1
|
Khodagholi F, Dezfouli MA, Yazdanfar N, Rashidi SK, Meymand AZ, Javadpour P, Mirbehbahani SH, Zare N. Prenatal Methamphetamine Exposure Impairs Helping Behaviour in Male Offspring: The Possible Role of miR-223 and NLRP3 Inflammasomes in the Amygdala. Int J Dev Neurosci 2025; 85:e10410. [PMID: 39723593 DOI: 10.1002/jdn.10410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 11/16/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
The increasing prevalence of methamphetamine abuse among women, particularly pregnant females, is a global concern. Methamphetamine can readily cross anatomical barriers like the blood-placenta barrier and cause detrimental impacts on the growing fetus. The current research evaluated the effects of prenatal methamphetamine exposure on helping behaviour and neuroinflammatory cascade in the amygdala of male offspring. On the tenth day of pregnancy, female rats received either saline or methamphetamine (5 mg/kg) until delivery. Once the offspring reached 21 days of age, the male ones were sep arated from their mothers and housed with normal male rats. An empathy-like behaviour test, which measured helping behaviour towards the cage mate, was conducted. The expression levels of miR-223-3p, NLRP3, Caspase 1, and gasdermin D (GSDMD) were evaluated in the amygdala of male offspring. Moreover, interleukin-1β (IL-1β) protein level was measured. Findings of this study revealed that male offspring exposed to methamphetamine during pregnancy had impaired helping behaviour. At the molecular level, prenatal methamphetamine exposure decreased miR-223-3p and increased inflammasome signaling by raising the levels of NLRP3, caspase-1, and GSDMD along with IL-1β levels. These findings indicate that prenatal methamphetamine exposure impairs emotional behaviour and activates inflammasome pathway in the amygdala.
Collapse
Affiliation(s)
- Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mitra Ansari Dezfouli
- Department of Neurology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Neda Yazdanfar
- Department of Neurology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Khalil Rashidi
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Pegah Javadpour
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Nayereh Zare
- Department of Anatomical Sciences and Cognitive Neurosciences, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
2
|
Dai Y, Shao M, Li L, Li H, Lu T, Lyu F. Molecular characterization of PANoptosis-related genes as novel signatures for peripheral nerve injury based on time-series transcriptome sequencing. Gene 2025; 933:148995. [PMID: 39393431 DOI: 10.1016/j.gene.2024.148995] [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: 06/25/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/13/2024]
Abstract
Programmed cell death (PCD) pathways play pivotal roles in the development and progression of peripheral nerve injury (PNI). PANoptosis, as a novel form of PCD pathway with key features of pyroptosis, apoptosis and necroptosis, is implicated in the pathogenesis of multiple neurologic diseases. This study aimed to identify PANoptosis-related biomarkers and characterize their molecular roles and immune landscape in PNI. PANoptosis-related genes (PRGs) were retrieved from Reactome pathway database and previous literatures. Differentially expressed PANoptosis-related genes (DEPRGs) were identified based on a time-series transcriptome sequencing dataset. DEPRGs were predicted to be enriched in inflammatory response, inflammatory complex, PCD and NOD-like receptor signaling pathway through GO, KEGG, Reactome and GSEA analysis. Hub genes, including Ripk3, Pycard and Il18, were then recognized through PPI network and multiple algorithms. The molecular regulatory mechanisms of hub genes were elucidated by transcription factor network and competing endogenous RNA network. Moreover, the immune cell landscape of hub genes was analyzed. Eventually, the expression levels of hub genes were verified through external dataset and animal model. Ripk3, Pycard and Il18 were remarkably upregulated in PNI samples, which were in consistent with the results of bioinformatic analysis. This study uncovered the molecular characterization of PANoptosis-related genes in PNI and illustrated the novel PANoptosis biomarkers for PNI.
