Keane RW, de Rivero Vaccari JP. Netosis and Inflammasomes in Large Vessel Occlusion Thrombi. Front Pharmacol 2021; 11:607287. [PMID: 33569001 PMCID: PMC7868597 DOI: 10.3389/fphar.2020.607287] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/18/2020] [Indexed: 12/20/2022] Open
Abstract
The inflammatory response appears to play a critical role in clotting in which neutrophil extracellular traps (NETs) are the major drivers of thrombosis in acute ischemic stroke (AIS). The inflammasome is an innate immune complex involved in the activation of interleukin (IL)-18 and IL-1β through caspase-1, but whether the inflammasome plays a role in NETosis in AIS remains poorly understood. Here we assessed the levels of inflammasome signaling proteins in NETs and their association with clinical and procedural outcomes of mechanical thrombectomy for AIS. Electron microscopy and immunofluorescence indicate the presence of NETs in thrombi of patients with AIS. Moreover, the inflammasome signaling proteins caspase-1 and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) were also present in clots associated with the marker of NETosis citrullinated histone 3H (CitH3). Analysis of protein levels by a simple plex assay show that caspase-1, ASC and interleukin (IL)-1β were significantly elevated in clots when compared to plasma of AIS patients and healthy controls, while IL-18 levels were lower. Moreover, multivariate analyses show that IL-1β levels in clots contribute to the number of passes to achieve complete recanalization, and that ASC, caspase-1 and IL-18 are significant contributors to time to recanalization. Thus, inflammasome proteins are elevated in NETs present in thrombi of patients with AIS that contribute to poor outcomes following stroke.
Collapse
Affiliation(s)
- Stephanie H Chen
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Xavier O Scott
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Yoandy Ferrer Marcelo
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Vania W Almeida
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Patricia L Blackwelder
- University of Miami Center for Advanced Microscopy (UMCAM) and Department of Chemistry, University of Miami, Coral Gables, FL, United States
| | - Dileep R Yavagal
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Eric C Peterson
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Robert M Starke
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - W Dalton Dietrich
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Robert W Keane
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States.,Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
43
|
de Rivero Vaccari JC, Dietrich WD, Keane RW, de Rivero Vaccari JP. The Inflammasome in Times of COVID-19. Front Immunol 2020; 11:583373. [PMID: 33149733 PMCID: PMC7580384 DOI: 10.3389/fimmu.2020.583373] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/07/2020] [Indexed: 12/15/2022] Open
Abstract
Coronaviruses (CoVs) are members of the genus Betacoronavirus and the Coronaviridiae family responsible for infections such as severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and more recently, coronavirus disease-2019 (COVID-19). CoV infections present mainly as respiratory infections that lead to acute respiratory distress syndrome (ARDS). However, CoVs, such as COVID-19, also present as a hyperactivation of the inflammatory response that results in increased production of inflammatory cytokines such as interleukin (IL)-1β and its downstream molecule IL-6. The inflammasome is a multiprotein complex involved in the activation of caspase-1 that leads to the activation of IL-1β in a variety of diseases and infections such as CoV infection and in different tissues such as lungs, brain, intestines and kidneys, all of which have been shown to be affected in COVID-19 patients. Here we review the literature regarding the mechanism of inflammasome activation by CoV infection, the role of the inflammasome in ARDS, ventilator-induced lung injury (VILI), and Disseminated Intravascular Coagulation (DIC) as well as the potential mechanism by which the inflammasome may contribute to the damaging effects of inflammation in the cardiac, renal, digestive, and nervous systems in COVID-19 patients.
