1
|
Medina-Rodriguez EM, Han D, Zeltzer SE, Moraskie Alvarez-Tabío MP, O'Connor G, Daunert S, Beurel E. Stress-induced VIPergic activation mediates microbiota/Th17cell-dependent depressive-like behaviors. Brain Behav Immun 2025; 123:739-751. [PMID: 39419356 DOI: 10.1016/j.bbi.2024.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024] Open
Abstract
Chronic stress often has deleterious effects leading to the development of psychiatric diseases. The gut-brain axis represents a novel avenue for stress research. The negative effects of stress on the gut physiology have been well-described, whereas the pathways whereby stress controls microbial composition to modulate behaviors remains mainly unknown. We discovered that vasoactive intestinal peptide (VIP) activation promoted stress-induced microbial changes leading to increased infiltration of T helper (Th) 17 cells and microglial activation in the hippocampus and depressive-like behaviors, uncovering a close crosstalk between intestinal VIPergic release and the gut microbiota during stress and providing a new interaction between the nervous system and the gut microbiome after stress. Neutralization of the signature cytokine of Th17 cells, interleukin (IL)-17A, was sufficient to block depressive-like behaviors, reduce neuronal VIPergic activation and microglia activation induced by VIPergic activation after stress, opening new potential therapeutic targets for depression.
Collapse
Affiliation(s)
- Eva M Medina-Rodriguez
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Dongmei Han
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Shanie E Zeltzer
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Michael P Moraskie Alvarez-Tabío
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Gregory O'Connor
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Eléonore Beurel
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States.
| |
Collapse
|
2
|
Ma J, Liu X, Zhao Y, Lu Q, Ding G, Wang Y, Cheng X. Th17/Treg balance is regulated during the suppression of experimental autoimmune encephalomyelitis treated by Astragalus polysaccharides via the microbiota-gut-brain axis. Brain Res Bull 2024; 220:111171. [PMID: 39675488 DOI: 10.1016/j.brainresbull.2024.111171] [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: 04/15/2024] [Revised: 10/16/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
The Th17/Treg imbalance is an important cause of immune cell infiltration into the central nervous system (CNS) in multiple sclerosis (MS). The gut microbiota affects the Th17/Treg balance in the gut and in distal areas, such as the CNS, which further contributes to the onset and progression of MS. Our previous studies have shown that Astragalus polysaccharide (APS) has a role in alleviating the clinical symptoms and demyelination of experimental autoimmune encephalomyelitis (EAE) mice, a classic MS model. However, the mechanism of action is not fully understood. In this study, we found that APS suppressed inflammation and regulated the Th17/Treg balance in the CNS and peripheral blood of EAE mice. It was further shown that APS inhibited gut inflammation and reduced Th17 function. The experiment with an antibiotic cocktail interfering with the gut microbiota proved that APS alleviated EAE by regulating the gut microbiota. Through 16S rRNA sequencing, we showed that APS regulated gut microbiota diversity and composition in EAE mice. Then, we found that APS regulated metabolite composition in feces and plasma, thus altering gut and blood metabolic functions. Metabolites related to this pathway, including sphingosine 1 phosphate (S1P), prostaglandin E2 (PGE2), ADP, and ATP, were downregulated by APS. The levels of bile acid metabolites such as taurochenodeoxycholate-7-sulfate and N-palmitoyl aspartic acid were upregulated by APS. In summary, our study demonstrated that APS exerts a suppressive effect on EAE by regulating gut microbiota composition, affecting metabolite composition, and improving the Th17/Treg balance in the peripheral blood and CNS.
Collapse
Affiliation(s)
- Jinyun Ma
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Xiaojun Liu
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Yan Zhao
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Qijin Lu
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Guiqing Ding
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Yuanhua Wang
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Xiaodong Cheng
- Institute of Clinical Immunology, Yue-yang Hospital of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| |
Collapse
|
3
|
Dou Y, You J, Wang J, Li X, Lin Y, Liu B, Ma L. IL-17A Mediates Depressive-Like Symptoms by Inducing Microglia Activation in Psoriasiform Dermatitis Mice. Immun Inflamm Dis 2024; 12:e70092. [PMID: 39660880 PMCID: PMC11632850 DOI: 10.1002/iid3.70092] [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: 06/25/2024] [Revised: 11/10/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Psoriasis is recognized as a systemic disease for its accompanying comorbidities, among which psychological disorders present a high incidence rate and affect patients' life quality. Interleukin (IL)-17A is the central pathological factor in the pathogenesis and development of psoriasis. OBJECTIVE To clarify if psoriasis-induced systemic IL-17A increase can mediate the neuronal inflammation and result in depressive-like symptoms. METHODS Psoriasiform dermatitis model was established by imiquimod (IMQ) application on male BALB/c mice and IL-17A intervention was performed by lateral ventricular catheterization. Skin structural, histopathological characteristics, and behavioral tests were assessed. Serum IL-17A levels were detected by Enzyme-linked immunosorbent assay. mRNA expression of pro-inflammatory factors IL-1β, IL-6, and tumor necrosis factor-α (TNF-α) as well as anti-inflammatory factors IL-4 and IL-10 in the hippocampus and cortex were measured by RT-qPCR. The number of microglia and hippocampal neurons was quantified by immunofluorescent assay. RESULTS IMQ treatment resulted in significant skin structural and histopathological characters of psoriasiform dermatitis with elevated serum IL-17A levels, obvious depressive-like behaviors, microglia activation with increased IL-1β, IL-6, and TNF-α expression levels in the hippocampus and cortex, and notable inhibition of hippocampal neurogenesis. While, IL-17A neutralization by intracerebroventricular injection of anti-IL-17A antibody can remarkably inhibit microglia activation and decrease the abnormally increased expression levels of IL-1β, IL-6, and TNF-α in the hippocampus and cortex of psoriasiform dermatitis mice, promote hippocampal neurogenesis, thus alleviate the depressive-like behaviors. CONCLUSION In the pathological condition of psoriasis, systemic IL-17A elevation can trigger microglia activation, provoke pro-inflammation mediators to release, evoke neuroinflammation, subsequently inhibit hippocampal neurogenesis, and result in depression. IL-17A, as an important pathogenic factor in psoriasis, contributes to its critical role in mediating systemic inflammation and depression comorbidity.
Collapse
Affiliation(s)
- Yue Dou
- Department of DermatologyBinzhou Medical University HospitalBinzhouShandongChina
| | - Jingjing You
- Department of DermatologyBinzhou Medical University HospitalBinzhouShandongChina
- Department of Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
| | - Jing Wang
- Department of DermatologyBinzhou Medical University HospitalBinzhouShandongChina
| | - Xinxin Li
- Department of DermatologyBinzhou Medical University HospitalBinzhouShandongChina
| | - Yawen Lin
- Department of DermatologyBinzhou Medical University HospitalBinzhouShandongChina
| | - Bin Liu
- Department of DermatologyBinzhou Medical University HospitalBinzhouShandongChina
- Department of Medical Research CenterBinzhou Medical University HospitalBinzhouShandongChina
| | - Lei Ma
- Department of DermatologyBinzhou Medical University HospitalBinzhouShandongChina
| |
Collapse
|
4
|
Tsuboi H, Sakakibara H, Minamida-Urata Y, Tsujiguchi H, Hara A, Suzuki K, Miyagi S, Nakamura M, Takazawa C, Kannon T, Zhao J, Shimizu Y, Shibata A, Ogawa A, Suzuki F, Kambayashi Y, Konoshita T, Tajima A, Nakamura H. Serum TNFα and IL-17A levels may predict increased depressive symptoms: findings from the Shika Study cohort project in Japan. Biopsychosoc Med 2024; 18:20. [PMID: 39358787 PMCID: PMC11446020 DOI: 10.1186/s13030-024-00317-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Low-grade systemic inflammation may be a key player in the immune activation that has been reported for mental health deterioration. We hypothesised that elevated serum levels of inflammatory cytokines increase neuroinflammation and exacerbate depressive symptoms. METHODS The participants were part of a cohort study for whom data was available for both 2015 and 2019. In 2015, blood samples were collected from 232 participants. Their depressive symptoms were assessed both 2015 and 2019 using the Centre for Epidemiologic Studies Depression Scale (CES-D) (n = 33). The multiplex immunoassay system (Luminex® 200) was used to measure the serum concentrations of IL-6, IL-10, IL-12, IL-17A and TNFα. Data were analysed using linear models with the level of significance considered to be p < 0.05. RESULTS After controlling for age, BMI, smoking and alcohol consumption, in 2015 the serum concentrations of IL-17A and TNFα in 2015 were significantly positively associated with the CES-D scores of women (standardised β (B) = .027, p < 0.01 and B = 0.26, p < 0.01, respectively). The serum concentrations of IL-17A and TNFα of men were significantly positively associated with the CES-D scores of 2019 (B = 0.62, p = 0.02 and B = 0.59, p = 0.02, respectively). CONCLUSIONS In this cross-sectional study, we found a significant positive correlation between the depressive symptoms and serum TNFα and IL-17A levels of women. In addition, our longitudinal findings suggest the possibility that TNFα and IL-17A could elevate the depressive symptoms of men.
Collapse
Affiliation(s)
- Hirohito Tsuboi
- Graduate School of Human Sciences, The University of Shiga Prefecture, 2500 Hassaka-Cho, Hikone, 522-8533, Japan.
- Research Group of Psychosomatic Medicine, Faculty of Pharmacy, Pharmaceutical and Health Sciences, Kanazawa University, 1 Kakuma-Machi, Kanazawa, 920-1192, Japan.
- Department of Hygiene and Public Health, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan.
| | - Hiroyuki Sakakibara
- Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai, Nada-ku, Kobe, 657-8501, Japan
| | - Yuuki Minamida-Urata
- Research Group of Psychosomatic Medicine, Faculty of Pharmacy, Pharmaceutical and Health Sciences, Kanazawa University, 1 Kakuma-Machi, Kanazawa, 920-1192, Japan
| | - Hiromasa Tsujiguchi
- Department of Hygiene and Public Health, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Akinori Hara
- Department of Hygiene and Public Health, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Keita Suzuki
- Department of Hygiene and Public Health, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Sakae Miyagi
- Innovative Clinical Research Center, Kanazawa University, 13-1 Takara-Machi, Kanazawa, Japan
| | - Masaharu Nakamura
- Department of Hygiene and Public Health, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Chie Takazawa
- Department of Hygiene and Public Health, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Takayuki Kannon
- Department of Biomedical Data Science, School of Medicine, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, 470-1192, Japan
| | - Jiaye Zhao
- Department of Hygiene and Public Health, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Yukari Shimizu
- Department of Nursing, Faculty of Health Sciences, Komatsu University, 10-10 Doihara-Machi, Komatsu, 923-0921, Japan
| | - Aki Shibata
- Department of Hygiene and Public Health, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Aya Ogawa
- Department of Hygiene and Public Health, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| | - Fumihiko Suzuki
- Department of Geriatric Dentistry, Ohu University School of Dentistry, 31-1 Misumido, Koriyama, 963-8611, Japan
| | - Yasuhiro Kambayashi
- Department of Public Health, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoinooka, Imabari, 794-8555, Japan
| | - Tadashi Konoshita
- Division of Diabetes Endocrinology and Metabolism, Yachiyo Medical Center, Tokyo Women's Medical University, 477-96 Owada-Shinden, Yachiyo, Japan
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8640, Japan
| | - Hiroyuki Nakamura
- Department of Hygiene and Public Health, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takara-Machi, Kanazawa, 920-8641, Japan
| |
Collapse
|
5
|
Kostic M, Zivkovic N, Cvetanovic A, Basic J, Stojanovic I. Dissecting the immune response of CD4 + T cells in Alzheimer's disease. Rev Neurosci 2024:revneuro-2024-0090. [PMID: 39238424 DOI: 10.1515/revneuro-2024-0090] [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: 07/04/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024]
Abstract
The formation of amyloid-β (Aβ) plaques is a neuropathological hallmark of Alzheimer's disease (AD), however, these pathological aggregates can also be found in the brains of cognitively unimpaired elderly population. In that context, individual variations in the Aβ-specific immune response could be key factors that determine the level of Aβ-induced neuroinflammation and thus the propensity to develop AD. CD4+ T cells are the cornerstone of the immune response that coordinate the effector functions of both adaptive and innate immunity. However, despite intensive research efforts, the precise role of these cells during AD pathogenesis is still not fully elucidated. Both pathogenic and beneficial effects have been observed in various animal models of AD, as well as in humans with AD. Although this functional duality of CD4+ T cells in AD can be simply attributed to the vast phenotype heterogeneity of this cell lineage, disease stage-specific effect have also been proposed. Therefore, in this review, we summarized the current understanding of the role of CD4+ T cells in the pathophysiology of AD, from the aspect of their antigen specificity, activation, and phenotype characteristics. Such knowledge is of practical importance as it paves the way for immunomodulation as a therapeutic option for AD treatment, given that currently available therapies have not yielded satisfactory results.