Collapse
Affiliation(s)
- Yuan Dai
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Minghao Shao
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Linli Li
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Hailong Li
- Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Tingwei Lu
- Center of Craniofacial Orthodontics, Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 210000, China.
| | - Feizhou Lyu
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai 200040, China; Department of Orthopedics, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China.
| |
Collapse
|
3
|
Fan H, Shen R, Yan J, Bai Y, Fu Q, Shi X, Du G, Wang D. Pyroptosis the Emerging Link Between Gut Microbiota and Multiple Sclerosis. Drug Des Devel Ther 2024; 18:6145-6164. [PMID: 39717200 PMCID: PMC11665440 DOI: 10.2147/dddt.s489454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/11/2024] [Indexed: 12/25/2024] Open
Abstract
This review elucidates the pivotal role of pyroptosis, triggered by gut microbiota, in the development of multiple sclerosis (MS), emphasizing its significance within the gut-brain axis. Our comprehensive analysis of recent literature reveals how dysbiosis in the gut microbiota of MS patients-characterized by reduced microbial diversity and shifts in bacterial populations-profoundly impacts immune regulation and the integrity of the central nervous system (CNS). Pyroptosis, an inflammatory form of programmed cell death, significantly exacerbates MS by promoting the release of inflammatory cytokines and causing substantial damage to CNS tissues. The gut microbiota facilitates this detrimental process through metabolites such as short-chain fatty acids and neuroactive compounds, or self-structural products like lipopolysaccharides (LPS), which modulate immune responses and influence neuronal survival. This review highlights the potential of modulating gut microbiota to regulate pyroptosis, thereby suggesting that targeting this pathway could be a promising therapeutic strategy to mitigate inflammatory responses and preserve neuronal integrity in patients with MS.
Collapse
Affiliation(s)
- Hua Fan
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| | - Ruile Shen
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| | - Junqiang Yan
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| | - Yongjie Bai
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| | - Qizhi Fu
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| | - Xiaofei Shi
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| | - Ganqin Du
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| | - Dongmei Wang
- The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471003, People’s Republic of China
| |
Collapse
|
4
|
Otálora-Alcaraz A, Reilly T, Oró-Nolla M, Sun MC, Costelloe L, Kearney H, Patra PH, Downer EJ. The NLRP3 inflammasome: A central player in multiple sclerosis. Biochem Pharmacol 2024; 232:116667. [PMID: 39647604 DOI: 10.1016/j.bcp.2024.116667] [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: 09/13/2024] [Revised: 11/06/2024] [Accepted: 11/25/2024] [Indexed: 12/10/2024]
Abstract
Multiple sclerosis (MS) is a neurological autoimmune condition associated with many symptoms including spasticity, pain, limb numbness and weakness. It is characterised by inflammatory demyelination and axonal degeneration of the brain and spinal cord. A range of disease-modifying therapies (DMTs) are available to suppress inflammatory disease activity in MS, however, there is a pressing need for new therapeutic avenues as DMTs have a limited ability to suppress confirmed disability progression. A body of literature indicates that innate immune inflammation is linked to MS progression. The nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing protein 3 (NLRP3) inflammasome has a well-established function in innate immunity which is closely associated with the pathogenesis of neuroinflammatory conditions. Evidence suggests that the inflammasome may be a therapeutic target in disorders such as MS and at present, inhibitors of the NLRP3 inflammasome are in pre-clinical development. Therefore, this review systematically highlights the pathogenic role of inflammasomes in MS, presenting an overview of research evidence linking inflammasome-related polymorphisms to MS susceptibility, and gathering evidence investigating NLRP3 biomarkers in MS. The role of the NLRP3 inflammasome in murine models of MS is furthermore discussed. Finally, a significant component of this review focuses on evidence that NLRP3 signalling components are novel drug targets in MS. Overall this review defines the role of the inflammasome in MS pathogenesis and identifies inflammasome inhibitor targets that warrant full investigation in MS and related disorders.