Collapse
Affiliation(s)
| | - W Dalton Dietrich
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Robert W Keane
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States.,Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
44
|
Chen YQ, Wang SN, Shi YJ, Chen J, Ding SQ, Tang J, Shen L, Wang R, Ding H, Hu JG, Lü HZ. CRID3, a blocker of apoptosis associated speck like protein containing a card, ameliorates murine spinal cord injury by improving local immune microenvironment. J Neuroinflammation 2020; 17:255. [PMID: 32861243 PMCID: PMC7456508 DOI: 10.1186/s12974-020-01937-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 08/20/2020] [Indexed: 01/25/2023] Open
Abstract
Background After spinal cord injury (SCI), destructive immune cell subsets are dominant in the local microenvironment, which are the important mechanism of injury. Studies have shown that inflammasomes play an important role in the inflammation following SCI, and apoptosis-associated speck-like protein containing a card (ASC) is the adaptor protein shared by inflammasomes. Therefore, we speculated that inhibiting ASC may improve the local microenvironment of injured spinal cord. Here, CRID3, a blocker of ASC oligomerization, was used to study its effect on the local microenvironment and the possible role in neuroprotection following SCI. Methods Murine SCI model was created using an Infinite Horizon impactor at T9 vertebral level with a force of 50 kdynes and CRID3 (50 mg/kg) was intraperitoneally injected following injury. ASC and its downstream molecules in inflammasome signaling pathway were measured by western blot. The immune cell subsets were detected by immunohistofluorescence (IHF) and flow cytometry (FCM). The spinal cord fibrosis area, neuron survival, myelin preservation, and functional recovery were assessed. Results Following SCI, CRID3 administration inhibited inflammasome-related ASC and caspase-1, IL-1β, and IL-18 activation, which consequently suppressed M1 microglia, Th1 and Th1Th17 differentiation, and increased M2 microglia and Th2 differentiation. Accordingly, the improved histology and behavior have also been found. Conclusions CRID3 may ameliorate murine SCI by inhibiting inflammasome activation, reducing proinflammatory factor production, restoring immune cell subset balance, and improving local immune microenvironment, and early administration may be a promising therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Yu-Qing Chen
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, 233004, Bengbu, Anhui, People's Republic of China.,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Bengbu, 233004, Anhui, People's Republic of China.,Department of Immunology, Bengbu Medical College, 233030, Bengbu, Anhui, People's Republic of China.,Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, 233030, Bengbu, Anhui, People's Republic of China
| | - Sai-Nan Wang
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, 233004, Bengbu, Anhui, People's Republic of China.,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Bengbu, 233004, Anhui, People's Republic of China.,Department of Immunology, Bengbu Medical College, 233030, Bengbu, Anhui, People's Republic of China.,Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, 233030, Bengbu, Anhui, People's Republic of China
| | - Yu-Jiao Shi
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, 233004, Bengbu, Anhui, People's Republic of China.,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Bengbu, 233004, Anhui, People's Republic of China
| | - Jing Chen
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, 233004, Bengbu, Anhui, People's Republic of China.,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Bengbu, 233004, Anhui, People's Republic of China.,Department of Immunology, Bengbu Medical College, 233030, Bengbu, Anhui, People's Republic of China.,Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, 233030, Bengbu, Anhui, People's Republic of China
| | - Shu-Qin Ding
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, 233004, Bengbu, Anhui, People's Republic of China
| | - Jie Tang
- Department of Immunology, Bengbu Medical College, 233030, Bengbu, Anhui, People's Republic of China.,Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, 233030, Bengbu, Anhui, People's Republic of China
| | - Lin Shen
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Bengbu, 233004, Anhui, People's Republic of China
| | - Rui Wang
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Bengbu, 233004, Anhui, People's Republic of China
| | - Hai Ding
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Bengbu, 233004, Anhui, People's Republic of China
| | - Jian-Guo Hu
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, 233004, Bengbu, Anhui, People's Republic of China. .,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Bengbu, 233004, Anhui, People's Republic of China.
| | - He-Zuo Lü
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, 233004, Bengbu, Anhui, People's Republic of China. .,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Bengbu, 233004, Anhui, People's Republic of China. .,Department of Immunology, Bengbu Medical College, 233030, Bengbu, Anhui, People's Republic of China. .,Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, 233030, Bengbu, Anhui, People's Republic of China.
