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
|
López-Hernández R, de la Torre-Álamo MM, García-Bueno B, Baroja-Mazo A, Fenoy FJ, Cuevas S. Inflammasomes in Alzheimer's Progression: Nrf2 as a Preventive Target. Antioxidants (Basel) 2025; 14:121. [PMID: 40002308 PMCID: PMC11851705 DOI: 10.3390/antiox14020121] [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: 12/19/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Current knowledge about Alzheimer's disease highlights the accumulation of β-amyloid plaques (Aβ1-42) and neurofibrillary tangles composed of hyperphosphorylated Tau, which lead to the loss of neuronal connections. Microglial activation and the release of inflammatory mediators play a significant role in the progression of Alzheimer's pathology. Recent advances have identified the involvement of inflammasomes, particularly NOD-like receptor NLR family pyrin domain containing 3 (NLRP3), whose activation promotes the release of proinflammatory cytokines and triggers pyroptosis, exacerbating neuroinflammation. Aggregates of Aβ1-42 and hyperphosphorylated Tau have been shown to activate these inflammasomes, while the apoptosis-associated speck-like protein (ASC) components form aggregates that further accelerate Aβ aggregation. Defects in the autophagic clearance of inflammasomes have also been implicated in Alzheimer's disease, contributing to sustained inflammation. This review explores strategies to counteract inflammation in Alzheimer's, emphasizing the degradation of ASC specks and the inhibition of NLRP3 inflammasome activation. Notably, the nuclear factor erythroid 2-related factor 2 (Nrf2) transcription factor emerges as a promising therapeutic target due to its dual role in mitigating oxidative stress and directly inhibiting NLRP3 inflammasome formation. By reducing inflammasome-driven inflammation, Nrf2 offers significant potential for addressing the neuroinflammatory aspects of Alzheimer's disease.
Collapse
Affiliation(s)
- Rubén López-Hernández
- Molecular Inflammation Group, Pathophysiology of the Inflammation and Oxidative Stress Lab, Biomedical Research Institute of Murcia (IMIB), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain;
| | - María Magdalena de la Torre-Álamo
- Molecular Inflammation Group, Digestive and Endocrine Surgery and Transplantation of Abdominal Organs, Biomedical Research Institute of Murcia (IMIB), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain; (M.M.d.l.T.-Á.); (B.G.-B.); (A.B.-M.)
| | - Belén García-Bueno
- Molecular Inflammation Group, Digestive and Endocrine Surgery and Transplantation of Abdominal Organs, Biomedical Research Institute of Murcia (IMIB), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain; (M.M.d.l.T.-Á.); (B.G.-B.); (A.B.-M.)
| | - Alberto Baroja-Mazo
- Molecular Inflammation Group, Digestive and Endocrine Surgery and Transplantation of Abdominal Organs, Biomedical Research Institute of Murcia (IMIB), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain; (M.M.d.l.T.-Á.); (B.G.-B.); (A.B.-M.)
| | - Francisco Jose Fenoy
- Department of Physiology, Faculty of Medicine, University of Murcia, 30120 Murcia, Spain;
| | - Santiago Cuevas
- Molecular Inflammation Group, Pathophysiology of the Inflammation and Oxidative Stress Lab, Biomedical Research Institute of Murcia (IMIB), University Clinical Hospital Virgen de la Arrixaca, 30120 Murcia, Spain;
| |
Collapse
|
3
|
Coll RC, Schroder K. Inflammasome components as new therapeutic targets in inflammatory disease. Nat Rev Immunol 2025; 25:22-41. [PMID: 39251813 DOI: 10.1038/s41577-024-01075-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2024] [Indexed: 09/11/2024]
Abstract
Inflammation drives pathology in many human diseases for which there are no disease-modifying drugs. Inflammasomes are signalling platforms that can induce pathological inflammation and tissue damage, having potential as an exciting new class of drug targets. Small-molecule inhibitors of the NLRP3 inflammasome that are now in clinical trials have demonstrated proof of concept that inflammasomes are druggable, and so drug development programmes are now focusing on other key inflammasome molecules. In this Review, we describe the potential of inflammasome components as candidate drug targets and the novel inflammasome inhibitors that are being developed. We discuss how the signalling biology of inflammasomes offers mechanistic insights for therapeutic targeting. We also discuss the major scientific and technical challenges associated with drugging these molecules during preclinical development and clinical trials.