Collapse
Affiliation(s)
- Almudena Otálora-Alcaraz
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Thomas Reilly
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Martí Oró-Nolla
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Melody Cui Sun
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Lisa Costelloe
- Department of Neurology, Beaumont Hospital, Dublin, Ireland
| | - Hugh Kearney
- MS Unit, Department of Neurology, St. James's Hospital, Dublin, Ireland; Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Ireland
| | - Pabitra H Patra
- Transpharmation Ltd., London Biosciences Innovation Centre, London, United Kingdom
| | - Eric J Downer
- Discipline of Physiology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
5
|
Fan H, Fu Q, Du G, Qin L, Shi X, Wang D, Yang Y. Microglial Mayhem NLRP3 Inflammasome's Role in Multiple Sclerosis Pathology. CNS Neurosci Ther 2024; 30:e70135. [PMID: 39690733 DOI: 10.1111/cns.70135] [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: 12/19/2023] [Revised: 09/29/2024] [Accepted: 11/05/2024] [Indexed: 12/19/2024] Open
Abstract
INTRODUCTION This review delves into the intricate relationship between NLR inflammasomes, particularly the NLRP3 inflammasome, and the immune-mediated neurodegenerative disease, multiple sclerosis (MS). While the precise etiology of MS remains elusive, compelling research underscores the pivotal role of the immune response in disease progression. Notably, recent investigations highlight the significant involvement of NLRP3 inflammasomes in various autoimmune diseases, prompting an in-depth exploration of their impact on MS. METHOD The review focuses on elucidating the activation mechanism of NLRP3 inflammasomes within microglia/macrophages (MG/MФ), examining how this activation promotes an inflammatory response that exacerbates neuronal damage in MS. A comprehensive analysis of existing literature and research findings forms the basis for understanding the intricate interplay between NLRP3 inflammasomes and MS pathogenesis. RESULTS Synthesizing current research, the review provides insight into the pivotal role played by NLR inflammasomes, specifically NLRP3, in MS. Emphasis is placed on the inflammatory response orchestrated by activated MG/MФ, elucidating the cascade that perpetuates neuronal damage in the disease. CONCLUSIONS This review concludes by consolidating key findings and offering a nuanced perspective on the role of NLRP3 inflammasomes in MS pathogenesis. The detailed exploration of the activation process within MG/MФ provides a foundation for understanding the disease's underlying mechanisms. Furthermore, the review sets the stage for potential therapeutic strategies targeting NLRP3 inflammasomes in the pursuit of MS treatment.
Collapse
Affiliation(s)
- Hua Fan
- Office of Research & Innovation, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Qizhi Fu
- Department of Intensive Medicine, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Ganqin Du
- Department of Neurology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Ling Qin
- Department of Hematology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Xiaofei Shi
- Department of Rheumatology and Immunology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Dongmei Wang
- School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Yanhui Yang
- Department of Emergency Medicine, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
6
|
Yang L, Xing W, Shi Y, Hu M, Li B, Hu Y, Zhang G. Stress-induced NLRP3 inflammasome activation and myelin alterations in the hippocampus of PTSD rats. Neuroscience 2024; 555:156-166. [PMID: 39043314 DOI: 10.1016/j.neuroscience.2024.07.028] [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: 01/31/2024] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024]
Abstract
Inflammatory and myelin changes may contribute to the pathophysiology of post-traumatic stress disorder (PTSD). The NOD-like receptor (NLR) family, pyrin domain-containing protein 3 (NLRP3), a brain inflammasome, is activated in the hippocampus of mice with PTSD. In other psychiatric disorders, NLRP3 expression has been associated with axonal myelination and demyelination. However, the association between NLRP3 and myelin in rats with PTSD remains unclear. Therefore, this study aims to investigate the relationship between the NLRP3 inflammasome and myelin in the hippocampus of rats with PTSD. A rat model of post-traumatic stress disorder was established using the single-prolonged stress (SPS) approach. Hippocampal tissues were collected for the detection of NLRP3 inflammasome-associated proteins and myelin basic protein at 3, 7, and 14 days after SPS. To further explore the relationship between NLRP3 and myelin, the NLRP3-specific inhibitor MCC950 was administered intraperitoneally to rats starting 72 h before SPS, and then alterations in NLRP3 inflammasome-associated proteins and myelin were observed in the PTSD and control groups. We found that NLRP3 and downstream related proteins were activated in the hippocampus of rats 3 days after SPS, and the myelin content in the hippocampus increased after SPS stress. MCC950 reduced the expression of NLRP3-related pathway proteins, improved anxiety behaviour and spatial learning memory impairment, and inhibited the increase in myelin content in the hippocampal region of rats after SPS. In conclusion the study indicates that NLRP3 has a significant role in the hippocampal region of rats with PTSD. Inhibition of the NLRP3 inflammasome could be a potential target for treating PTSD.