| |
Collapse
|
45
|
Noori L, Arabzadeh S, Mohamadi Y, Mojaverrostami S, Mokhtari T, Akbari M, Hassanzadeh G. Intrathecal administration of the extracellular vesicles derived from human Wharton's jelly stem cells inhibit inflammation and attenuate the activity of inflammasome complexes after spinal cord injury in rats. Neurosci Res 2020; 170:87-98. [PMID: 32717259 DOI: 10.1016/j.neures.2020.07.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 07/10/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023]
Abstract
Activation of inflammasome complexes during spinal cord injury (SCI) lead to conversion of pro-inflammatory cytokines, interleukin-1beta (IL-1β) and interleukin-18 (IL-18) to their active form to initiates the neuroinflammation. Mesenchymal stem cells (MSCs) showed anti-inflammatory properties through their extracellular vehicles (EVs). We investigated immunomodulatory potential of human Wharton's jelly mesenchymal stem cells derived extracellular vesicles (hWJ-MSC-EVs) on inflammasome activity one week after SCI in rats. The gene expression and protein level of IL-1β, IL-18, tumor necrosis factor alpha (TNF-α) and caspase1, were assessed by QPCR and western blotting. Immunohistochemistry (IHC) was done to measure the glial fibrillary acidic protein (GFAP) and Nestin expression. Cell death, histological evaluation and hind limb locomotion was studied by TUNEL assay, Nissl staining and Basso, Beattie, Bresnaham (BBB), respectively. Our finding represented that intrathecally administrated of hWJ-MSC-EVs significantly attenuated expression of the examined factors in both mRNA (P < 0.05 and P ≤ 0.01) and protein levels (P < 0.05 and P ≤ 0.01), decreased GFAP and increased Nestin expression (P < 0.05), reduced cell death and revealed the higher number of typical neurons in ventral horn of spinal cord. Consequently, progress in locomotion. We came to the conclusion that hWJ-MSC-EVs has the potential to control the inflammasome activity after SCI in rats. Moreover, EVs stimulated the neural progenitor cells and modulate the astrocyte activity.
Collapse
Affiliation(s)
- Leila Noori
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Arabzadeh
- Department of Biology, School of Basic Sciences, Ale Taha Institute of Higher Education, Tehran, Iran
| | - Yousef Mohamadi
- Department of Anatomy, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Sina Mojaverrostami
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Mokhtari
- CAS Key Laboratory of Mental Health, Institute of Psychology, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Mohammad Akbari
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Hassanzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Neuroscience and addiction studies, School of advanced technologies in medicine, Tehran University of Medical Sciences, Tehran, Iran; Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran.
| |
Collapse
|
46
|
The expressions of NLRP1, NLRP3, and AIM2 inflammasome complexes in the contusive spinal cord injury rat model and their responses to hormonal therapy. Cell Tissue Res 2020; 381:397-410. [DOI: 10.1007/s00441-020-03250-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
|
47
|
The Inflammasome Adaptor Protein ASC in Mild Cognitive Impairment and Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21134674. [PMID: 32630059 PMCID: PMC7370034 DOI: 10.3390/ijms21134674] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 12/23/2022] Open
Abstract
Mild cognitive impairment (MCI) is characterized by memory loss in the absence of dementia and is considered the translational stage between normal aging and early Alzheimer’s disease (AD). Patients with MCI have a greater risk of advancing to AD. Thus, identifying early markers of MCI has the potential to increase the therapeutic window to treat and manage the disease. Protein levels of the inflammasome signaling proteins apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and interleukin (IL)-18 were analyzed in the serum of patients with MCI, AD and healthy age-matched donors as possible biomarkers, as well as levels of soluble amyloid precursor proteins α/β (sAPP α/β) and neurofilament light (NfL). Cut-off points and positive and negative predictive values, as well as receiver operator characteristic (ROC) curves, likelihood ratios and accuracy were determined for these proteins. Although the levels of ASC were higher in MCI and AD than in age-matched controls, protein levels of ASC were higher in MCI than in AD cases. For control vs. MCI, the area under the curve (AUC) for ASC was 0.974, with a cut-off point of 264.9 pg/mL. These data were comparable to the AUC for sAPP α and β of 0.9687 and 0.9068, respectively, as well as 0.7734 for NfL. Moreover, similar results were obtained for control vs. AD and MCI vs. AD. These results indicate that ASC is a promising biomarker of MCI and AD.