Collapse
Affiliation(s)
- Rebecca C Coll
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK.
| | - Kate Schroder
- Institute for Molecular Bioscience (IMB), The University of Queensland, St Lucia, Queensland, Australia.
| |
Collapse
|
4
|
Scott XO, Kerr NA, Sanchez-Molano J, de Rivero Vaccari JP, Hadad R, De La Cruz A, Larsson HP, Dietrich WD, Keane RW. Catecholamine-Induced Inflammasome Activation in the Heart Following Photothrombotic Stroke. Transl Stroke Res 2024:10.1007/s12975-024-01311-3. [PMID: 39556309 DOI: 10.1007/s12975-024-01311-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/23/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
Cerebrovascular stroke patients exhibit an increased incidence of cardiac arrhythmias. The pathomechanisms underlying post-traumatic cardiac dysfunction include a surge of catecholamines and an increased systemic inflammatory response, but whether inflammasome activation contributes to cardiac dysfunction remains unexplored. Here, we used a mouse model of photothrombotic stroke (PTS) to investigate the role of inflammasome activation in post-stroke cardiac dysfunction by catecholamines and to evaluate the effectiveness of the inflammasome inhibitor IC100 on inflammasome activation. To evaluate functional electrophysiological changes in the heart by catecholamine treatment, we recorded action potential duration in excised zebrafish hearts with and without IC100 treatment. We show that PTS induced AIM2 inflammasome activation in atria and ventricles that was significantly reduced by administration of IC100. Injection of epinephrine into naïve mice induced a significant increase in AIM2, IL-1b and caspase-8 in atria. Treatment of excised zebrafish hearts with epinephrine shortened the action potential duration and this shortening that was reduced by IC100. These findings indicate that stroke initiates a catecholamine surge that induces inflammasome activation and pyroptosis in the heart that is blocked by IC100, thus providing a framework for the development of therapeutics for stroke-related cardiovascular injury.
Collapse
Affiliation(s)
- Xavier O Scott
- Department of Molecular Physiology and Cellular Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Nadine A Kerr
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, 33136, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juliana Sanchez-Molano
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, 33136, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Molecular Physiology and Cellular Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, 33136, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Roey Hadad
- Department of Molecular Physiology and Cellular Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, 33136, USA
| | - Alicia De La Cruz
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - H Peter Larsson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, 33136, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Robert W Keane
- Department of Molecular Physiology and Cellular Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA.
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL, 33136, USA.
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
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
|
C L B Ferreira B, Hannard M, Lozano-Garcia M, Aston L, Tejeda G, Domena JB, Bernard B, Chen J, Bartoli M, Rech Tondin A, Zhou Y, Scorzari A, Perrone CS, Tagliaferro A, Deo S, Daunert S, Dumont CM, Leblanc RM. Investigating the Significances of Thiol Functionalities in SARS-CoV-2 Using Carbon Dots for Viral Inhibition. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58439-58451. [PMID: 39422222 DOI: 10.1021/acsami.4c14482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
While the World Health Organization has declared the end of the SARS-CoV-2 public health emergency, studies related to corona viruses are still under course. As of 2024, the severity of COVID-19 has diminished with current treatments and vaccinations. However, individuals can still face severe complications, highlighting the importance of ongoing research into innovative treatments for current and future coronavirus-related diseases. This study approaches the mechanism of viral entrance into the host cells and the current evidence on the use of sulfhydryl groups for the COVID-19 treatment. Certain thiol drugs, a key contributor to inflammatory processes, exhibit both viral inhibition properties and the potential to regulate cellular oxidative stress by scavenging free radicals. Herein, we developed biocompatible thiol-functionalized carbon dots (CDs) and investigated the correlation between the number of thiols and pseudo-SARS-CoV-2 inhibition, reactive oxygen species (ROS) scavenging, and anti-inflammatory response. The free-radical scavenging experiment and the ROS cellular assay indicate that thiolated CDs serve as effective reducing agents and potential regulators of cellular oxidative stress. The CDs also demonstrated good cell viability alongside significant antiviral capabilities, with inhibition levels up to 60.4%. Furthermore, the flow cytometry results suggest that in an inflammatory environment, the presence of thiolated CDs promotes an anti-inflammatory response. Overall, the results demonstrate a strong correlation between the number of thiols and the increased efficacy observed across experiments, presenting thiolated CDs as promising candidates to prevent and treat COVID-19 infection.