Collapse
Affiliation(s)
- Luodong Yang
- First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Wenlong Xing
- First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Yan Shi
- Shihezi University, Shihezi, China
| | - Min Hu
- First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Bin Li
- Shihezi University, Shihezi, China
| | - Yuanyuan Hu
- First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Guiqing Zhang
- First Affiliated Hospital of Shihezi University, Shihezi, China.
| |
Collapse
|
7
|
Hedayati N, Safaei Naeini M, Ale Sahebfosoul MM, Mafi A, Eshaghi Milasi Y, Rizaneh A, Nabavi N, Farahani N, Alimohammadi M, Ghezelbash B. MicroRNA dysregulation and its impact on apoptosis-related signaling pathways in myelodysplastic syndrome. Pathol Res Pract 2024; 261:155478. [PMID: 39079383 DOI: 10.1016/j.prp.2024.155478] [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: 04/12/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 08/18/2024]
Abstract
Myelodysplastic syndrome (MDS) holds a unique position among blood cancers, encompassing a spectrum of blood-related disorders marked by impaired maturation of blood cell precursors, bone marrow abnormalities, genetic instability, and a higher likelihood of progressing to acute myeloid leukemia. MicroRNAs (miRNAs), short non-coding RNA molecules typically 18-24 nucleotides in length, are known to regulate gene expression and contribute to various biological processes, including cellular differentiation and programmed cell death. Additionally, miRNAs are involved in many aspects of cancer development, influencing cell growth, transformation, and apoptosis. In this study, we explore the impact of microRNAs on cellular apoptosis in MDS.
Collapse
Affiliation(s)
- Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Mobina Safaei Naeini
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran; Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yaser Eshaghi Milasi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Anahita Rizaneh
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, Canada.
| | - Najma Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Behrooz Ghezelbash
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
8
|
Benoit RY, Zagrodnik JL, Carew SJ, Moore CS. Bruton Tyrosine Kinase Inhibition Decreases Inflammation and Differentially Impacts Phagocytosis and Cellular Metabolism in Mouse- and Human-derived Myeloid Cells. Immunohorizons 2024; 8:652-667. [PMID: 39259208 PMCID: PMC11447691 DOI: 10.4049/immunohorizons.2400045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 09/12/2024] Open
Abstract
Bruton tyrosine kinase (BTK) is a kinase expressed by various immune cells and is often activated under proinflammatory states. Although the majority of BTK-related research has historically focused on B cells, understanding the role of BTK in non-B cell populations is critical given myeloid cells also express BTK at comparable levels. In this study, we investigated and compared how BTK inhibition in human and murine myeloid cells alters cell phenotype and function. All experiments were performed using two BTK inhibitors (evobrutinib and tolebrutinib) that are currently in late-stage clinical trials for the treatment of multiple sclerosis. Assays were performed to assess the impact of BTK inhibition on cytokine and microRNA expression, phagocytic capacity, and cellular metabolism. In all cells, both evobrutinib and tolebrutinib significantly decreased phosphorylated BTK and LPS-induced cytokine release. BTK inhibition also significantly decreased the oxygen consumption rate and extracellular acidification rate in myeloid cells, and significantly decreased phagocytosis in murine-derived cells, but not human macrophages. To further elucidate the mechanism, we also investigated the expression of microRNAs known to impact the function of myeloid cells. BTK inhibition resulted in an altered microRNA expression profile (i.e., decreased miR-155-5p and increased miR-223-3p), which is consistent with a decreased proinflammatory myeloid cell phenotype. In summary, these results provide further insights into the mechanism of action of BTK inhibitors in the context of immune-related diseases, while also highlighting important species-specific and cell-specific differences that should be considered when interpreting and comparing results between preclinical and human studies.