Collapse
Affiliation(s)
- Milos Kostic
- Department of Immunology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Nikola Zivkovic
- Department of Pathology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ana Cvetanovic
- Department of Oncology, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Jelena Basic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| | - Ivana Stojanovic
- Department of Biochemistry, Medical Faculty of Nis, University of Nis, Blvd. dr Zorana Djindjica 81, Nis, 18000, Serbia
| |
Collapse
|
6
|
Łaszczych D, Czernicka A, Gostomczyk K, Szylberg Ł, Borowczak J. The role of IL-17 in the pathogenesis and treatment of glioblastoma-an update on the state of the art and future perspectives. Med Oncol 2024; 41:187. [PMID: 38918274 PMCID: PMC11199243 DOI: 10.1007/s12032-024-02434-1] [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: 05/06/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
Glioblastoma (GBM) is the most common malignant brain tumor, which, despite significant progress made in the last years in the field of neuro-oncology, remains an incurable disease. GBM has a poor prognosis with a median survival of 12-15 months, and its aggressive clinical course is related to rapid growth, extensive infiltration of adjacent tissues, resistance to chemotherapy, radiotherapy and immunotherapy, and frequent relapse. Currently, several molecular biomarkers are used in clinical practice to predict patient prognosis and response to treatment. However, due to the overall unsatisfactory efficacy of standard multimodal treatment and the remaining poor prognosis, there is an urgent need for new biomarkers and therapeutic strategies for GBM. Recent evidence suggests that GBM tumorigenesis is associated with crosstalk between cancer, immune and stromal cells mediated by various cytokines. One of the key factors involved in this process appears to be interleukin-17 (IL-17), a pro-inflammatory cytokine that is significantly upregulated in the serum and tissue of GBM patients. IL-17 plays a key role in tumorigenesis, angiogenesis, and recurrence of GBM by activating pro-oncogenic signaling pathways and promoting cell survival, proliferation, and invasion. IL-17 facilitates the immunomodulation of the tumor microenvironment by promoting immune cells infiltration and cytokine secretion. In this article we review the latest scientific reports to provide an update on the role of IL-17 role in tumorigenesis, tumor microenvironment, diagnosis, prognosis, and treatment of GBM.
Collapse
Affiliation(s)
- Dariusz Łaszczych
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum, Nicolaus Copernicus University in Bydgoszcz, Ujejskiego 75 street, 85-168, Bydgoszcz, Poland.
| | - Aleksandra Czernicka
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum, Nicolaus Copernicus University in Bydgoszcz, Ujejskiego 75 street, 85-168, Bydgoszcz, Poland
| | - Karol Gostomczyk
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum, Nicolaus Copernicus University in Bydgoszcz, Ujejskiego 75 street, 85-168, Bydgoszcz, Poland
| | - Łukasz Szylberg
- Department of Obstetrics, Gynaecology and Oncology, Collegium Medicum, Nicolaus Copernicus University in Bydgoszcz, Ujejskiego 75 street, 85-168, Bydgoszcz, Poland
- Department of Tumor Pathology and Pathomorphology, Oncology Centre - Prof. Franciszek Łukaszczyk Memorial Hospital, dr Izabeli Romanowskiej 2 street, 85-796, Bydgoszcz, Poland
| | - Jędrzej Borowczak
- Department of Clinical Oncology, Oncology Centre - Prof. Franciszek Łukaszczyk Memorial Hospital, dr Izabeli Romanowskiej 2 street, 85-796, Bydgoszcz, Poland
| |
Collapse
|
7
|
Holt EA, Tyler A, Lakusta-Wong T, Lahue KG, Hankes KC, Teuscher C, Lynch RM, Ferris MT, Mahoney JM, Krementsov DN. Probing the basis of disease heterogeneity in multiple sclerosis using genetically diverse mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597205. [PMID: 38895248 PMCID: PMC11185616 DOI: 10.1101/2024.06.03.597205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Multiple sclerosis (MS) is a complex disease with significant heterogeneity in disease course and progression. Genetic studies have identified numerous loci associated with MS risk, but the genetic basis of disease progression remains elusive. To address this, we leveraged the Collaborative Cross (CC), a genetically diverse mouse strain panel, and experimental autoimmune encephalomyelitis (EAE). The thirty-two CC strains studied captured a wide spectrum of EAE severity, trajectory, and presentation, including severe-progressive, monophasic, relapsing remitting, and axial rotary (AR)-EAE, accompanied by distinct immunopathology. Sex differences in EAE severity were observed in six strains. Quantitative trait locus analysis revealed distinct genetic linkage patterns for different EAE phenotypes, including EAE severity and incidence of AR-EAE. Machine learning-based approaches prioritized candidate genes for loci underlying EAE severity ( Abcc4 and Gpc6 ) and AR-EAE ( Yap1 and Dync2h1 ). This work expands the EAE phenotypic repertoire and identifies novel loci controlling unique EAE phenotypes, supporting the hypothesis that heterogeneity in MS disease course is driven by genetic variation. Summary The genetic basis of disease heterogeneity in multiple sclerosis (MS) remains elusive. We leveraged the Collaborative Cross to expand the phenotypic repertoire of the experimental autoimmune encephalomyelitis (EAE) model of MS and identify loci controlling EAE severity, trajectory, and presentation.
Collapse
|
8
|
Santiago-Balmaseda A, Aguirre-Orozco A, Valenzuela-Arzeta IE, Villegas-Rojas MM, Pérez-Segura I, Jiménez-Barrios N, Hurtado-Robles E, Rodríguez-Hernández LD, Rivera-German ER, Guerra-Crespo M, Martinez-Fong D, Ledesma-Alonso C, Diaz-Cintra S, Soto-Rojas LO. Neurodegenerative Diseases: Unraveling the Heterogeneity of Astrocytes. Cells 2024; 13:921. [PMID: 38891053 PMCID: PMC11172252 DOI: 10.3390/cells13110921] [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: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
The astrocyte population, around 50% of human brain cells, plays a crucial role in maintaining the overall health and functionality of the central nervous system (CNS). Astrocytes are vital in orchestrating neuronal development by releasing synaptogenic molecules and eliminating excessive synapses. They also modulate neuronal excitability and contribute to CNS homeostasis, promoting neuronal survival by clearance of neurotransmitters, transporting metabolites, and secreting trophic factors. Astrocytes are highly heterogeneous and respond to CNS injuries and diseases through a process known as reactive astrogliosis, which can contribute to both inflammation and its resolution. Recent evidence has revealed remarkable alterations in astrocyte transcriptomes in response to several diseases, identifying at least two distinct phenotypes called A1 or neurotoxic and A2 or neuroprotective astrocytes. However, due to the vast heterogeneity of these cells, it is limited to classify them into only two phenotypes. This review explores the various physiological and pathophysiological roles, potential markers, and pathways that might be activated in different astrocytic phenotypes. Furthermore, we discuss the astrocyte heterogeneity in the main neurodegenerative diseases and identify potential therapeutic strategies. Understanding the underlying mechanisms in the differentiation and imbalance of the astrocytic population will allow the identification of specific biomarkers and timely therapeutic approaches in various neurodegenerative diseases.
Collapse
Affiliation(s)
- Alberto Santiago-Balmaseda
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| | - Annai Aguirre-Orozco
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (I.E.V.-A.); (N.J.-B.); (D.M.-F.)
| | - Irais E. Valenzuela-Arzeta
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (I.E.V.-A.); (N.J.-B.); (D.M.-F.)
| | - Marcos M. Villegas-Rojas
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de Mexico 11340, Mexico
| | - Isaac Pérez-Segura
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| | - Natalie Jiménez-Barrios
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (I.E.V.-A.); (N.J.-B.); (D.M.-F.)
| | - Ernesto Hurtado-Robles
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| | - Luis Daniel Rodríguez-Hernández
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| | - Erick R. Rivera-German
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| | - Magdalena Guerra-Crespo
- Laboratorio de Medicina Regenerativa, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de Mexico, Mexico City 04510, Mexico;
| | - Daniel Martinez-Fong
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (I.E.V.-A.); (N.J.-B.); (D.M.-F.)
| | - Carlos Ledesma-Alonso
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de Mexico, Querétaro 76230, Mexico;
| | - Sofía Diaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de Mexico, Querétaro 76230, Mexico;
| | - Luis O. Soto-Rojas
- Laboratorio de Patogénesis Molecular, Laboratorio 4 Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico City 54090, Mexico; (A.S.-B.); (A.A.-O.); (M.M.V.-R.); (I.P.-S.); (E.H.-R.); (L.D.R.-H.); (E.R.R.-G.)
| |
Collapse
|
9
|
Jonischkies K, del Angel M, Demiray YE, Loaiza Zambrano A, Stork O. The NDR family of kinases: essential regulators of aging. Front Mol Neurosci 2024; 17:1371086. [PMID: 38803357 PMCID: PMC11129689 DOI: 10.3389/fnmol.2024.1371086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Aging is defined as a progressive decline of cognitive and physiological functions over lifetime. Since the definition of the nine hallmarks of aging in 2013 by López-Otin, numerous studies have attempted to identify the main regulators and contributors in the aging process. One interesting group of proteins whose participation has been implicated in several aging hallmarks are the nuclear DBF2-related (NDR) family of serine-threonine AGC kinases. They are one of the core components of the Hippo signaling pathway and include NDR1, NDR2, LATS1 and LATS2 in mammals, along with its highly conserved metazoan orthologs; Trc in Drosophila melanogaster, SAX-1 in Caenorhabditis elegans, CBK1, DBF20 in Saccharomyces cerevisiae and orb6 in Saccharomyces pombe. These kinases have been independently linked to the regulation of widely diverse cellular processes disrupted during aging such as the cell cycle progression, transcription, intercellular communication, nutrient homeostasis, autophagy, apoptosis, and stem cell differentiation. However, a comprehensive overview of the state-of-the-art knowledge regarding the post-translational modifications of and by NDR kinases in aging has not been conducted. In this review, we summarize the current understanding of the NDR family of kinases, focusing on their relevance to various aging hallmarks, and emphasize the growing body of evidence that suggests NDR kinases are essential regulators of aging across species.
Collapse
Affiliation(s)
- Kevin Jonischkies
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Miguel del Angel
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Yunus Emre Demiray
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Allison Loaiza Zambrano
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Oliver Stork
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Science, Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
- German Center for Mental Health (DZPG), Jena-Magdeburg-Halle, Germany
| |
Collapse
|
10
|
Dai HY, Zhang ZX, Tan C, Xian X, Ji D, Yang J, Sun J, Yao H. Propionic acid ameliorates cognitive function through immunomodulatory effects on Th17 cells in perioperative neurocognitive disorders. Heliyon 2024; 10:e28817. [PMID: 38699705 PMCID: PMC11063405 DOI: 10.1016/j.heliyon.2024.e28817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 05/05/2024] Open
Abstract
Background Elderly patients undergoing surgery are prone to cognitive decline known as perioperative neurocognitive disorders (PND). Several studies have shown that the microglial activation and the decrease of short-chain fatty acids (SCFAs) in gut induced by surgery may be related to the pathogenesis of PND. The purpose of this study was to determine whether microglia and short-chain fatty acids were involved in cognitive dysfunction in aged rats. Methods Male wild-type Wistar rats aged 11-12 months were randomly divided into control group (Ctrl: Veh group), propionic acid group (Ctrl: PA group), exploratory laparotomy group (LP: Veh group) and propionic acid + exploratory laparotomy group (LP: PA group) according to whether exploratory laparotomy (LP) or PA pretreatment for 21 days was performed. The motor ability of the rats was evaluated by open field test on postoperative day 3 (POD3), and then the cognitive function was evaluated by Y-maze test and fear conditioning test. The expression of IL-1β, IL-6, RORγt and IL-17A mRNA in hippocampus was detected by RT-qPCR, the expression of IL-17A and IL-17RA in hippocampus was detected by Western blot, and the activation of microglia was detected by immunofluorescence. Results The PND rat model was successfully established by laparotomy. Compared with Ctrl: Veh group, the body weight of LP: Veh group decreased, the percentage of spontaneous alternations in Y maze decreased (P < 0.001), and the percentage of freezing time in contextual fear test decreased (P < 0.001). Surgery triggers neuroinflammation, manifested as the elevated levels of the inflammatory cytokines IL-1β (P < 0.001) and IL-6 (P < 0.001), the increased expression of the transcription factor RORγt (P = 0.0181, POD1; P = 0.0073, POD5)and major inflammatory cytokines IL-17A (P = 0.0215, POD1; P = 0.0071, POD5), and the increased average fluorescence intensity of Iba1 (P < 0.001, POD1; P < 0.001, POD5). After PA preconditioning, the recovery of rats in LP: PA group was faster than that in LP: Veh group as the body weight lost on POD1 (P = 0.0148) was close to the baseline level on POD5 (P = 0.1846), and they performed better in behavioral tests. The levels of IL-1β (P < 0.001) and IL-6 (P = 0.0035) inflammatory factors in hippocampus decreased on POD1 and the average fluorescence intensity of Iba1 decreased (P = 0.0024, POD1; P < 0.001, POD5), representing the neuroinflammation was significantly improved. Besides, the levels of RORγt mRNA (P = 0.0231, POD1; P = 0.0251, POD5) and IL-17A mRNA (P = 0.0208, POD1; P = 0.0071, POD5) in hippocampus as well as the expression of IL-17A (P = 0.0057, POD1; P < 0.001, POD5) and IL-17RA (P = 0.0388) decreased. Conclusion PA pretreatment results in reduced postoperative neuroinflammation and improved cognitive function, potentially attributed to the regulatory effects of PA on Th17-mediated immune responses.