Collapse
Affiliation(s)
- Braulio C L B Ferreira
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Maxence Hannard
- Empa - Swiss Federal Laboratories for Materials Science and Technology, Dübendorf 8600, Switzerland
| | - Mercedes Lozano-Garcia
- Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States
| | - Lillian Aston
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Giancarlo Tejeda
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Justin B Domena
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Brianna Bernard
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Jiuyan Chen
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Mattia Bartoli
- Center for Sustainable Future Technologies, Italian Institute of Technology, Via Livorno 60, Turin 10144, Italy
| | - Arthur Rech Tondin
- Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States
| | - Yiqun Zhou
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Annalise Scorzari
- Department of Chemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Caitlyn S Perrone
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Alberto Tagliaferro
- Department of Applied Science and Technology, Politecnico di Torino, Torino 10129 Italy
| | - Sapna Deo
- Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States
| | - Sylvia Daunert
- Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Roger M Leblanc
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| |
Collapse
|
7
|
Losa M, Emmenegger M, De Rossi P, Schürch PM, Serdiuk T, Pengo N, Capron D, Bieli D, Bargenda N, Rupp NJ, Carta MC, Frontzek KJ, Lysenko V, Reimann RR, Schwarz P, Nuvolone M, Westermark GT, Nilsson KPR, Polymenidou M, Theocharides AP, Hornemann S, Picotti P, Aguzzi A. The ASC inflammasome adapter governs SAA-derived protein aggregation in inflammatory amyloidosis. EMBO Mol Med 2024; 16:2024-2042. [PMID: 39080493 PMCID: PMC11393341 DOI: 10.1038/s44321-024-00107-0] [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: 05/28/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 09/14/2024] Open
Abstract
Extracellularly released molecular inflammasome assemblies -ASC specks- cross-seed Aβ amyloid in Alzheimer's disease. Here we show that ASC governs the extent of inflammation-induced amyloid A (AA) amyloidosis, a systemic disease caused by the aggregation and peripheral deposition of the acute-phase reactant serum amyloid A (SAA) in chronic inflammatory conditions. Using super-resolution microscopy, we found that ASC colocalized tightly with SAA in human AA amyloidosis. Recombinant ASC specks accelerated SAA fibril formation and mass spectrometry after limited proteolysis showed that ASC interacts with SAA via its pyrin domain (PYD). In a murine model of inflammatory AA amyloidosis, splenic amyloid load was conspicuously decreased in Pycard-/- mice which lack ASC. Treatment with anti-ASCPYD antibodies decreased amyloid loads in wild-type mice suffering from AA amyloidosis. The prevalence of natural anti-ASC IgG (-logEC50 ≥ 2) in 19,334 hospital patients was <0.01%, suggesting that anti-ASC antibody treatment modalities would not be confounded by natural autoimmunity. These findings expand the role played by ASC and IL-1 independent inflammasome employments to extraneural proteinopathies and suggest that anti-ASC immunotherapy may contribute to resolving such diseases.