Collapse
Affiliation(s)
- Rochelle Y. Benoit
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland and Labrador, Canada
| | - Jennifer L. Zagrodnik
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland and Labrador, Canada
| | - Samantha J. Carew
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland and Labrador, Canada
| | - Craig S. Moore
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland and Labrador, Canada
| |
Collapse
|
9
|
Gosetti di Sturmeck T, Malimpensa L, Ferrazzano G, Belvisi D, Leodori G, Lembo F, Brandi R, Pascale E, Cattaneo A, Salvetti M, Conte A, D’Onofrio M, Arisi I. Exploring miRNAs' Based Modeling Approach for Predicting PIRA in Multiple Sclerosis: A Comprehensive Analysis. Int J Mol Sci 2024; 25:6342. [PMID: 38928049 PMCID: PMC11203572 DOI: 10.3390/ijms25126342] [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: 05/08/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
The current hypothesis on the pathophysiology of multiple sclerosis (MS) suggests the involvement of both inflammatory and neurodegenerative mechanisms. Disease Modifying Therapies (DMTs) effectively decrease relapse rates, thus reducing relapse-associated disability in people with MS. In some patients, disability progression, however, is not solely linked to new lesions and clinical relapses but can manifest independently. Progression Independent of Relapse Activity (PIRA) significantly contributes to long-term disability, stressing the urge to unveil biomarkers to forecast disease progression. Twenty-five adult patients with relapsing-remitting multiple sclerosis (RRMS) were enrolled in a cohort study, according to the latest McDonald criteria, and tested before and after high-efficacy Disease Modifying Therapies (DMTs) (6-24 months). Through Agilent microarrays, we analyzed miRNA profiles from peripheral blood mononuclear cells. Multivariate logistic and linear models with interactions were generated. Robustness was assessed by randomization tests in R. A subset of miRNAs, correlated with PIRA, and the Expanded Disability Status Scale (EDSS), was selected. To refine the patient stratification connected to the disease trajectory, we computed a robust logistic classification model derived from baseline miRNA expression to predict PIRA status (AUC = 0.971). We built an optimal multilinear model by selecting four other miRNA predictors to describe EDSS changes compared to baseline. Multivariate modeling offers a promising avenue to uncover potential biomarkers essential for accurate prediction of disability progression in early MS stages. These models can provide valuable insights into developing personalized and effective treatment strategies.
Collapse
Affiliation(s)
- Tommaso Gosetti di Sturmeck
- European Brain Research Institute (EBRI) Rita Levi-Montalcini, 00161 Rome, Italy; (T.G.d.S.); (R.B.); (A.C.)
| | - Leonardo Malimpensa
- IRCCS Istituto Neurologico Mediterraneo Neuromed, 86077 Pozzilli, Italy; (L.M.); (D.B.); (G.L.); (M.S.); (A.C.)
| | - Gina Ferrazzano
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy; (G.F.); (F.L.)
| | - Daniele Belvisi
- IRCCS Istituto Neurologico Mediterraneo Neuromed, 86077 Pozzilli, Italy; (L.M.); (D.B.); (G.L.); (M.S.); (A.C.)
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy; (G.F.); (F.L.)
| | - Giorgio Leodori
- IRCCS Istituto Neurologico Mediterraneo Neuromed, 86077 Pozzilli, Italy; (L.M.); (D.B.); (G.L.); (M.S.); (A.C.)
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy; (G.F.); (F.L.)
| | - Flaminia Lembo
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy; (G.F.); (F.L.)
| | - Rossella Brandi
- European Brain Research Institute (EBRI) Rita Levi-Montalcini, 00161 Rome, Italy; (T.G.d.S.); (R.B.); (A.C.)
| | - Esterina Pascale
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy;
| | - Antonino Cattaneo
- European Brain Research Institute (EBRI) Rita Levi-Montalcini, 00161 Rome, Italy; (T.G.d.S.); (R.B.); (A.C.)
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore (SNS), 56126 Pisa, Italy
| | - Marco Salvetti
- IRCCS Istituto Neurologico Mediterraneo Neuromed, 86077 Pozzilli, Italy; (L.M.); (D.B.); (G.L.); (M.S.); (A.C.)