Collapse
Affiliation(s)
- Hong-yu Dai
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ze-xin Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cheng Tan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xian Xian
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dong Ji
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Yang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jie Sun
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Hao Yao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
11
|
Arenas YM, López-Gramaje A, Montoliu C, Llansola M, Felipo V. Increased levels and activation of the IL-17 receptor in microglia contribute to enhanced neuroinflammation in cerebellum of hyperammonemic rats. Biol Res 2024; 57:18. [PMID: 38671534 PMCID: PMC11055256 DOI: 10.1186/s40659-024-00504-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Patients with liver cirrhosis may show minimal hepatic encephalopathy (MHE) with mild cognitive impairment and motor incoordination. Rats with chronic hyperammonemia reproduce these alterations. Motor incoordination in hyperammonemic rats is due to increased GABAergic neurotransmission in cerebellum, induced by neuroinflammation, which enhances TNFα-TNFR1-S1PR2-CCL2-BDNF-TrkB pathway activation. The initial events by which hyperammonemia triggers activation of this pathway remain unclear. MHE in cirrhotic patients is triggered by a shift in inflammation with increased IL-17. The aims of this work were: (1) assess if hyperammonemia increases IL-17 content and membrane expression of its receptor in cerebellum of hyperammonemic rats; (2) identify the cell types in which IL-17 receptor is expressed and IL-17 increases in hyperammonemia; (3) assess if blocking IL-17 signaling with anti-IL-17 ex-vivo reverses activation of glia and of the TNFα-TNFR1-S1PR2-CCL2-BDNF-TrkB pathway. RESULTS IL-17 levels and membrane expression of the IL-17 receptor are increased in cerebellum of rats with hyperammonemia and MHE, leading to increased activation of IL-17 receptor in microglia, which triggers activation of STAT3 and NF-kB, increasing IL-17 and TNFα levels, respectively. TNFα released from microglia activates TNFR1 in Purkinje neurons, leading to activation of NF-kB and increased IL-17 and TNFα also in these cells. Enhanced TNFR1 activation also enhances activation of the TNFR1-S1PR2-CCL2-BDNF-TrkB pathway which mediates microglia and astrocytes activation. CONCLUSIONS All these steps are triggered by enhanced activation of IL-17 receptor in microglia and are prevented by ex-vivo treatment with anti-IL-17. IL-17 and IL-17 receptor in microglia would be therapeutic targets to treat neurological impairment in patients with MHE.
Collapse
Affiliation(s)
- Yaiza M Arenas
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain
- Departamento de Patología, Facultad de Medicina, Universidad Valencia, Valencia, Spain
- INCLIVA Instituto de Investigación Sanitaria, Valencia, Spain
| | - Adrià López-Gramaje
- Departamento de Patología, Facultad de Medicina, Universidad Valencia, Valencia, Spain
- INCLIVA Instituto de Investigación Sanitaria, Valencia, Spain
| | - Carmina Montoliu
- Departamento de Patología, Facultad de Medicina, Universidad Valencia, Valencia, Spain
- INCLIVA Instituto de Investigación Sanitaria, Valencia, Spain
| | - Marta Llansola
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain
| | - Vicente Felipo
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Eduardo Primo-Yufera 3, 46012, Valencia, Spain.
| |
Collapse
|
12
|
Tamberi L, Belloni A, Pugnaloni A, Rippo MR, Olivieri F, Procopio AD, Bronte G. The Influence of Myeloid-Derived Suppressor Cell Expansion in Neuroinflammation and Neurodegenerative Diseases. Cells 2024; 13:643. [PMID: 38607083 PMCID: PMC11011419 DOI: 10.3390/cells13070643] [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: 02/22/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024] Open
Abstract
The neuro-immune axis has a crucial function both during physiological and pathological conditions. Among the immune cells, myeloid-derived suppressor cells (MDSCs) exert a pivotal role in regulating the immune response in many pathological conditions, influencing neuroinflammation and neurodegenerative disease progression. In chronic neuroinflammation, MDSCs could lead to exacerbation of the inflammatory state and eventually participate in the impairment of cognitive functions. To have a complete overview of the role of MDSCs in neurodegenerative diseases, research on PubMed for articles using a combination of terms made with Boolean operators was performed. According to the search strategy, 80 papers were retrieved. Among these, 44 papers met the eligibility criteria. The two subtypes of MDSCs, monocytic and polymorphonuclear MDSCs, behave differently in these diseases. The initial MDSC proliferation is fundamental for attenuating inflammation in Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS), but not in amyotrophic lateral sclerosis (ALS), where MDSC expansion leads to exacerbation of the disease. Moreover, the accumulation of MDSC subtypes in distinct organs changes during the disease. The proliferation of MDSC subtypes occurs at different disease stages and can influence the progression of each neurodegenerative disorder differently.
Collapse
Affiliation(s)
- Lorenza Tamberi
- Department of Clinical and Molecular Sciences (DISCLIMO), Polytechnic University of Marche, 60121 Ancona, Italy; (L.T.); (A.P.); (M.R.R.); (F.O.); (A.D.P.); (G.B.)
| | - Alessia Belloni
- Department of Clinical and Molecular Sciences (DISCLIMO), Polytechnic University of Marche, 60121 Ancona, Italy; (L.T.); (A.P.); (M.R.R.); (F.O.); (A.D.P.); (G.B.)
| | - Armanda Pugnaloni
- Department of Clinical and Molecular Sciences (DISCLIMO), Polytechnic University of Marche, 60121 Ancona, Italy; (L.T.); (A.P.); (M.R.R.); (F.O.); (A.D.P.); (G.B.)
| | - Maria Rita Rippo
- Department of Clinical and Molecular Sciences (DISCLIMO), Polytechnic University of Marche, 60121 Ancona, Italy; (L.T.); (A.P.); (M.R.R.); (F.O.); (A.D.P.); (G.B.)
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences (DISCLIMO), Polytechnic University of Marche, 60121 Ancona, Italy; (L.T.); (A.P.); (M.R.R.); (F.O.); (A.D.P.); (G.B.)
- Clinic of Laboratory and Precision Medicine, National Institute of Health and Sciences on Ageing (IRCCS INRCA), 60124 Ancona, Italy
| | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences (DISCLIMO), Polytechnic University of Marche, 60121 Ancona, Italy; (L.T.); (A.P.); (M.R.R.); (F.O.); (A.D.P.); (G.B.)
- Clinic of Laboratory and Precision Medicine, National Institute of Health and Sciences on Ageing (IRCCS INRCA), 60124 Ancona, Italy
| | - Giuseppe Bronte
- Department of Clinical and Molecular Sciences (DISCLIMO), Polytechnic University of Marche, 60121 Ancona, Italy; (L.T.); (A.P.); (M.R.R.); (F.O.); (A.D.P.); (G.B.)
- Clinic of Laboratory and Precision Medicine, National Institute of Health and Sciences on Ageing (IRCCS INRCA), 60124 Ancona, Italy
| |
Collapse
|
13
|
Vacharasin JM, Ward JA, McCord MM, Cox K, Imitola J, Lizarraga SB. Neuroimmune mechanisms in autism etiology - untangling a complex problem using human cellular models. OXFORD OPEN NEUROSCIENCE 2024; 3:kvae003. [PMID: 38665176 PMCID: PMC11044813 DOI: 10.1093/oons/kvae003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 01/13/2024] [Accepted: 01/31/2024] [Indexed: 04/28/2024]
Abstract
Autism spectrum disorder (ASD) affects 1 in 36 people and is more often diagnosed in males than in females. Core features of ASD are impaired social interactions, repetitive behaviors and deficits in verbal communication. ASD is a highly heterogeneous and heritable disorder, yet its underlying genetic causes account only for up to 80% of the cases. Hence, a subset of ASD cases could be influenced by environmental risk factors. Maternal immune activation (MIA) is a response to inflammation during pregnancy, which can lead to increased inflammatory signals to the fetus. Inflammatory signals can cross the placenta and blood brain barriers affecting fetal brain development. Epidemiological and animal studies suggest that MIA could contribute to ASD etiology. However, human mechanistic studies have been hindered by a lack of experimental systems that could replicate the impact of MIA during fetal development. Therefore, mechanisms altered by inflammation during human pre-natal brain development, and that could underlie ASD pathogenesis have been largely understudied. The advent of human cellular models with induced pluripotent stem cell (iPSC) and organoid technology is closing this gap in knowledge by providing both access to molecular manipulations and culturing capability of tissue that would be otherwise inaccessible. We present an overview of multiple levels of evidence from clinical, epidemiological, and cellular studies that provide a potential link between higher ASD risk and inflammation. More importantly, we discuss how stem cell-derived models may constitute an ideal experimental system to mechanistically interrogate the effect of inflammation during the early stages of brain development.
Collapse
Affiliation(s)
- Janay M Vacharasin
- Department of Biological Sciences, and Center for Childhood Neurotherapeutics, Univ. of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA
- Department of Biological Sciences, Francis Marion University, 4822 East Palmetto Street, Florence, S.C. 29506, USA
| | - Joseph A Ward
- Department of Molecular Biology, Cell Biology, & Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute of Brain Science, Brown University, 70 Ship Street, Providence, RI 02903, USA
| | - Mikayla M McCord
- Department of Biological Sciences, and Center for Childhood Neurotherapeutics, Univ. of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA
| | - Kaitlin Cox
- Department of Biological Sciences, and Center for Childhood Neurotherapeutics, Univ. of South Carolina, 715 Sumter Street, Columbia, SC 29208, USA
| | - Jaime Imitola
- Laboratory of Neural Stem Cells and Functional Neurogenetics, UConn Health, Departments of Neuroscience, Neurology, Genetics and Genome Sciences, UConn Health, 263 Farmington Avenue, Farmington, CT 06030-5357, USA
| | - Sofia B Lizarraga
- Department of Molecular Biology, Cell Biology, & Biochemistry, Brown University, 185 Meeting Street, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute of Brain Science, Brown University, 70 Ship Street, Providence, RI 02903, USA
| |
Collapse
|
14
|
Yu Y, Weiss RM, Wei S. Interleukin 17A Contributes to Blood-Brain Barrier Disruption of Hypothalamic Paraventricular Nucleus in Rats With Myocardial Infarction. J Am Heart Assoc 2024; 13:e032533. [PMID: 38240234 PMCID: PMC11056165 DOI: 10.1161/jaha.123.032533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 12/12/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND Elevated inflammatory cytokines in the periphery have been identified as active contributors to neuroinflammation and sympathetic overactivity in heart failure (HF). Yet, the exact mechanisms by which these cytokines breach the blood-brain barrier (BBB) to exert their effects on the brain remain elusive. Interleukin 17A has been linked to BBB disruption in various neurologic disorders, and its levels were significantly augmented in circulation and the brain in HF. The present study aimed to determine whether the BBB integrity was compromised within the hypothalamic paraventricular nucleus (PVN), and if so, whether interleukin 17A contributes to BBB disruption in myocardial infarction-induced HF. METHODS AND RESULTS Male Sprague-Dawley rats underwent coronary artery ligation to induce HF or sham surgery. Some HF rats received bilateral PVN microinjections of an interleukin 17 receptor A small interfering RNA or a scrambled small interfering RNA adeno-associated virus. Four weeks after coronary artery ligation, the permeability of the BBB was evaluated by intracarotid injection of fluorescent dyes (fluorescein isothiocyanate-dextran 10 kDa+rhodamine-dextran 70 kDa). Compared with sham-operated rats, HF rats exhibited an elevated extravasation of fluorescein isothiocyanate-dextran 10 kDa within the PVN but not in the brain cortex. The plasma interleukin 17A levels were positively correlated with fluorescein isothiocyanate 10 kDa extravasation in the PVN. The expression of caveolin-1, a transcytosis marker, was augmented, whereas the expression of tight junction proteins was diminished in HF rats. Interleukin 17 receptor A was identified within the endothelium of PVN microvessels. Treatment with interleukin 17 receptor A small interfering RNA led to a significant attenuation of fluorescein isothiocyanate 10 kDa extravasation in the PVN and reversed expression of caveolin-1 and tight junction-associated proteins in the PVN. CONCLUSIONS Collectively, these data indicate that BBB permeability within the PVN is enhanced in HF and is likely attributable to increased interleukin 17A/interleukin 17 receptor A signaling in the BBB endothelium, by promoting caveolar transcytosis and degradation of tight junction complexes.