Collapse
Affiliation(s)
- Marco Losa
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Marc Emmenegger
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Pierre De Rossi
- Department of Quantitative Biomedicine, University of Zürich, Zurich, Switzerland
| | - Patrick M Schürch
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Tetiana Serdiuk
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | | | | | | | - Niklas Bargenda
- Department of Quantitative Biomedicine, University of Zürich, Zurich, Switzerland
| | - Niels J Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Manfredi C Carta
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Karl J Frontzek
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Veronika Lysenko
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Regina R Reimann
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Petra Schwarz
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Mario Nuvolone
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
- Amyloidosis Research and Treatment Center, Fondazione Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Policlinico San Matteo, University of Pavia, Pavia, Italy
| | | | - K Peter R Nilsson
- Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | | | | | - Simone Hornemann
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Paola Picotti
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
8
|
Brint A, Greene S, Fennig-Victor AR, Wang S. Multiple sclerosis: the NLRP3 inflammasome, gasdermin D, and therapeutics. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:62. [PMID: 39118955 PMCID: PMC11304424 DOI: 10.21037/atm-23-1960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/26/2024] [Indexed: 08/10/2024]
Abstract
Multiple sclerosis (MS) stands as a chronic inflammatory disease characterized by its neurodegenerative impacts on the central nervous system. The complexity of MS and the significant challenges it poses to patients have made the exploration of effective treatments a crucial area of research. Among the various mechanisms under investigation, the role of inflammation in MS progression is of particular interest. Inflammatory responses within the body are regulated by various cellular mechanisms, one of which involves the nucleotide-binding oligomerization domain (NOD)-, leucine-rich repeat (LRR)-, and pyrin domains (PYD)-containing protein 3 (NLRP3). NLRP3 acts as a sensor within cells, playing a pivotal role in controlling the inflammatory response. Its activation is a critical step leading to the assembly of the NLRP3 inflammasome complex, a process that has profound implications for inflammatory diseases like MS. The NLRP3 inflammasome's activation is intricately linked to the subsequent activation of caspase 1 and gasdermin D (GsdmD), signaling pathways that are central to the inflammatory process. GsdmD, a prominent member of the Gasdermin protein family, is particularly noteworthy for its role in pyroptotic cell death, a form of programmed cell death that is distinct from apoptosis and is characterized by its inflammatory nature. This pathway's activation contributes significantly to the pathology of MS by exacerbating inflammatory responses within the nervous system. Given the detrimental effects of unregulated inflammation in MS, therapeutics targeting these inflammatory processes offer a promising avenue for alleviating the symptoms experienced by patients. This review delves into the intricacies of the pyroptotic pathways, highlighting how the formation of the NLRP3 inflammasome induces such pathways and the potential intervention points for therapeutic agents. By inhibiting key steps within these pathways, it is possible to mitigate the inflammatory response, thereby offering relief to those suffering from MS. Understanding these mechanisms not only sheds light on the pathophysiology of MS but also paves the way for the development of novel therapeutic strategies aimed at controlling the disease's progression through the modulation of the body's inflammatory response.
Collapse
Affiliation(s)
- Amie Brint
- Chemistry Department, University of Arkansas at Little Rock, Little Rock, AR, USA
- College of Medicine and Graduate School, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Seth Greene
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, USA
| | - Alyssa R. Fennig-Victor
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, USA
| | - Shanzhi Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, USA
| |
Collapse
|
9
|
Lu KP, Zhou XZ. Pin1-catalyzed conformational regulation after phosphorylation: A distinct checkpoint in cell signaling and drug discovery. Sci Signal 2024; 17:eadi8743. [PMID: 38889227 PMCID: PMC11409840 DOI: 10.1126/scisignal.adi8743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 05/30/2024] [Indexed: 06/20/2024]
Abstract
Protein phosphorylation is one of the most common mechanisms regulating cellular signaling pathways, and many kinases and phosphatases are proven drug targets. Upon phosphorylation, protein functions can be further regulated by the distinct isomerase Pin1 through cis-trans isomerization. Numerous protein targets and many important roles have now been elucidated for Pin1. However, no tools are available to detect or target cis and trans conformation events in cells. The development of Pin1 inhibitors and stereo- and phospho-specific antibodies has revealed that cis and trans conformations have distinct and often opposing cellular functions. Aberrant conformational changes due to the dysregulation of Pin1 can drive pathogenesis but can be effectively targeted in age-related diseases, including cancers and neurodegenerative disorders. Here, we review advances in understanding the roles of Pin1 signaling in health and disease and highlight conformational regulation as a distinct signal transduction checkpoint in disease development and treatment.