- Centre for Experimental Neurological Therapies (CENTERS), Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University of Rome, 00189 Rome, Italy
| | - Antonella Conte
- IRCCS Istituto Neurologico Mediterraneo Neuromed, 86077 Pozzilli, Italy; (L.M.); (D.B.); (G.L.); (M.S.); (A.C.)
- Department of Human Neurosciences, Sapienza University of Rome, 00185 Rome, Italy; (G.F.); (F.L.)
| | - Mara D’Onofrio
- European Brain Research Institute (EBRI) Rita Levi-Montalcini, 00161 Rome, Italy; (T.G.d.S.); (R.B.); (A.C.)
| | - Ivan Arisi
- European Brain Research Institute (EBRI) Rita Levi-Montalcini, 00161 Rome, Italy; (T.G.d.S.); (R.B.); (A.C.)
- Institute of Translational Pharmacology, National Research Council, 00133 Rome, Italy
| |
Collapse
|
10
|
Shadab A, Abbasi-Kolli M, Saharkhiz M, Ahadi SH, Shokouhi B, Nahand JS. The interplay between mitochondrial dysfunction and NLRP3 inflammasome in multiple sclerosis: Therapeutic implications and animal model studies. Biomed Pharmacother 2024; 175:116673. [PMID: 38713947 DOI: 10.1016/j.biopha.2024.116673] [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: 01/27/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/09/2024] Open
Abstract
Multiple sclerosis (MS) is a complex autoimmune disorder that impacts the central nervous system (CNS), resulting in inflammation, demyelination, and neurodegeneration. The NOD-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome, a multiprotein complex of the innate immune system, serves an essential role in the pathogenesis of MS by regulating the production of pro-inflammatory cytokines (IL-1β & IL-18) and the induction of pyroptotic cell death. Mitochondrial dysfunction is one of the main potential factors that can trigger NLRP3 inflammasome activation and lead to inflammation and axonal damage in MS. This highlights the importance of understanding how mitochondrial dynamics modulate NLRP3 inflammasome activity and contribute to the inflammatory and neurodegenerative features of MS. The lack of a comprehensive understanding of the pathogenesis of MS and the urge for the introduction of new therapeutic strategies led us to review the therapeutic potential of targeting the interplay between mitochondrial dysfunction and the NLRP3 inflammasome in MS. This paper also evaluates the natural and synthetic compounds that can improve mitochondrial function and/or inhibit the NLRP3 inflammasome, thereby providing neuroprotection. Moreover, it summarizes the evidence from animal models of MS that demonstrate the beneficial effects of these compounds on reducing inflammation, demyelination, and neurodegeneration. Finally, this review advocates for a deeper investigation into the molecular crosstalk between mitochondrial dynamics and the NLRP3 inflammasome as a means to refine therapeutic targets for MS.
Collapse
Affiliation(s)
- Alireza Shadab
- Deputy of Health, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Mohammad Abbasi-Kolli
- Deputy of Health, Iran University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mansoore Saharkhiz
- Department of immunology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran; Cellular and molecular research center, Birjand University of medical sciences, Birjand, Iran
| | | | - Behrooz Shokouhi
- Pathology Department, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
11
|
Ravichandran KA, Heneka MT. Inflammasomes in neurological disorders - mechanisms and therapeutic potential. Nat Rev Neurol 2024; 20:67-83. [PMID: 38195712 DOI: 10.1038/s41582-023-00915-x] [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: 12/04/2023] [Indexed: 01/11/2024]
Abstract
Inflammasomes are molecular scaffolds that are activated by damage-associated and pathogen-associated molecular patterns and form a key element of innate immune responses. Consequently, the involvement of inflammasomes in several diseases that are characterized by inflammatory processes, such as multiple sclerosis, is widely appreciated. However, many other neurological conditions, including Alzheimer disease, Parkinson disease, amyotrophic lateral sclerosis, stroke, epilepsy, traumatic brain injury, sepsis-associated encephalopathy and neurological sequelae of COVID-19, all involve persistent inflammation in the brain, and increasing evidence suggests that inflammasome activation contributes to disease progression in these conditions. Understanding the biology and mechanisms of inflammasome activation is, therefore, crucial for the development of inflammasome-targeted therapies for neurological conditions. In this Review, we present the current evidence for and understanding of inflammasome activation in neurological diseases and discuss current and potential interventional strategies that target inflammasome activation to mitigate its pathological consequences.