Collapse
Affiliation(s)
- Yang Yu
- Department of Internal MedicineUniversity of Iowa Carver College of MedicineIowa CityIA
| | - Robert M. Weiss
- Department of Internal MedicineUniversity of Iowa Carver College of MedicineIowa CityIA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of MedicineIowa CityIA
- Veteran Affairs Medical CenterIowa CityIA
| | - Shun‐Guang Wei
- Department of Internal MedicineUniversity of Iowa Carver College of MedicineIowa CityIA
- Abboud Cardiovascular Research Center, University of Iowa Carver College of MedicineIowa CityIA
- Iowa Neuroscience Institute, University of Iowa Carver College of MedicineIowa CityIA
- Veteran Affairs Medical CenterIowa CityIA
| |
Collapse
|
15
|
Kazemi R, Yazdanpanah E, Esmaeili SA, Yousefi B, Baharlou R, Haghmorad D. Thymoquinone improves experimental autoimmune encephalomyelitis by regulating both pro-inflammatory and anti-inflammatory cytokines. Mol Biol Rep 2024; 51:256. [PMID: 38302802 DOI: 10.1007/s11033-023-09148-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/13/2023] [Indexed: 02/03/2024]
Abstract
Introduction Multiple sclerosis (MS) is an autoimmune condition marked by inflammation and the loss of myelin in the central nervous system (CNS). The aim of this research was to understand how Thymoquinone regulate the molecular and cellular processes involved in controlling experimental autoimmune encephalomyelitis (EAE), which is an animal model often used to study MS. Methods Female C57BL/6 mice were split into different groups receiving different doses (low, medium, and high) of Thymoquinone simultaneously with EAE induction. Clinical scores and other measurements were observed daily throughout the 25-day post immunization. We assessed lymphocyte infiltration and demyelination in the spinal cord through histological staining, analyzed T-cell profiles using ELISA, and quantified the expression levels of transcription factors in the CNS using Real-time PCR. Results Thymoquinone prevented the development of EAE. Histological experiments revealed only a small degree of leukocyte infiltration into the CNS. Thymoquinone resulted in a notable reduction in the generation of IFN-γ, IL-17, and IL-6, while simultaneously increasing the production of IL-4, IL-10, and TGF-β in Th2 and Treg cells. Results from Real-time PCR suggested Treatment with Thymoquinone decreased the expression of T-bet and ROR-γt while increasing the expression of Foxp3 and GATA3. Conclusion These findings showed that Thymoquinone could decrease both disease incidence and severity.
Collapse
Affiliation(s)
- Roya Kazemi
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Esmaeil Yazdanpanah
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Bahman Yousefi
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Rasoul Baharlou
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Dariush Haghmorad
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
16
|
Amoriello R, Memo C, Ballerini L, Ballerini C. The brain cytokine orchestra in multiple sclerosis: from neuroinflammation to synaptopathology. Mol Brain 2024; 17:4. [PMID: 38263055 PMCID: PMC10807071 DOI: 10.1186/s13041-024-01077-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/18/2024] [Indexed: 01/25/2024] Open
Abstract
The central nervous system (CNS) is finely protected by the blood-brain barrier (BBB). Immune soluble factors such as cytokines (CKs) are normally produced in the CNS, contributing to physiological immunosurveillance and homeostatic synaptic scaling. CKs are peptide, pleiotropic molecules involved in a broad range of cellular functions, with a pivotal role in resolving the inflammation and promoting tissue healing. However, pro-inflammatory CKs can exert a detrimental effect in pathological conditions, spreading the damage. In the inflamed CNS, CKs recruit immune cells, stimulate the local production of other inflammatory mediators, and promote synaptic dysfunction. Our understanding of neuroinflammation in humans owes much to the study of multiple sclerosis (MS), the most common autoimmune and demyelinating disease, in which autoreactive T cells migrate from the periphery to the CNS after the encounter with a still unknown antigen. CNS-infiltrating T cells produce pro-inflammatory CKs that aggravate local demyelination and neurodegeneration. This review aims to recapitulate the state of the art about CKs role in the healthy and inflamed CNS, with focus on recent advances bridging the study of adaptive immune system and neurophysiology.
Collapse
Affiliation(s)
- Roberta Amoriello
- International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy.
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, 50139, Florence, Italy.
| | - Christian Memo
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, 50139, Florence, Italy
| | - Laura Ballerini
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, 50139, Florence, Italy
| | - Clara Ballerini
- International School for Advanced Studies (SISSA/ISAS), 34136, Trieste, Italy.
| |
Collapse
|
17
|
Iitani Y, Miki R, Imai K, Fuma K, Ushida T, Tano S, Yoshida K, Yokoi A, Kajiyama H, Kotani T. Interleukin-17A stimulation induces alterations in Microglial microRNA expression profiles. Pediatr Res 2024; 95:167-173. [PMID: 37758861 DOI: 10.1038/s41390-023-02825-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Increased maternal interleukin (IL)-17A and activated microglia are pivotal factors contributing to the pathological phenotypes of maternal immune activation (MIA), developing neurodevelopmental disorders in offspring. This study aimed to determine whether IL-17A affects the microglial microRNA (miRNA) profiles. METHODS The miRNA expression profiles of primary cultured microglia stimulated with recombinant IL-17A were examined comprehensively using miRNA sequencing and validated through qRT-PCR. The expressions of miRNAs target genes identified using bioinformatics, were investigated in microglia transfected with mimic miRNA. The target gene's expression was also examined in the fetal brains of the MIA mouse model induced by maternal lipopolysaccharide (LPS) administration. RESULTS Primary cultured microglia expressed the IL-17A receptor and increased proinflammatory cytokines and nitric oxide synthase 2 upon treatment with IL-17A. Among the three miRNAs with |log2FC | >1, only mmu-miR-206-3p expression was significantly up-regulated by IL-17A. Transfection with the mmu-miR-206-3p mimic resulted in a significant decrease in the expression of Hdac4 and Igf1, target genes of mmu-miR-206-3p. Hdac4 expression also significantly decreased in the LPS-induced MIA model. CONCLUSIONS IL-17A affected microglial miRNA profiles with upregulated mmu-miR-206-3p. These findings suggest that targeting the IL-17A/mmu-miR-206-3p pathway may be a new strategy for predicting MIA-related neurodevelopmental deficits and providing preventive interventions. IMPACT Despite the growing evidence of interleukin (IL)-17A and microglia in the pathology of maternal immune activation (MIA), the downstream of IL-17A in microglia is not fully known. IL-17A altered microRNA profiles and upregulated the mmu-miR-206-3p expression in microglia. The mmu-miR-206-3p reduced autism spectrum disorder (ASD) related gene expressions, Hdac4 and Igf1. The Hdac4 expression was also reduced in the brain of MIA offspring. The hsa-miR-206 sequence is consistent with that of mmu-miR-206-3p. This study may provide clues to pathological mechanisms leading to predictions and interventions for ASD children born to mothers with IL-17A-related disorders.
Collapse
Affiliation(s)
- Yukako Iitani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
| | - Rika Miki
- Laboratory of Bell Research Center‑Department of Obstetrics and Gynecology Collaborative Research, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
| | - Kenji Imai
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
| | - Kazuya Fuma
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
| | - Takafumi Ushida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
- Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Aichi, 466‑8560, Japan
| | - Sho Tano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
| | - Kosuke Yoshida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
- Nagoya University Institute for Advanced Research, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Akira Yokoi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
- Nagoya University Institute for Advanced Research, Furo-cho, Chikusa-ku, Nagoya, 464-8603, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan
| | - Tomomi Kotani
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, 466‑8550, Japan.
- Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Aichi, 466‑8560, Japan.
| |
Collapse
|
18
|
Mora P, Chapouly C. Astrogliosis in multiple sclerosis and neuro-inflammation: what role for the notch pathway? Front Immunol 2023; 14:1254586. [PMID: 37936690 PMCID: PMC10627009 DOI: 10.3389/fimmu.2023.1254586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
Multiple sclerosis is an autoimmune inflammatory disease of the central nervous system leading to neurodegeneration. It affects 2.3 million people worldwide, generally younger than 50. There is no known cure for the disease, and current treatment options - mainly immunotherapies to limit disease progression - are few and associated with serious side effects. In multiple sclerosis, disruption of the blood-brain barrier is an early event in the pathogenesis of lesions, predisposing to edema, excito-toxicity and inflammatory infiltration into the central nervous system. Recently, the vision of the blood brain barrier structure and integrity has changed and include contributions from all components of the neurovascular unit, among which astrocytes. During neuro-inflammation, astrocytes become reactive. They undergo morphological and molecular changes named "astrogliosis" driving the conversion from acute inflammatory injury to a chronic neurodegenerative state. Astrogliosis mechanisms are minimally explored despite their significance in regulating the autoimmune response during multiple sclerosis. Therefore, in this review, we take stock of the state of knowledge regarding astrogliosis in neuro-inflammation and highlight the central role of NOTCH signaling in the process of astrocyte reactivity. Indeed, a very detailed nomenclature published in nature neurosciences in 2021, listing all the reactive astrocyte markers fully identified in the literature, doesn't cover the NOTCH signaling. Hence, we discuss evidence supporting NOTCH1 receptor as a central regulator of astrogliosis in the pathophysiology of neuro-inflammation, notably multiple sclerosis, in human and experimental models.
Collapse
Affiliation(s)
- Pierre Mora
- Université de Bordeaux, Institut national de la santé et de la recherche médicale (INSERM), Biology of Cardiovascular Diseases, Pessac, France
| | | |
Collapse
|
19
|
Fathallah S, Abdellatif A, Saadeldin MK. Unleashing nature's potential and limitations: Exploring molecular targeted pathways and safe alternatives for the treatment of multiple sclerosis (Review). MEDICINE INTERNATIONAL 2023; 3:42. [PMID: 37680650 PMCID: PMC10481116 DOI: 10.3892/mi.2023.102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/14/2023] [Indexed: 09/09/2023]
Abstract
Driven by the limitations and obstacles of the available approaches and medications for multiple sclerosis (MS) that still cannot treat the disease, but only aid in accelerating the recovery from its attacks, the use of naturally occurring molecules as a potentially safe and effective treatment for MS is being explored in model organisms. MS is a devastating disease involving the brain and spinal cord, and its symptoms vary widely. Multiple molecular pathways are involved in the pathogenesis of the disease. The present review showcases the recent advancements in harnessing nature's resources to combat MS. By deciphering the molecular pathways involved in the pathogenesis of the disease, a wealth of potential therapeutic agents is uncovered that may revolutionize the treatment of MS. Thus, a new hope can be envisioned in the future, aiming at paving the way toward identifying novel safe alternatives to improve the lives of patients with MS.