Collapse
Affiliation(s)
- Kun Ping Lu
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry
- Robarts Research Institute, Schulich School of Medicine & Dentistry
| | - Xiao Zhen Zhou
- Departments of Biochemistry and Oncology, Schulich School of Medicine & Dentistry
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine & Dentistry
- Lawson Health Research Institute, Western University, London, ON N6G 2V4, Canada
| |
Collapse
|
10
|
Vontell RT, Gober R, Dallmeier J, Brzostowicki D, Barreda A, Blennow K, Zetterberg H, Kvartsberg H, Gultekin SH, de Rivero Vaccari JP, Bramlett HM, Dietrich WD, Keane RW, Davis DA, Rundek T, Sun X. Association of region-specific hippocampal reduction of neurogranin with inflammasome proteins in post mortem brains of Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12444. [PMID: 38356472 PMCID: PMC10865487 DOI: 10.1002/trc2.12444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 02/16/2024]
Abstract
INTRODUCTION Neurogranin (Ng) is considered a biomarker for synaptic dysfunction in Alzheimer's disease (AD). In contrast, the inflammasome complex has been shown to exacerbate AD pathology. METHODS We investigated the protein expression, morphological differences of Ng, and correlated Ng to hyperphosphorylated tau in the post mortem brains of 17 AD cases and 17 age- and sex-matched controls. In addition, we correlated the Ng expression with two different epitopes of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC). RESULTS We show a reduction of Ng immunopositive neurons and morphological differences in AD compared to controls. Ng immunostaining was negatively correlated with neurofibrillary tangles, humanized anti-ASC (IC100) positive neurons and anti-ASC positive microglia, in AD. DISCUSSION The finding of a negative correlation between Ng and ASC speck protein expression in post mortem brains of AD suggests that the activation of inflammasome/ASC speck pathway may play an important role in synaptic degeneration in AD. Highlights We show the role that neurogranin plays on post-synaptic signaling in specific hippocampal regions.We demonstrate that there could be clinical implications of using neurogranin as a biomarker for dementia.We describe the loss of plasticity and neuronal scaffolding proteins in the present of AD pathology.We show the response of neuroinflammation when tau proteins phosphorylate in hippocampal neurons.We show that there is a potential therapeutic target for the inflammasome, and future studies may show that IC100, a humanized monoclonal antibody directed against ASC, may slow the progression of neurodegeneration.
Collapse
Affiliation(s)
- Regina T. Vontell
- Department of Neurology and Evelyn F. McKnight Brain InstituteUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Brain Endowment BankUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Ryan Gober
- Brain Endowment BankUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Julian Dallmeier
- Brain Endowment BankUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Daniel Brzostowicki
- Brain Endowment BankUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Ayled Barreda
- Brain Endowment BankUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalSahlgrenska University Hospital/Molndal V‐husetMolndalSweden
- Paris Brain InstituteICMPitié‐Salpêtrière HospitalSorbonne UniversityParisFrance
- Neurodegenerative Disorder Research CenterDivision of Life Sciences and Medicineand Department of NeurologyInstitute on Aging and Brain DisordersUniversity of Science and Technology of China and First Affiliated Hospital of USTCHefeiP.R. China
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalSahlgrenska University Hospital/Molndal V‐husetMolndalSweden
- Department of Neurodegenerative DiseaseUCL Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- Wisconsin Alzheimer's Disease Research CenterUniversity of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin–MadisonMadisonWisconsinUSA
| | - Hlin Kvartsberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgMölndalSweden
| | - Sakir Humayun Gultekin
- Brain Endowment BankUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Department of PathologyUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure ParalysisUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Department of Physiology and BiophysicsUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Center for Cognitive Neuroscience and AgingUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Helen M. Bramlett
- Department of Neurological Surgery and The Miami Project to Cure ParalysisUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Bruce W. Carter Department of Veterans Affairs Medical CenterMiamiFloridaUSA
| | - W. Dalton Dietrich
- Department of Neurological Surgery and The Miami Project to Cure ParalysisUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Robert W. Keane
- Department of Neurological Surgery and The Miami Project to Cure ParalysisUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Center for Cognitive Neuroscience and AgingUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - David A. Davis
- Department of Neurology and Evelyn F. McKnight Brain InstituteUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Brain Endowment BankUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Tatjana Rundek
- Department of Neurology and Evelyn F. McKnight Brain InstituteUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Xiaoyan Sun
- Department of Neurology and Evelyn F. McKnight Brain InstituteUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Brain Endowment BankUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| |
Collapse
|
11
|
Huang Y, Gao X, He QY, Liu W. A Interacting Model: How TRIM21 Orchestrates with Proteins in Intracellular Immunity. SMALL METHODS 2024; 8:e2301142. [PMID: 37922533 DOI: 10.1002/smtd.202301142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/12/2023] [Indexed: 11/07/2023]
Abstract
Tripartite motif-containing protein 21 (TRIM21), identified as both a cytosolic E3 ubiquitin ligase and FcR (Fragment crystallizable receptor), primarily interacts with proteins via its PRY/SPRY domains and promotes their proteasomal degradation to regulate intracellular immunity. But how TRIM21 involves in intracellular immunity still lacks systematical understanding. Herein, it is probed into the TRIM21-related literature and raises an interacting model about how TRIM21 orchestrates proteins in cytosol. In this novel model, TRIM21 generally interacts with miscellaneous protein in intracellular immunity in two ways: For one, TRIM21 solely plays as an E3, ubiquitylating a glut of proteins that contain specific interferon-regulatory factor, nuclear transcription factor kappaB, virus sensors and others, and involving inflammatory responses. For another, TRIM21 serves as both E3 and specific FcR that detects antibody-complexes and facilitates antibody destroying target proteins. Correspondingly delineated as Fc-independent signaling and Fc-dependent signaling in this review, how TRIM21's interactions contribute to intracellular immunity, expecting to provide a systematical understanding of this important protein and invest enlightenment for further research on the pathogenesis of related diseases and its prospective application is elaborated.