Collapse
Affiliation(s)
- Kishore Aravind Ravichandran
- Department of Neuroinflammation, Institute of innate immunity, University of Bonn Medical Center Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Esch-sur-Alzette, Luxembourg.
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, North Worcester, MA, USA.
| |
Collapse
|
12
|
Zhan F, Zhang J, He P, Chen W, Ouyang Y. Macrophage-derived exosomal miRNA-141 triggers endothelial cell pyroptosis by targeting NLRP3 to accelerate sepsis progression. Int J Immunopathol Pharmacol 2024; 38:3946320241234736. [PMID: 38652556 PMCID: PMC11041538 DOI: 10.1177/03946320241234736] [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: 09/12/2023] [Accepted: 02/07/2024] [Indexed: 04/25/2024] Open
Abstract
Sepsis, critical condition marked by severe organ dysfunction from uncontrolled infection, involves the endothelium significantly. Macrophages, through paracrine actions, play a vital role in sepsis, but their mechanisms in sepsis pathogenesis remain elusive. Objective: We aimed to explore how macrophage-derived exosomes with low miR-141 expression promote pyroptosis in endothelial cells (ECs). Exosomes from THP-1 cell supernatant were isolated and characterized. The effects of miR-141 mimic/inhibitor on apoptosis, proliferation, and invasion of Human Umbilical Vein Endothelial Cells (HUVECs) were assessed using flow cytometry, CCK-8, and transwell assays. Key pyroptosis-related proteins, including caspase-1, IL-18, IL-1β, NLR Family Pyrin Domain Containing 3 (NLRP3), ASC, and cleaved-GSDMD, were analyzed via Western blot. The interaction between miR-141 and NLRP3 was studied using RNAhybrid v2.2 and dual-Luciferase reporter assays. The mRNA and protein level of NLRP3 after exosomal miR-141 inhibitor treatment was detected by qPCR and Western blot, respectively. Exosomes were successfully isolated. miR-141 mimic reduced cell death and pyroptosis-related protein expression in HUVECs, while the inhibitor had opposite effects, increasing cell death, and enhancing pyroptosis protein expression. Additionally, macrophage-derived exosomal miR-141 inhibitor increased cell death and pyroptosis-related proteins in HUVECs. miR-141 inhibits NLRP3 transcription. Macrophages facilitate sepsis progression by secreting miR-141 decreased exosomes to activate NLRP3-mediated pyroptosis in ECs, which could be a potentially valuable target of sepsis therapy.
Collapse
Affiliation(s)
| | | | - Ping He
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Wenteng Chen
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yanhong Ouyang
- Department of Emergency Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
13
|
Yao J, Wang Z, Song W, Zhang Y. Targeting NLRP3 inflammasome for neurodegenerative disorders. Mol Psychiatry 2023; 28:4512-4527. [PMID: 37670126 DOI: 10.1038/s41380-023-02239-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 09/07/2023]
Abstract
Neuroinflammation is a key pathological feature in neurological diseases, including Alzheimer's disease (AD). The nucleotide-binding domain leucine-rich repeat-containing proteins (NLRs) belong to the pattern recognition receptors (PRRs) family that sense stress signals, which play an important role in inflammation. As a member of NLRs, the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) is predominantly expressed in microglia, the principal innate immune cells in the central nervous system (CNS). Microglia release proinflammatory cytokines to cause pyroptosis through activating NLRP3 inflammasome. The active NLRP3 inflammasome is involved in a variety of neurodegenerative diseases (NDs). Recent studies also indicate the key role of neuronal NLRP3 in the pathogenesis of neurological disorders. In this article, we reviewed the mechanisms of NLRP3 expression and activation and discussed the role of active NLRP3 inflammasome in the pathogenesis of NDs, particularly focusing on AD. The studies suggest that targeting NLRP3 inflammasome could be a novel approach for the disease modification.