Collapse
Affiliation(s)
- Sara Fathallah
- Biotechnology Program, School of Science and Engineering, American University in Cairo, New Cairo 11835, Egypt
| | - Ahmed Abdellatif
- Biotechnology Program, School of Science and Engineering, American University in Cairo, New Cairo 11835, Egypt
- Biology Department, School of Science and Engineering, American University in Cairo, New Cairo 11835, Egypt
| | - Mona Kamal Saadeldin
- Biotechnology Program, School of Science and Engineering, American University in Cairo, New Cairo 11835, Egypt
- Biology Department, School of Science and Engineering, American University in Cairo, New Cairo 11835, Egypt
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
20
|
Wakabayashi C, Kunugi H. Possible involvement of Interleukin-17A in the deterioration of prepulse inhibition on acoustic startle response in mice. Neuropsychopharmacol Rep 2023; 43:365-372. [PMID: 37280178 PMCID: PMC10496063 DOI: 10.1002/npr2.12351] [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: 01/05/2023] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 06/08/2023] Open
Abstract
AIM Proinflammatory cytokines such as interleukin-6 (IL-6) and IL-17A have been implicated in the pathophysiology of schizophrenia which often shows sensorimotor gating abnormalities. This study aimed to examine whether a proinflammatory cytokine, IL-17A, induces impairment in sensorimotor gating in mice. We also examined whether IL-17A administration affects GSK3α/β protein level or phosphorylation in the striatum. METHODS Recombinant mouse IL-17A (low-dose: 0.5 ng/mL and high-dose: 50 ng/mL with 10 μL/g mouse body weight, respectively) or vehicle was intraperitoneally administered into C57BL/6 male mice 10 times in 3 weeks (sub-chronic administration). Prepulse inhibition test using acoustic startle stimulus was conducted 4 weeks after the final IL-17A administration. We evaluated the effect of IL-17A administration on protein level or phosphorylation of GSK3α/β in the striatum by using Western blot analysis. RESULTS Administration of IL-17A induced significant PPI deterioration. Low-dose of IL-17A administration significantly decreased both GSK3α (Ser21) and GSK3β (Ser9) phosphorylation in mouse striatum. There was no significant alteration of GSK3α/β protein levels except for GSK3α in low-dose IL-17A administration group. CONCLUSION We demonstrated for the first time that sub-chronic IL-17A administration induced PPI disruption and that IL-17A administration resulted in decreased phosphorylation of GSKα/β at the striatum. These results suggest that IL-17A could be a target molecule in the prevention and treatment of sensorimotor gating abnormalities observed in schizophrenia.
Collapse
Affiliation(s)
- Chisato Wakabayashi
- Department of Mental Disorder Research, National Institute of NeuroscienceNational Center of Neurology and PsychiatryKodairaJapan
- Faculty of Pharmaceutical SciencesHimeji Dokkyo UniversityHimejiJapan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of NeuroscienceNational Center of Neurology and PsychiatryKodairaJapan
- Department of PsychiatryTeikyo University School of MedicineItabashiJapan
| |
Collapse
|
21
|
Zolfaghari Baghbadorani P, Rayati Damavandi A, Moradi S, Ahmadi M, Bemani P, Aria H, Mottedayyen H, Rayati Damavandi A, Eskandari N, Fathi F. Current advances in stem cell therapy in the treatment of multiple sclerosis. Rev Neurosci 2023; 34:613-633. [PMID: 36496351 DOI: 10.1515/revneuro-2022-0102] [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: 08/10/2022] [Accepted: 11/18/2022] [Indexed: 08/04/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory disease related to the central nervous system (CNS) with a significant global burden. In this illness, the immune system plays an essential role in its pathophysiology and progression. The currently available treatments are not recognized as curable options and, at best, might slow the progression of MS injuries to the CNS. However, stem cell treatment has provided a new avenue for treating MS. Stem cells may enhance CNS healing and regulate immunological responses. Likewise, stem cells can come from various sources, including adipose, neuronal, bone marrow, and embryonic tissues. Choosing the optimal cell source for stem cell therapy is still a difficult verdict. A type of stem cell known as mesenchymal stem cells (MSCs) is obtainable from different sources and has a strong immunomodulatory impact on the immune system. According to mounting data, the umbilical cord and adipose tissue may serve as appropriate sources for the isolation of MSCs. Human amniotic epithelial cells (hAECs), as novel stem cell sources with immune-regulatory effects, regenerative properties, and decreased antigenicity, can also be thought of as a new upcoming contender for MS treatment. Overall, the administration of stem cells in different sets of animal and clinical trials has shown immunomodulatory and neuroprotective results. Therefore, this review aims to discuss the different types of stem cells by focusing on MSCs and their mechanisms, which can be used to treat and improve the outcomes of MS disease.
Collapse
Affiliation(s)
| | - Amirmasoud Rayati Damavandi
- Students' Scientific Research Center, Exceptional Talents Development Center, Tehran University of Medical Sciences, Keshavarz Blvrd, Vesal Shirazi St., Tehran 1417613151, Iran
| | - Samira Moradi
- School of Medicine, Hormozgan University of Medical Sciences Chamran Blvrd., Hormozgan 7919693116, Bandar Abbass, Iran
| | - Meysam Ahmadi
- School of Medicine, Shiraz University of Medical Sciences, Fars, Zand St., Shiraz 7134814336, Iran
| | - Peyman Bemani
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib St., Isfahan 8174673461, Iran
| | - Hamid Aria
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib St., Isfahan 8174673461, Iran
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fars, Ibn Sina Sq., Fasa 7461686688, Iran
| | - Hossein Mottedayyen
- Department of Immunology, School of Medicine, Kashan University of Medical Sciences, Ravandi Blvrd, Isfahan, Kashan 8715988141, Iran
| | - Amirhossein Rayati Damavandi
- Student's Research Committee, Pharmaceutical Sciences Branch, Islamic Azad University, Yakhchal St., Tehran 193951498, Iran
| | - Nahid Eskandari
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib St., Isfahan 8174673461, Iran
| | - Farshid Fathi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Hezar Jerib St., Isfahan 8174673461, Iran
| |
Collapse
|
22
|
Meyer-Arndt L, Kerkering J, Kuehl T, Infante AG, Paul F, Rosiewicz KS, Siffrin V, Alisch M. Inflammatory Cytokines Associated with Multiple Sclerosis Directly Induce Alterations of Neuronal Cytoarchitecture in Human Neurons. J Neuroimmune Pharmacol 2023; 18:145-159. [PMID: 36862362 PMCID: PMC10485132 DOI: 10.1007/s11481-023-10059-w] [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: 08/20/2021] [Accepted: 01/16/2023] [Indexed: 03/03/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) coined by inflammation and neurodegeneration. The actual cause of the neurodegenerative component of the disease is however unclear. We investigated here the direct and differential effects of inflammatory mediators on human neurons. We used embryonic stem cell-derived (H9) human neuronal stem cells (hNSC) to generate neuronal cultures. Neurons were subsequently treated with tumour necrosis factor alpha (TNFα), interferon gamma (IFNγ), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin 17A (IL-17A) and interleukin 10 (IL-10) separately or in combination. Immunofluorescence staining and quantitative polymerase chain reaction (qPCR) were used to assess cytokine receptor expression, cell integrity and transcriptomic changes upon treatment. H9-hNSC-derived neurons expressed cytokine receptors for IFNγ, TNFα, IL-10 and IL-17A. Neuronal exposure to these cytokines resulted in differential effects on neurite integrity parameters with a clear decrease for TNFα- and GM-CSF-treated neurons. The combinatorial treatment with IL-17A/IFNγ or IL-17A/TNFα induced a more pronounced effect on neurite integrity. Furthermore, combinatorial treatments with two cytokines induced several key signalling pathways, i.e. NFκB-, hedgehog and oxidative stress signalling, stronger than any of the cytokines alone. This work supports the idea of immune-neuronal crosstalk and the need to focus on the potential role of inflammatory cytokines on neuronal cytoarchitecture and function.
Collapse
Affiliation(s)
- Lil Meyer-Arndt
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humbolt-Universität Zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humbolt-Universität Zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Janis Kerkering
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Tess Kuehl
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Ana Gil Infante
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Friedemann Paul
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
- NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humbolt-Universität Zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany
| | - Kamil Sebastian Rosiewicz
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Volker Siffrin
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany.
- Department of Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humbolt-Universität Zu Berlin, and Berlin Institute of Health, 10117, Berlin, Germany.
| | - Marlen Alisch
- Experimental and Clinical Research Center (ECRC), Charité - Universitätsmedizin Berlin und Max Delbrück Center for Molecular Medicine in the Helmholtz Association Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
| |
Collapse
|
23
|
Usui N, Kobayashi H, Shimada S. Neuroinflammation and Oxidative Stress in the Pathogenesis of Autism Spectrum Disorder. Int J Mol Sci 2023; 24:ijms24065487. [PMID: 36982559 PMCID: PMC10049423 DOI: 10.3390/ijms24065487] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder (NDD) characterized by impairments in social communication, repetitive behaviors, restricted interests, and hyperesthesia/hypesthesia caused by genetic and/or environmental factors. In recent years, inflammation and oxidative stress have been implicated in the pathogenesis of ASD. In this review, we discuss the inflammation and oxidative stress in the pathophysiology of ASD, particularly focusing on maternal immune activation (MIA). MIA is a one of the common environmental risk factors for the onset of ASD during pregnancy. It induces an immune reaction in the pregnant mother’s body, resulting in further inflammation and oxidative stress in the placenta and fetal brain. These negative factors cause neurodevelopmental impairments in the developing fetal brain and subsequently cause behavioral symptoms in the offspring. In addition, we also discuss the effects of anti-inflammatory drugs and antioxidants in basic studies on animals and clinical studies of ASD. Our review provides the latest findings and new insights into the involvements of inflammation and oxidative stress in the pathogenesis of ASD.
Collapse
Affiliation(s)
- Noriyoshi Usui
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
- United Graduate School of Child Development, Osaka University, Suita 565-0871, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Suita 565-0871, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
- Correspondence: ; Tel.: +81-668-79-3124
| | - Hikaru Kobayashi
- SANKEN (Institute of Scientific and Industrial Research), Osaka University, Suita 567-0047, Japan
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
- United Graduate School of Child Development, Osaka University, Suita 565-0871, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Suita 565-0871, Japan
- Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
| |
Collapse
|
24
|
Sun JL, Dai WJ, Shen XY, Lü N, Zhang YQ. Interleukin-17 is involved in neuropathic pain and spinal synapse plasticity on mice. J Neuroimmunol 2023; 377:578068. [PMID: 36948094 DOI: 10.1016/j.jneuroim.2023.578068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/08/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023]
Abstract
Neuropathic pain seriously affects people's life, but its mechanism is not clear. Interleukin-17 (IL-17) is a proinflammation cytokine and involved in pain regulation. Our previous study found that IL-17 markedly enhanced the excitatory activity of spinal dorsal neurons in mice spinal slices. The present study attempts to explore if IL-17 contributes to neuropathic pain and spinal synapse plasticity. A model of spared nerve injury (SNI) was established in C57BL/6 J mice and IL-17a mutant mice. The pain-like behaviors was tested by von Frey test and dynamic mechanical stimuli, and the expression of IL-17 and its receptor, IL-17RA, was detected by immunohistochemical staining. C-fiber evoked field potentials were recorded in vivo. In the spinal dorsal horn, IL-17 predominantly expressed in the superficial spinal astrocytes and IL-17RA expressed mostly in neurons and slightly in astrocytes. The SNI-induced static and dynamic allodynia was significantly prevented by pretreatment of neutralizing IL-17 antibody (intrathecal injection, 2 μg/10 μL) and attenuated in IL-17a mutant mice. Post-treatment of IL-17 neutralizing antibody also partially relieved the established mechanical allodynia. Moreover, spinal long-term potentiation (LTP) of C-fiber evoked field potentials, a substrate for central sensitization, was suppressed by IL-17 neutralizing antibody. Intrathecal injection of IL-17 recombinant protein (0.2 μg/10 μL) mimicked the mechanical allodynia and facilitated the spinal LTP. These data implied that IL-17 in the spinal cord played a crucial role in neuropathic pain and central sensitization.
Collapse
Affiliation(s)
- Jia-Lu Sun
- Department of Translational Neuroscience, Jing'an District Center Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Wen-Jing Dai
- Department of Translational Neuroscience, Jing'an District Center Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Xin-Yuan Shen
- Department of Translational Neuroscience, Jing'an District Center Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Ning Lü
- Department of Translational Neuroscience, Jing'an District Center Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| | - Yu-Qiu Zhang
- Department of Translational Neuroscience, Jing'an District Center Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
25
|
Singh Gautam A, Kumar Singh R. Therapeutic potential of targeting IL-17 and its receptor signaling in neuroinflammation. Drug Discov Today 2023; 28:103517. [PMID: 36736763 DOI: 10.1016/j.drudis.2023.103517] [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: 08/27/2022] [Revised: 12/26/2022] [Accepted: 01/26/2023] [Indexed: 02/04/2023]
Abstract
T helper 17 cells are thought to significantly contribute to the neuroinflammation process during neurogenerative diseases via their signature cytokine, interleukin (IL)-17. Recently, an emerging key role of IL-17 and its receptors has been documented in inflammatory and autoimmune diseases. The clinical studies conducted on patients with neurodegenerative disease have also shown an increase in IL-17 levels in serum as well as cerebrospinal fluid samples. Therapeutic targeting of either IL-17 receptors or direct IL-17 neutralizing antibodies has shown a promising preclinical and clinical proof of concept for treating chronic autoimmune neurodegenerative diseases such as multiple sclerosis. Thus, IL-17 and its receptors have a central role in regulation of neuroinflammation and can be considered as one of the major therapeutic targets in chronic neuroinflammatory diseases.