Collapse
Affiliation(s)
- Yisha Huang
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Xuejuan Gao
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Qing-Yu He
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Wanting Liu
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
12
|
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
|
13
|
Wang HY, Lin X, Huang GG, Zhou R, Lei SY, Ren J, Zhang KR, Feng CL, Wu YW, Tang W. Atranorin inhibits NLRP3 inflammasome activation by targeting ASC and protects NLRP3 inflammasome-driven diseases. Acta Pharmacol Sin 2023; 44:1687-1700. [PMID: 36964308 PMCID: PMC10374890 DOI: 10.1038/s41401-023-01054-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/09/2023] [Indexed: 03/26/2023]
Abstract
Aberrant NLRP3 activation has been implicated in the pathogenesis of numerous inflammation-associated diseases. However, no small molecular inhibitor that directly targets NLRP3 inflammasome has been approved so far. In this study, we show that Atranorin (C19H18O8), the secondary metabolites of lichen family, effectively prevents NLRP3 inflammasome activation in macrophages and dendritic cells. Mechanistically, Atranorin inhibits NLRP3 activation induced cytokine secretion and cell pyroptosis through binding to ASC protein directly and therefore restraining ASC oligomerization. The pharmacological effect of Atranorin is evaluated in NLRP3 inflammasome-driven disease models. Atranorin lowers serum IL-1β and IL-18 levels in LPS induced mice acute inflammation model. Also, Atranorin protects against MSU crystal induced mice gouty arthritis model and lowers ankle IL-1β level. Moreover, Atranorin ameliorates intestinal inflammation and epithelial barrier dysfunction in DSS induced mice ulcerative colitis and inhibits NLRP3 inflammasome activation in colon. Altogether, our study identifies Atranorin as a novel NLRP3 inhibitor that targets ASC protein and highlights the potential therapeutic effects of Atranorin in NLRP3 inflammasome-driven diseases including acute inflammation, gouty arthritis and ulcerative colitis.
Collapse
Affiliation(s)
- Hao-Yu Wang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xi Lin
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Guan-Gen Huang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rong Zhou
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shu-Yue Lei
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jing Ren
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Kai-Rong Zhang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science, Nanchang University, Nanchang, 330006, China
| | - Chun-Lan Feng
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yan-Wei Wu
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Medicine, Shanghai University, Shanghai, 200444, China.