Collapse
Affiliation(s)
- Jing Yao
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China
| | - Zhe Wang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China
| | - Weihong Song
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China.
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Clinical Research Center for Mental Disorders, School of Mental Health and The Affiliated Kangning Hospital, Wenzhou Medical University, Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, 325000, Zhejiang, China.
| | - Yun Zhang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, 100053, Beijing, China.
| |
Collapse
|
14
|
Kunze R, Fischer S, Marti HH, Preissner KT. Brain alarm by self-extracellular nucleic acids: from neuroinflammation to neurodegeneration. J Biomed Sci 2023; 30:64. [PMID: 37550658 PMCID: PMC10405513 DOI: 10.1186/s12929-023-00954-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 07/22/2023] [Indexed: 08/09/2023] Open
Abstract
Neurological disorders such as stroke, multiple sclerosis, as well as the neurodegenerative diseases Parkinson's or Alzheimer's disease are accompanied or even powered by danger associated molecular patterns (DAMPs), defined as endogenous molecules released from stressed or damaged tissue. Besides protein-related DAMPs or "alarmins", numerous nucleic acid DAMPs exist in body fluids, such as cell-free nuclear and mitochondrial DNA as well as different species of extracellular RNA, collectively termed as self-extracellular nucleic acids (SENAs). Among these, microRNA, long non-coding RNAs, circular RNAs and extracellular ribosomal RNA constitute the majority of RNA-based DAMPs. Upon tissue injury, necrosis or apoptosis, such SENAs are released from neuronal, immune and other cells predominantly in association with extracellular vesicles and may be translocated to target cells where they can induce intracellular regulatory pathways in gene transcription and translation. The majority of SENA-induced signaling reactions in the brain appear to be related to neuroinflammatory processes, often causally associated with the onset or progression of the respective disease. In this review, the impact of the diverse types of SENAs on neuroinflammatory and neurodegenerative diseases will be discussed. Based on the accumulating knowledge in this field, several specific antagonistic approaches are presented that could serve as therapeutic interventions to lower the pathological outcome of the indicated brain disorders.
Collapse
Affiliation(s)
- Reiner Kunze
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Ruprecht-Karls-University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Silvia Fischer
- Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
| | - Hugo H. Marti
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Ruprecht-Karls-University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Klaus T. Preissner
- Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany
- Kerckhoff-Heart-Research-Institute, Department of Cardiology, Medical School, Justus-Liebig-University, Giessen, Germany
| |
Collapse
|
15
|
Maciak K, Dziedzic A, Saluk J. Remyelination in multiple sclerosis from the miRNA perspective. Front Mol Neurosci 2023; 16:1199313. [PMID: 37333618 PMCID: PMC10270307 DOI: 10.3389/fnmol.2023.1199313] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
Remyelination relies on the repair of damaged myelin sheaths, involving microglia cells, oligodendrocyte precursor cells (OPCs), and mature oligodendrocytes. This process drives the pathophysiology of autoimmune chronic disease of the central nervous system (CNS), multiple sclerosis (MS), leading to nerve cell damage and progressive neurodegeneration. Stimulating the reconstruction of damaged myelin sheaths is one of the goals in terms of delaying the progression of MS symptoms and preventing neuronal damage. Short, noncoding RNA molecules, microRNAs (miRNAs), responsible for regulating gene expression, are believed to play a crucial role in the remyelination process. For example, studies showed that miR-223 promotes efficient activation and phagocytosis of myelin debris by microglia, which is necessary for the initiation of remyelination. Meanwhile, miR-124 promotes the return of activated microglia to the quiescent state, while miR-204 and miR-219 promote the differentiation of mature oligodendrocytes. Furthermore, miR-138, miR-145, and miR-338 have been shown to be involved in the synthesis and assembly of myelin proteins. Various delivery systems, including extracellular vesicles, hold promise as an efficient and non-invasive way for providing miRNAs to stimulate remyelination. This article summarizes the biology of remyelination as well as current challenges and strategies for miRNA molecules in potential diagnostic and therapeutic applications.
Collapse
|