Collapse
Affiliation(s)
- Avtar Singh Gautam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Transit Campus, Bijnour-sisendi Road, Sarojini Nagar, Lucknow 226002, Uttar Pradesh, India
| | - Rakesh Kumar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Transit Campus, Bijnour-sisendi Road, Sarojini Nagar, Lucknow 226002, Uttar Pradesh, India.
| |
Collapse
|
26
|
Liu S, Deng S, Ding Y, Flores JJ, Zhang X, Jia X, Hu X, Peng J, Zuo G, Zhang JH, Gong Y, Tang J. Secukinumab attenuates neuroinflammation and neurobehavior defect via PKCβ/ERK/NF-κB pathway in a rat model of GMH. Exp Neurol 2023; 360:114276. [PMID: 36402169 DOI: 10.1016/j.expneurol.2022.114276] [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: 03/24/2022] [Revised: 11/05/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022]
Abstract
AIMS Germinal matrix hemorrhage (GMH) is a disastrous clinical event for newborns. Neuroinflammation plays an important role in the development of neurological deficits after GMH. The purpose of this study is to investigate the anti-inflammatory role of secukinumab after GMH and its underlying mechanisms involving PKCβ/ERK/NF-κB signaling pathway. METHODS A total of 154 Sprague-Dawley P7 rat pups were used. GMH was induced by intraparenchymal injection of bacterial collagenase. Secukinumab was administered intranasally post-GMH. PKCβ activator PMA and p-ERK activator Ceramide C6 were administered intracerebroventricularly at 24 h prior to GMH induction, respectively. Neurobehavioral tests, western blot and immunohistochemistry were used to evaluate the efficacy of Secukinumab in both short-term and long-term studies. RESULTS Endogenous IL-17A, IL-17RA, PKCβ and p-ERK were increased after GMH. Secukinumab treatment improved short- and long-term neurological outcomes, reduced the synthesis of MPO and Iba-1 in the perihematoma area, and inhibited the synthesis of proinflammatory factors, such as NF-κB, IL-1β, TNF-α and IL-6. Additionally, PMA and ceramide C6 abolished the beneficial effects of Secukinumab. CONCLUSION Secukinumab treatment suppressed neuroinflammation and attenuated neurological deficits after GMH, which was mediated through the downregulation of the PKCβ/ERK/NF-κB pathway. Secukinumab treatment may provide a promising therapeutic strategy for GMH patients.
Collapse
Affiliation(s)
- Shengpeng Liu
- Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, China; Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Guangdong, China
| | - Shuixiang Deng
- Department of Critical Care Medicine, HuaShan Hospital, Fudan University, Shanghai 200040, China; Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Yan Ding
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Jerry J Flores
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Xiaoli Zhang
- Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, China; Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Guangdong, China
| | - Xiaojing Jia
- Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, China; Department of Pediatrics, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Guangdong, China
| | - Xiao Hu
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Jun Peng
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Gang Zuo
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA; Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Ye Gong
- Department of Critical Care Medicine, HuaShan Hospital, Fudan University, Shanghai 200040, China; Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Jiping Tang
- Department of Physiology and Pharmacology, Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| |
Collapse
|
27
|
Feng W, Zhang Y, Sun P, Xiao M. Acquired immunity and Alzheimer's disease. J Biomed Res 2023; 37:15-29. [PMID: 36165328 PMCID: PMC9898041 DOI: 10.7555/jbr.36.20220083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by progressive cognitive defects. The role of the central immune system dominated by microglia in the progression of AD has been extensively investigated. However, little is known about the peripheral immune system in AD pathogenesis. Recently, with the discovery of the meningeal lymphatic vessels and glymphatic system, the roles of the acquired immunity in the maintenance of central homeostasis and neurodegenerative diseases have attracted an increasing attention. The T cells not only regulate the function of neurons, astrocytes, microglia, oligodendrocytes and brain microvascular endothelial cells, but also participate in the clearance of β-amyloid (Aβ) plaques. Apart from producing antibodies to bind Aβ peptides, the B cells affect Aβ-related cascades via a variety of antibody-independent mechanisms. This review systemically summarizes the recent progress in understanding pathophysiological roles of the T cells and B cells in AD.
Collapse
Affiliation(s)
- Weixi Feng
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China,Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Chinese Academy of Sciences, Shanghai 200031, China,Weixi Feng, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, 101 Longmian Avenue, Jiangning District, Nanjing, Jiangsu 211166, China. Tel: +86-25-86869338; E-mail:
| | - Yanli Zhang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China,Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Peng Sun
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China,Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu 211166, China,Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Chinese Academy of Sciences, Shanghai 200031, China,Brain Institute, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
28
|
Mohamed YT, Salama A, Rabie MA, Abd El Fattah MA. Neuroprotective effect of secukinumab against rotenone induced Parkinson's disease in rat model: Involvement of IL-17, HMGB-1/TLR4 axis and BDNF/TrKB cascade. Int Immunopharmacol 2023; 114:109571. [PMID: 36527875 DOI: 10.1016/j.intimp.2022.109571] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/23/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
Neuroinflammatory status produced via activation of toll like receptor-4 (TLR-4) and interleukin-17 receptor (IL-17R) is one of the principal mechanisms involved in dopaminergic neuronal loss in Parkinson's disease (PD). Activation of TLR-4 and IL-17R stimulates reactive oxygen species (ROS) and proinflammatory cytokines (IL-17, IL-1β, TNFα, IL-6) production that augments neurodegeneration and reduces neuro-survival axis (TrKB/Akt/CREB/BDNF). So, reducing IL-17-driven neuroinflammation via secukinumab, monoclonal antibody against IL-17A, may be one of therapeutic approach for PD. Moreover, the aim was extended to delineate the possible neuroprotective mechanism involved against neuronal loss in rotenone induced PD in rats. Rats received 11 subcutaneous injection of rotenone (1.5 mg/kg) every other day for 21 consecutive days and treated with 2 subcutaneous injections of secukinumab (15 mg/kg) on day 9 and 15, one hour after rotenone administration. Treatment with secukinumab improved motor impairment and muscle incoordination induced by rotenone, as verified by open field and rotarod tests. Moreover, secukinumab attenuated neuronal loss and improve histopathological profile. Noteworthy, secukinumab reduces neuro-inflammatory status by hindering the interaction between IL and 17A and IL-17RA together with inhibiting the activation of TLR-4 and its downstream cascade including pS536-NFκB p65, IL-1β and HMGB-1. Additionally, secukinumab stimulated neuro-survival signalling cascade via activation pY515-TrKB receptor and triggered upsurge in its downstream targets (pS473-Akt/pS133-CREB/BDNF). Furthermore, secukinumab increased striatal tyrosine hydroxylase immunoexpression, the rate limiting step in dopamine biosynthesis, to guard against dopaminergic neuronal loss. In conclusion, secukinumab exerts a neuroprotective effect against rotenone induced neuronal loss via inhibition IL17A/IL17RA interaction and HMGB-1/TLR-4/NF-κBp65/IL1β signalling cascade, together with activation of TrKB/ Akt/CREB/BDNF axis.
Collapse
Affiliation(s)
- Yara T Mohamed
- Maintenance & Calibration unit, Technical Support Department, National Organization of Research & Biologicals, Egyptian Drug Authority, Giza, Egypt
| | - Abeer Salama
- Department of Pharmacology, National Research Centre, 33 El Buhouth St., Dokki, Cairo, 12622, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Mai A Abd El Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
29
|
Otero AM, Antonson AM. At the crux of maternal immune activation: Viruses, microglia, microbes, and IL-17A. Immunol Rev 2022; 311:205-223. [PMID: 35979731 PMCID: PMC9804202 DOI: 10.1111/imr.13125] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Inflammation during prenatal development can be detrimental to neurodevelopmental processes, increasing the risk of neuropsychiatric disorders. Prenatal exposure to maternal viral infection during pregnancy is a leading environmental risk factor for manifestation of these disorders. Preclinical animal models of maternal immune activation (MIA), established to investigate this link, have revealed common immune and microbial signaling pathways that link mother and fetus and set the tone for prenatal neurodevelopment. In particular, maternal intestinal T helper 17 cells, educated by endogenous microbes, appear to be key drivers of effector IL-17A signals capable of reaching the fetal brain and causing neuropathologies. Fetal microglial cells are particularly sensitive to maternally derived inflammatory and microbial signals, and they shift their functional phenotype in response to MIA. Resulting cortical malformations and miswired interneuron circuits cause aberrant offspring behaviors that recapitulate core symptoms of human neurodevelopmental disorders. Still, the popular use of "sterile" immunostimulants to initiate MIA has limited translation to the clinic, as these stimulants fail to capture biologically relevant innate and adaptive inflammatory sequelae induced by live pathogen infection. Thus, there is a need for more translatable MIA models, with a focus on relevant pathogens like seasonal influenza viruses.
Collapse
Affiliation(s)
- Ashley M. Otero
- Neuroscience ProgramUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Adrienne M. Antonson
- Department of Animal SciencesUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
30
|
Jiang X, Zhou R, Zhang Y, Zhu T, Li Q, Zhang W. Interleukin-17 as a potential therapeutic target for chronic pain. Front Immunol 2022; 13:999407. [PMID: 36248896 PMCID: PMC9556763 DOI: 10.3389/fimmu.2022.999407] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic pain remains to be a clinical challenge and is recognized as a major health problem with varying impacts on quality of life. Currently, the first-line therapy for chronic pain is opioids, which are often accompanied by unwanted psychoactive side effects. Thus, new and effective treatments for chronic pain are urgently needed and eagerly pursued. Inflammatory cytokines, especially interleukin-17 (IL-17), are reportedly potential therapeutic targets owing to their pivotal role in chronic pain from the neuroinflammation perspective. Recently, substantial evidence confirmed that IL-17 and IL-17 receptors (IL-17Rs) were increased in neuropathic, inflammatory, and cancer pain models. Notably, IL-17/IL-17R antibodies also reportedly relieve or cure inflammatory- and pain-related diseases. However, existing studies have reported controversial results regarding IL-17/IL-17Rs as potential therapeutic targets in diverse animal models of chronic pain. In this review, we present a summary of published studies and discuss the evidence, from basic to clinical to research, regarding the role and mechanism of action between IL-17 and diverse kinds of chronic pain in animal models and clinical patients. Furthermore, we evaluated IL-17-based therapy as a potential therapeutic strategy for inflammatory- and pain-related disease. Importantly, we also discussed clinical trials of IL-17/IL-17R targeting monoclonal antibodies. Overall, we found that IL-17 is a potential therapeutic target for chronic pain from the perspective of neuroinflammation.
Collapse
Affiliation(s)
- Xiaojuan Jiang
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu, China
| | - Ruihao Zhou
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu, China
| | - Yujun Zhang
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Zhu
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Li
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Qian Li, ; Weiyi Zhang,
| | - Weiyi Zhang
- Department of Anesthesiology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Qian Li, ; Weiyi Zhang,
| |
Collapse
|
31
|
Park JC, Im SH. The gut-immune-brain axis in neurodevelopment and neurological disorders. MICROBIOME RESEARCH REPORTS 2022; 1:23. [PMID: 38046904 PMCID: PMC10688819 DOI: 10.20517/mrr.2022.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
The gut-brain axis is gaining momentum as an interdisciplinary field addressing how intestinal microbes influence the central nervous system (CNS). Studies using powerful tools, including germ-free, antibiotic-fed, and fecal microbiota transplanted mice, demonstrate how gut microbiota perturbations alter the fate of neurodevelopment. Probiotics are also becoming more recognized as potentially effective therapeutic agents in alleviating symptoms of neurological disorders. While gut microbes may directly communicate with the CNS through their effector molecules, including metabolites, their influence on neuroimmune populations, including newly discovered brain-resident T cells, underscore the host immunity as a potent mediator of the gut-brain axis. In this review, we examine the unique immune populations within the brain, the effects of the gut microbiota on the CNS, and the efficacy of specific probiotic strains to propose the novel concept of the gut-immune-brain axis.
Collapse
Affiliation(s)
- John Chulhoon Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea.
| | - Sin-Hyeog Im
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea.