| | - Wei Tang
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
14
|
Johnson NH, Kerr NA, de Rivero Vaccari JP, Bramlett HM, Keane RW, Dietrich WD. Genetic predisposition to Alzheimer's disease alters inflammasome activity after traumatic brain injury. Transl Res 2023; 257:66-77. [PMID: 36758791 PMCID: PMC10192027 DOI: 10.1016/j.trsl.2023.02.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 02/01/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023]
Abstract
Traumatic Brain Injury (TBI) is a major cause of death and disability in the US and a recognized risk factor for the development of Alzheimer's disease (AD). The relationship between these conditions is not completely understood, but the conditions may share additive or synergistic pathological hallmarks that may serve as novel therapeutic targets. Heightened inflammasome signaling plays a critical role in the pathogenesis of central nervous system injury (CNS) and the release of apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) speck from neurons and activated microglia contribute significantly to TBI and AD pathology. This study investigated whether inflammasome signaling after TBI was augmented in AD and whether this signaling pathway impacted biochemical and neuropathological outcomes and overall cognitive function. Five-month-old, 3xTg mice and respective wild type controls were randomized and underwent moderate controlled cortical impact (CCI) injury or served as sham/uninjured controls. Animals were sacrificed at 1 hour, 1 day, or 1 week after TBI to assess acute pathology or at 12 weeks after assessing cognitive function. The ipsilateral cerebral cortex was processed for inflammasome protein expression by immunoblotting. Mice were evaluated for behavior by open field (3 days), novel object recognition (2 weeks), and Morris water maze (6 weeks) testing after TBI. There was a statistically significant increase in the expression of inflammasome signaling proteins Caspase-1, Caspase-8, ASC, and interleukin (IL)-1β after TBI in both wild type and 3xTg animals. At 1-day post injury, significant increases in ASC and IL-1β protein expression were measured in AD TBI mice compared to WT TBI. Behavioral testing showed that injured AD mice had altered cognitive function when compared to injured WT mice. Elevated Aβ was seen in the ipsilateral cortex and hippocampus of sham and injured AD when compared to respective groups at 12 weeks post injury. Moreover, treatment of injured AD mice with IC100, an anti-ASC monoclonal antibody, inhibited the inflammasome, as evidenced by IL-1β reduction in the injured cortex at 1-week post injury. These findings show that the inflammasome response is heightened in mice genetically predisposed to AD and suggests that AD may exacerbate TBI pathology. Thus, dampening inflammasome signaling may offer a novel approach for the treatment of AD and TBI.
Collapse
Affiliation(s)
- Nathan H Johnson
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida
| | - Nadine A Kerr
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Juan P de Rivero Vaccari
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Helen M Bramlett
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida
| | - Robert W Keane
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida; Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida
| | - W Dalton Dietrich
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida.
| |
Collapse
|
15
|
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
|
16
|
McKee CG, Hoffos M, Vecchiarelli HA, Tremblay MÈ. Microglia: A pharmacological target for the treatment of age-related cognitive decline and Alzheimer's disease. Front Pharmacol 2023; 14:1125982. [PMID: 36969855 PMCID: PMC10034122 DOI: 10.3389/fphar.2023.1125982] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/03/2023] [Indexed: 03/29/2023] Open
Abstract
As individuals age, microglia, the resident immune cells of the central nervous system (CNS), become less effective at preserving brain circuits. Increases in microglial inflammatory activity are thought to contribute to age-related declines in cognitive functions and to transitions toward mild cognitive impairment (MCI) and Alzheimer's disease (AD). As microglia possess receptors for communicating with the CNS environment, pharmacological therapies targeting these pathways hold potential for promoting homeostatic microglial functions within the aging CNS. Preclinical and early phase clinical trials investigating the therapeutic effects of pharmacological agents acting on microglia, including reactive oxygen species, TREM2, fractalkine signaling, the complement cascade, and the NLRP3 inflammasome, are currently underway; however, important questions remain unanswered. Current challenges include target selectivity, as many of the signaling pathways are expressed in other cell types. Furthermore, microglia are a heterogenous cell population with transcriptomic, proteomic, and microscopy studies revealing distinct microglial states, whose activities and abundance shift across the lifespan. For example, homeostatic microglia can transform into pathological states characterized by markers of oxidative stress. Selective pharmacological targeting aimed at limiting transitions to pathological states or promoting homeostatic or protective states, could help to avoid potentially harmful off-target effects on beneficial states or other cell types. In this mini-review we cover current microglial pathways of interest for the prevention and treatment of age-related cognitive decline and CNS disorders of aging focusing on MCI and AD. We also discuss the heterogeneity of microglia described in these conditions and how pharmacological agents could target specific microglial states.
Collapse
Affiliation(s)
- Chloe G. McKee
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | - Madison Hoffos
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | | | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Département de Médecine Moléculaire, Université Laval, Québec City, QC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
- *Correspondence: Marie-Ève Tremblay,
| |
Collapse
|
17
|
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
|