Collapse
|
48
|
Chen Y, Ding S, Zhang H, Sun Z, Shen X, Sun L, Yin Y, Qun S, Li W. Protective effects of ginsenoside Rg1 on neuronal senescence due to inhibition of NOX2 and NLRP1 inflammasome activation in SAMP8 mice. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103713] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
49
|
Salmeron KE, Maniskas ME, Edwards DN, Wong R, Rajkovic I, Trout A, Rahman AA, Hamilton S, Fraser JF, Pinteaux E, Bix GJ. Interleukin 1 alpha administration is neuroprotective and neuro-restorative following experimental ischemic stroke. J Neuroinflammation 2019; 16:222. [PMID: 31727174 PMCID: PMC6857151 DOI: 10.1186/s12974-019-1599-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 09/24/2019] [Indexed: 01/06/2023] Open
Abstract
Background Stroke remains a leading cause of death and disability worldwide despite recent treatment breakthroughs. A primary event in stroke pathogenesis is the development of a potent and deleterious local and peripheral inflammatory response regulated by the pro-inflammatory cytokine interleukin-1 (IL-1). While the role of IL-1β (main released isoform) has been well studied in stroke, the role of the IL-1α isoform remains largely unknown. With increasing utilization of intravenous tissue plasminogen activator (t-PA) or thrombectomy to pharmacologically or mechanically remove ischemic stroke causing blood clots, respectively, there is interest in pairing successful cerebrovascular recanalization with neurotherapeutic pharmacological interventions (Fraser et al., J Cereb Blood Flow Metab 37:3531–3543, 2017; Hill et al., Lancet Neurol 11:942–950, 2012; Amaro et al., Stroke 47:2874–2876, 2016). Methods Transient stroke was induced in mice via one of two methods. One group of mice were subjected to tandem ipsilateral common carotid artery and middle cerebral artery occlusion, while another group underwent the filament-based middle cerebral artery occlusion. We have recently developed an animal model of intra-arterial (IA) drug administration after recanalization (Maniskas et al., J Neurosci Met 240:22–27, 2015). Sub groups of the mice were treated with either saline or Il-1α, wherein the drug was administered either acutely (immediately after surgery) or subacutely (on the third day after stroke). This was followed by behavioral and histological analyses. Results We now show in the above-mentioned mouse stroke models (transient tandem ipsilateral common carotid artery (CCA) and middle cerebral artery occlusion (MCA) occlusion, MCA suture occlusion) that IL-1α is neuroprotective when acutely given either intravenously (IV) or IA at low sub-pathologic doses. Furthermore, while IV administration induces transient hemodynamic side effects without affecting systemic markers of inflammation, IA delivery further improves overall outcomes while eliminating these side effects. Additionally, we show that delayed/subacute IV IL-1α administration ameliorates functional deficit and promotes neurorepair. Conclusions Taken together, our present study suggests for the first time that IL-1α could, unexpectedly, be an effective ischemic stroke therapy with a broad therapeutic window.
Collapse
Affiliation(s)
- Kathleen E Salmeron
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA.,Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA
| | - Michael E Maniskas
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA.,Department of Neurosurgery, University of Kentucky, Lexington, KY, 40536, USA.,Department of Neurology, University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Danielle N Edwards
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA.,Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA
| | - Raymond Wong
- Faculty of Biology, Medicine and Health, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Ivana Rajkovic
- Faculty of Biology, Medicine and Health, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Amanda Trout
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA.,Department of Neurology, University of Kentucky, Lexington, KY, 40536, USA.,Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, 40536, USA
| | - Abir A Rahman
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA.,Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, 40536, USA
| | - Samantha Hamilton
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Justin F Fraser
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA.,Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA.,Department of Neurosurgery, University of Kentucky, Lexington, KY, 40536, USA.,Department of Neurology, University of Kentucky, Lexington, KY, 40536, USA.,Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, 40536, USA
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, A.V. Hill Building, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Gregory J Bix
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA. .,Department of Neuroscience, University of Kentucky, Lexington, KY, 40536, USA. .,Department of Neurosurgery, University of Kentucky, Lexington, KY, 40536, USA. .,Department of Neurology, University of Kentucky, Lexington, KY, 40536, USA. .,Center for Advanced Translational Stroke Science, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
50
|
Ameliorative effects of echinacoside against spinal cord injury via inhibiting NLRP3 inflammasome signaling pathway. Life Sci 2019; 237:116978. [DOI: 10.1016/j.lfs.2019.116978] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/07/2019] [Accepted: 10/15/2019] [Indexed: 01/09/2023]
|