- Institute for Convergence Research and Education, Yonsei University, Seoul 03722, Republic of Korea
- ImmunoBiome Inc., POSTECH Biotech Center, Pohang 37673, Republic of Korea
| |
Collapse
|
32
|
Wen Y, Feng S, Dai H, Mao M, Zhou Z, Li B, Wang C, Cai X, Li S, Yang J, Ren Q, Sun J. Intestinal dysbacteriosis-propelled T helper 17 cells activation mediate the perioperative neurocognitive disorder induced by anesthesia/surgery in aged rats. Neurosci Lett 2022; 783:136741. [PMID: 35716962 DOI: 10.1016/j.neulet.2022.136741] [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: 02/08/2022] [Revised: 04/18/2022] [Accepted: 06/13/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Perioperative neurocognitive disorders (PND) is a common postoperative disease in elderly patients, but its pathogenesis remains unclear. METHODS Exploratory laparotomy was performed to establish PND model under sevoflurane anesthesia. 16S rRNA high-throughput sequencing was used to detect the changes of intestinal flora. Antibiotics were used to relatively eliminate intestinal flora before anesthesia/surgery, and behavior tests, such as open field, Y maze, and fear conditioning tests were applied to detect the changes of memory ability. The number of Th17 cells and Foxp3 cells was detected by flow cytometry in the Peyer's patches (PP), mesenteric lymph nodes (MLN), blood and brain. Western blot was used to detect the expression of IL17, IL17RA, IL6 and IL10 in the hippocampus. Immunofluorescence was used to detect the expression of IL17, IL17R and IBA1 (ionized calcium binding adaptor molecule1) in the hippocampus. RESULTS Anesthesia/surgery caused intestinal flora imbalance and induced neurocognitive impairment, increased the number of Th17 cells in the PP, MLN, blood and brain, increased the level of IL17, IL17R and inflammatory factors production in the hippocampus. Antibiotics administration before anesthesia/surgery significantly decreased the number of Th17 cells and the level of IL17, IL17R and inflammatory factors production, and improved the memory function. In addition, we found that IL17R was co-labeled with IBA1 in a large amount in the hippocampus through immunofluorescence double-staining. CONCLUSION Our study suggested that intestinal dysbacteriosis-propelled T helper 17 cells activation and IL17 secretion might play an important role in the pathogenesis of PND induced by anesthesia/surgery in aged rats.
Collapse
Affiliation(s)
- Yazhou Wen
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China; Department of Anesthesiology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, Jiangsu 210004, China
| | - Shanwu Feng
- Department of Anesthesiology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, Jiangsu 210004, China
| | - Hongyu Dai
- Department of Anesthesiology, Nanjing Medical University, Nanjing, Jiangsu 210004, China
| | - Meng Mao
- Department of Anesthesiology, Nanjing Medical University, Nanjing, Jiangsu 210004, China
| | - Zhenhui Zhou
- Department of Anesthesiology, Nanjing Medical University, Nanjing, Jiangsu 210004, China
| | - Bin Li
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Chaoran Wang
- Department of Anesthesiology, Nanjing Medical University, Nanjing, Jiangsu 210004, China
| | - Xuechun Cai
- Department of Anesthesiology, Nanjing Medical University, Nanjing, Jiangsu 210004, China
| | - Shuming Li
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Jiaojiao Yang
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Quan Ren
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Jie Sun
- Department of Anesthesiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
33
|
Brummer T, Zipp F, Bittner S. T cell-neuron interaction in inflammatory and progressive multiple sclerosis biology. Curr Opin Neurobiol 2022; 75:102588. [PMID: 35732103 DOI: 10.1016/j.conb.2022.102588] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/18/2022] [Accepted: 05/18/2022] [Indexed: 11/03/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune condition of the central nervous system (CNS) characterized by acute inflammatory relapses, chronic neuro-axonal degeneration, and subsequent disability progression. T cells - in interaction with B cells and CNS-resident glial cells - are key initiators and drivers of neurodegeneration in MS. However, it is not entirely clear how encephalitogenic T cells orchestrate the local immune response within the brain and how they overtake disease stage-specific roles in MS pathogenesis. This review highlights recent advances in understanding direct and indirect T cell-neuron interactions in inflammatory and progressive MS. Finally, we discuss new diagnostic tools such as neurofilament light chain (NfL), which is on the cusp of becoming a key factor in clinical and therapeutic decision-making.
Collapse
Affiliation(s)
- Tobias Brummer
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Frauke Zipp
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience (FTN) and Immunotherapy (FZI), Rhine Main Neuroscience Network (rmn(2)), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
34
|
Zhang Y, Lian L, Fu R, Liu J, Shan X, Jin Y, Xu S. Microglia: The Hub of Intercellular Communication in Ischemic Stroke. Front Cell Neurosci 2022; 16:889442. [PMID: 35518646 PMCID: PMC9062186 DOI: 10.3389/fncel.2022.889442] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/30/2022] [Indexed: 12/14/2022] Open
Abstract
Communication between microglia and other cells has recently been at the forefront of research in central nervous system (CNS) disease. In this review, we provide an overview of the neuroinflammation mediated by microglia, highlight recent studies of crosstalk between microglia and CNS resident and infiltrating cells in the context of ischemic stroke (IS), and discuss how these interactions affect the course of IS. The in-depth exploration of microglia-intercellular communication will be beneficial for therapeutic tools development and clinical translation for stroke control.
Collapse
Affiliation(s)
- Yunsha Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, China
| | - Lu Lian
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rong Fu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jueling Liu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoqian Shan
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yang Jin
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| |
Collapse
|
35
|
Beurel E, Medina-Rodriguez EM, Jope RS. Targeting the Adaptive Immune System in Depression: Focus on T Helper 17 Cells. Pharmacol Rev 2022; 74:373-386. [PMID: 35302045 PMCID: PMC8973514 DOI: 10.1124/pharmrev.120.000256] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
There is a vital need to understand mechanisms contributing to susceptibility to depression to improve treatments for the 11% of Americans who currently suffer from this debilitating disease. The adaptive immune system, comprising T and B cells, has emerged as a potential contributor to depression, as demonstrated in the context of lymphopenic mice. Overall, patients with depression have reduced circulating T and regulatory B cells, "immunosuppressed" T cells, and alterations in the relative abundance of T cell subtypes. T helper (Th) cells have the capacity to differentiate to various lineages depending on the cytokine environment, antigen stimulation, and costimulation. Regulatory T cells are decreased, and the Th1/Th2 ratio and the Th17 cells are increased in patients with depression. Evidence for changes in each Th lineage has been reported to some extent in patients with depression. However, the evidence is strongest for the association of depression with changes in Th17 cells. Th17 cells produce the inflammatory cytokine interleukin (IL)-17A, and the discovery of Th17 cell involvement in depression evolved from the well established link that IL-6, which is required for Th17 cell differentiation, contributes to the onset, and possibly maintenance, of depression. One intriguing action of Th17 cells is their participation in the gut-brain axis to mediate stress responses. Although the mechanisms of action of Th17 cells in depression remain unclear, neutralization of IL-17A by anti-IL-17A antibodies, blocking stress-induced production, or release of gut Th17 cells represent feasible therapeutic approaches and might provide a new avenue to improve depression symptoms. SIGNIFICANCE STATEMENT: Th17 cells appear as a promising therapeutic target for depression, for which efficacious therapeutic options are limited. The use of neutralizing antibodies targeting Th17 cells has provided encouraging results in depressed patients with comorbid autoimmune diseases.
Collapse
Affiliation(s)
- Eléonore Beurel
- Department of Psychiatry and Behavioral Sciences (E.B., E.M.M.-R., R.S.J.) and Department of Biochemistry and Molecular Biology Miller School of Medicine (E.B., R.S.J.), University of Miami, Miami, Florida and Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida (E.M.M.-R., R.S.J.)
| | - Eva M Medina-Rodriguez
- Department of Psychiatry and Behavioral Sciences (E.B., E.M.M.-R., R.S.J.) and Department of Biochemistry and Molecular Biology Miller School of Medicine (E.B., R.S.J.), University of Miami, Miami, Florida and Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida (E.M.M.-R., R.S.J.)
| | - Richard S Jope
- Department of Psychiatry and Behavioral Sciences (E.B., E.M.M.-R., R.S.J.) and Department of Biochemistry and Molecular Biology Miller School of Medicine (E.B., R.S.J.), University of Miami, Miami, Florida and Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, Florida (E.M.M.-R., R.S.J.)
| |
Collapse
|
36
|
Kunkl M, Amormino C, Tedeschi V, Fiorillo MT, Tuosto L. Astrocytes and Inflammatory T Helper Cells: A Dangerous Liaison in Multiple Sclerosis. Front Immunol 2022; 13:824411. [PMID: 35211120 PMCID: PMC8860818 DOI: 10.3389/fimmu.2022.824411] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/13/2022] [Indexed: 11/15/2022] Open
Abstract
Multiple Sclerosis (MS) is a neurodegenerative autoimmune disorder of the central nervous system (CNS) characterized by the recruitment of self-reactive T lymphocytes, mainly inflammatory T helper (Th) cell subsets. Once recruited within the CNS, inflammatory Th cells produce several inflammatory cytokines and chemokines that activate resident glial cells, thus contributing to the breakdown of blood-brain barrier (BBB), demyelination and axonal loss. Astrocytes are recognized as key players of MS immunopathology, which respond to Th cell-defining cytokines by acquiring a reactive phenotype that amplify neuroinflammation into the CNS and contribute to MS progression. In this review, we summarize current knowledge of the astrocytic changes and behaviour in both MS and experimental autoimmune encephalomyelitis (EAE), and the contribution of pathogenic Th1, Th17 and Th1-like Th17 cell subsets, and CD8+ T cells to the morphological and functional modifications occurring in astrocytes and their pathological outcomes.
Collapse
Affiliation(s)
- Martina Kunkl
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Carola Amormino
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Valentina Tedeschi
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Maria Teresa Fiorillo
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| |
Collapse
|
37
|
Krishnarajah S, Becher B. T H Cells and Cytokines in Encephalitogenic Disorders. Front Immunol 2022; 13:822919. [PMID: 35320935 PMCID: PMC8934849 DOI: 10.3389/fimmu.2022.822919] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/15/2022] [Indexed: 12/14/2022] Open
Abstract
The invasion of immune cells into the central nervous system (CNS) is a hallmark of the process we call neuroinflammation. Diseases such as encephalitides or multiple sclerosis (MS) are characterised by the dramatic influx of T lymphocytes and monocytes. The communication between inflammatory infiltrates and CNS resident cells is primarily mediated through cytokines. Over the years, numerous cytokine networks have been assessed to better understand the development of immunopathology in neuroinflammation. In MS for instance, many studies have shown that CD4+ T cells infiltrate the CNS and subsequently lead to immunopathology. Inflammatory CD4+ T cells, such as TH1, TH17, GM-CSF-producing helper T cells are big players in chronic neuroinflammation. Conversely, encephalitogenic or meningeal regulatory T cells (TREGs) and TH2 cells have been shown to drive a decrease in inflammatory functions in microglial cells and thus promote a neuroprotective microenvironment. Recent studies report overlapping as well as differential roles of these cells in tissue inflammation. Taken together, this suggests a more complex relationship between effector T cell subsets in neuroinflammation than has hitherto been established. In this overview, we review the interplay between helper T cell subsets infiltrating the CNS and how they actively contribute to neuroinflammation and degeneration. Importantly, in this context, we will especially focus on the current knowledge regarding the contribution of various helper cell subsets to neuroinflammation by referring to their helper T cell profile in the context of their target cell.
Collapse
|
38
|
Lee TL, Tsai TF. Non-immune functions of inflammatory cytokines targeted by anti-psoriatic biologics: a review. Inflamm Res 2022; 71:157-168. [PMID: 34981130 DOI: 10.1007/s00011-021-01528-0] [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: 08/13/2021] [Revised: 11/25/2021] [Accepted: 11/25/2021] [Indexed: 12/01/2022] Open
Abstract
PURPOSE Psoriasis is an inflammatory disease characterized by skin thickening with silvery white desquamation due to dysregulated inflammatory pathways and elevated levels of inflammatory cytokines. Biologic agents targeting these inflammatory cytokines have brought about significant improvement in clearing psoriatic lesions in patients with moderate-to-severe psoriasis. Moreover, biologics exert both beneficial and detrimental effects on comorbidities in psoriasis, which include increased risk of cardiovascular events, metabolic syndrome, among other conditions. However, non-immune functions of cytokines targeted by biologics, and, hence, the potential risks and benefits of biologics for psoriasis to different organs/systems and comorbidities, have not been well elucidated. RESULTS This review summarizes current understanding of the pathogenesis of psoriasis-related comorbidities and emerging discoveries of roles of cytokines targeted in psoriasis treatment, including tumor necrosis factor α and interleukins 12, 23, and 17, aiming to complete the safety profile of each biologics and provide therapeutic implications on psoriasis-related comorbidities, and on diseases involving other organs or systems.
Collapse
Affiliation(s)
- Tung-Lin Lee
- Department of Medical Education, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsen-Fang Tsai
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, 7 Chung-Shan S. Rd., Taipei, 100, Taiwan.
| |
Collapse
|
39
|
Fujitani M, Miyajima H, Otani Y, Liu X. Maternal and Adult Interleukin-17A Exposure and Autism Spectrum Disorder. Front Psychiatry 2022; 13:836181. [PMID: 35211045 PMCID: PMC8861354 DOI: 10.3389/fpsyt.2022.836181] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/14/2022] [Indexed: 11/26/2022] Open
Abstract
Epidemiological evidence in humans has suggested that maternal infections and maternal autoimmune diseases are involved in the pathogenesis of autism spectrum disorder. Animal studies supporting human results have shown that maternal immune activation causes brain and behavioral alterations in offspring. Several underlying mechanisms, including interleukin-17A imbalance, have been identified. Apart from the pro-inflammatory effects of interleukin-17A, there is also evidence to support the idea that it activates neuronal function and defines cognitive behavior. In this review, we examined the signaling pathways in both immunological and neurological contexts that may contribute to the improvement of autism spectrum disorder symptoms associated with maternal blocking of interleukin-17A and adult exposure to interleukin-17A. We first describe the epidemiology of maternal immune activation then focus on molecular signaling of the interleukin-17 family regarding its physiological and pathological roles in the embryonic and adult brain. In the future, it may be possible to use interleukin-17 antibodies to prevent autism spectrum disorder.
Collapse
Affiliation(s)
- Masashi Fujitani
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shimane University, Shimane, Japan
| | - Hisao Miyajima
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shimane University, Shimane, Japan
| | - Yoshinori Otani
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shimane University, Shimane, Japan
| | - Xinlang Liu
- Department of Anatomy and Neuroscience, Faculty of Medicine, Shimane University, Shimane, Japan
| |
Collapse
|
40
|
The Efficacy of Fecal Microbiota Transplantation in Experimental Autoimmune Encephalomyelitis: Transcriptome and Gut Microbiota Profiling. J Immunol Res 2021; 2021:4400428. [PMID: 34938813 PMCID: PMC8687821 DOI: 10.1155/2021/4400428] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/19/2021] [Accepted: 11/25/2021] [Indexed: 12/14/2022] Open
Abstract
Objective To study the protective effect of fecal microbiota transplantation (FMT) on experimental autoimmune encephalomyelitis (EAE) and reveal its potential intestinal microflora-dependent mechanism through analyses of the intestinal microbiota and spinal cord transcriptome in mice. Method We measured the severity of disease by clinical EAE scores and H&E staining. Gut microbiota alteration in the gut and differentially expressed genes (DEGs) in the spinal cord were analyzed through 16S rRNA and transcriptome sequencing. Finally, we analyzed associations between the relative abundance of intestinal microbiota constituents and DEGs. Results We observed that clinical EAE scores were lower in the EAE+FMT group than in the EAE group. Meanwhile, mice in the EAE+FMT group also had a lower number of infiltrating cells. The results of 16S rRNA sequence analysis showed that FMT increased the relative abundance of Firmicutes and Proteobacteria and reduced the abundance of Bacteroides and Actinobacteria. Meanwhile, FMT could modulate gut microbiota balance, especially via increasing the relative abundance of g_Adlercreutzia, g_Sutterella, g_Prevotella_9, and g_Tyzzerella_3 and decreasing the relative abundance of g_Turicibacter. Next, we analyzed the transcriptome of mouse spinal cord tissue and found that 1476 genes were differentially expressed between the EAE and FMT groups. The analysis of these genes showed that FMT mainly participated in the inflammatory response. Correlation analysis between gut microbes and transcriptome revealed that the relative abundance of Adlercreutzia was correlated with the expression of inflammation-related genes negatively, including Casp6, IL1RL2 (IL-36R), IL-17RA, TNF, CCL3, CCR5, and CCL8, and correlated with the expression of neuroprotection-related genes positively, including Snap25, Edil3, Nrn1, Cpeb3, and Gpr37. Conclusion Altogether, FMT may selectively regulate gene expression to improve inflammation and maintain the stability of the intestinal environment in a gut microbiota-dependent manner.
Collapse
|
41
|
Cao Y, Yu Y, Xue B, Wang Y, Chen X, Beltz TG, Johnson AK, Wei SG. IL (Interleukin)-17A Acts in the Brain to Drive Neuroinflammation, Sympathetic Activation, and Hypertension. Hypertension 2021; 78:1450-1462. [PMID: 34628936 DOI: 10.1161/hypertensionaha.121.18219] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Yiling Cao
- Department of Internal Medicine (Y.C., Y.Y., S.-G.W.), University of Iowa Carver College of Medicine
| | - Yang Yu
- Department of Internal Medicine (Y.C., Y.Y., S.-G.W.), University of Iowa Carver College of Medicine
| | - Baojian Xue
- Psychological and Brain Sciences (B.X., T.G.B., A.K.J.), University of Iowa Carver College of Medicine
| | - Ye Wang
- Department of Cardiology, the First Affiliated Hospital of Shandong First Medical University, China (Y.W.)
| | - Xiaolei Chen
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China (X.C.)
| | - Terry G Beltz
- Psychological and Brain Sciences (B.X., T.G.B., A.K.J.), University of Iowa Carver College of Medicine
| | - Alan Kim Johnson
- Psychological and Brain Sciences (B.X., T.G.B., A.K.J.), University of Iowa Carver College of Medicine.,Abboud Cardiovascular Research Center (A.K.J., S.-G.W.), University of Iowa Carver College of Medicine.,Iowa Neuroscience Institute (A.K.J., S.-G.W.), University of Iowa Carver College of Medicine
| | - Shun-Guang Wei
- Department of Internal Medicine (Y.C., Y.Y., S.-G.W.), University of Iowa Carver College of Medicine.,Abboud Cardiovascular Research Center (A.K.J., S.-G.W.), University of Iowa Carver College of Medicine.,Department of Cardiology, the First Affiliated Hospital of Shandong First Medical University, China (Y.W.)
| |
Collapse
|
42
|
Brigas HC, Ribeiro M, Coelho JE, Gomes R, Gomez-Murcia V, Carvalho K, Faivre E, Costa-Pereira S, Darrigues J, de Almeida AA, Buée L, Dunot J, Marie H, Pousinha PA, Blum D, Silva-Santos B, Lopes LV, Ribot JC. IL-17 triggers the onset of cognitive and synaptic deficits in early stages of Alzheimer's disease. Cell Rep 2021; 36:109574. [PMID: 34469732 DOI: 10.1016/j.celrep.2021.109574] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 06/09/2021] [Accepted: 07/30/2021] [Indexed: 11/18/2022] Open
Abstract
Neuroinflammation in patients with Alzheimer's disease (AD) and related mouse models has been recognized for decades, but the contribution of the recently described meningeal immune population to AD pathogenesis remains to be addressed. Here, using the 3xTg-AD model, we report an accumulation of interleukin-17 (IL-17)-producing cells, mostly γδ T cells, in the brain and the meninges of female, but not male, mice, concomitant with the onset of cognitive decline. Critically, IL-17 neutralization into the ventricles is sufficient to prevent short-term memory and synaptic plasticity deficits at early stages of disease. These effects precede blood-brain barrier disruption and amyloid-beta or tau pathology, implying an early involvement of IL-17 in AD pathology. When IL-17 is neutralized at later stages of disease, the onset of short-memory deficits and amyloidosis-related splenomegaly is delayed. Altogether, our data support the idea that cognition relies on a finely regulated balance of "inflammatory" cytokines derived from the meningeal immune system.
Collapse
Affiliation(s)
- Helena C Brigas
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Miguel Ribeiro
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Joana E Coelho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Rui Gomes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal; Faculdade de Ciências de Lisboa, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Victoria Gomez-Murcia
- Université Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, 59000 Lille, France; Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Kevin Carvalho
- Université Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, 59000 Lille, France; Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Emilie Faivre
- Université Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, 59000 Lille, France; Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Sara Costa-Pereira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Julie Darrigues
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Afonso Antunes de Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Luc Buée
- Université Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, 59000 Lille, France; Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Jade Dunot
- Université Côte d'Azur, CNRS, UMR 7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne, France
| | - Hélène Marie
- Université Côte d'Azur, CNRS, UMR 7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne, France
| | - Paula A Pousinha
- Université Côte d'Azur, CNRS, UMR 7275, Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne, France
| | - David Blum
- Université Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, 59000 Lille, France; Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Luísa V Lopes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal.
| | - Julie C Ribot
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal.
| |
Collapse
|
43
|
Influence of a High-Impact Multidimensional Rehabilitation Program on the Gut Microbiota of Patients with Multiple Sclerosis. Int J Mol Sci 2021; 22:ijms22137173. [PMID: 34281224 PMCID: PMC8268819 DOI: 10.3390/ijms22137173] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 01/04/2023] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative inflammatory condition mediated by autoreactive immune processes. Due to its potential to influence host immunity and gut-brain communication, the gut microbiota has been suggested to be involved in the onset and progression of MS. To date, there is no definitive cure for MS, and rehabilitation programs are of the utmost importance, especially in the later stages. However, only a few people generally participate due to poor support, knowledge, and motivation, and no information is available on gut microbiota changes. Herein we evaluated the potential of a brief high-impact multidimensional rehabilitation program (B-HIPE) in a leisure environment to affect the gut microbiota, mitigate MS symptoms and improve quality of life. B-HIPE resulted in modulation of the MS-typical dysbiosis, with reduced levels of pathobionts and the replenishment of beneficial short-chain fatty acid producers. This partial recovery of a eubiotic profile could help counteract the inflammatory tone typically observed in MS, as supported by reduced circulating lipopolysaccharide levels and decreased populations of pro-inflammatory lymphocytes. Improved physical performance and fatigue relief were also found. Our findings pave the way for integrating clinical practice with holistic approaches to mitigate MS symptoms and improve patients’ quality of life.
Collapse
|
44
|
Li Y, Zhang Y, Zeng X. γδ T Cells Participating in Nervous Systems: A Story of Jekyll and Hyde. Front Immunol 2021; 12:656097. [PMID: 33868300 PMCID: PMC8044362 DOI: 10.3389/fimmu.2021.656097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/15/2021] [Indexed: 11/18/2022] Open
Abstract
γδ T cells are distributed in various lymphoid and nonlymphoid tissues, and act as early responders in many conditions. Previous studies have proven their significant roles in infection, cancer, autoimmune diseases and tissue maintenance. Recently, accumulating researches have highlighted the crosstalk between γδ T cells and nervous systems. In these reports, γδ T cells maintain some physiological functions of central nervous system by secreting interleukin (IL) 17, and neurons like nociceptors can in turn regulate the activity of γδ T cells. Moreover, γδ T cells are involved in neuroinflammation such as stroke and multiple sclerosis. This review illustrates the relationship between γδ T cells and nervous systems in physiological and pathological conditions.
Collapse
Affiliation(s)
| | | | - Xun Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
45
|
Sasaki T, Nagata R, Takahashi S, Takei Y. Effects of RORγt overexpression on the murine central nervous system. Neuropsychopharmacol Rep 2021; 41:102-110. [PMID: 33547881 PMCID: PMC8182958 DOI: 10.1002/npr2.12162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 01/07/2021] [Accepted: 01/19/2021] [Indexed: 12/18/2022] Open
Abstract
Objective T helper 17 (Th17) cells are a subset of CD4+ T cells that produce interleukin (IL)‐17A. Recent studies showed that an increase in circulating IL‐17A causes cognitive dysfunction, although it is unknown how increased systemic IL‐17A affects brain function. Using transgenic mice overexpressing RORγt, a transcription factor essential for differentiation of Th17 cells (RORγt Tg mice), we examined changes in the brain caused by chronically increased IL‐17A resulting from excessive activation of Th17 cells. Results RORγt Tg mice exhibited elevated Rorc and IL‐17A mRNA expression in the colon, as well as a chronic increase in circulating IL‐17A. We found that the immunoreactivity of Iba1 and density of microglia were lower in the dentate gyrus of RORγt Tg mice compared with wild‐type mice. However, GFAP+ astrocytes were unchanged in the hippocampi of RORγt Tg mice. Levels of synaptic proteins were not significantly different between RORγt Tg and wild‐type mouse brains. In addition, novel object location test results indicated no difference in preference between these mice. Conclusion Our findings indicate that a continuous increase of IL‐17A in response to RORγt overexpression resulted in decreased microglia activity in the dentate gyrus, but had only a subtle effect on murine hippocampal functions. Using transgenic mice overexpressing RORγt, a transcription factor essential for differentiation of Th17 cells, we examined changes in the brain caused by chronically increased IL‐17A resulting from excessive activation of Th17 cells. RORγt Tg mice exhibited elevated Rorc and IL‐17A mRNA expression in the colon, as well as a chronic increase in circulating IL‐17A. Our findings indicate that a continuous increase of IL‐17A in response to RORγt overexpression resulted in decreased microglia activity in the dentate gyrus but had a subtle effect on murine hippocampal functions.![]()
Collapse
Affiliation(s)
- Tetsuya Sasaki
- Faculty of Medicine, Department of Anatomy and Neuroscience, University of Tsukuba, Tsukuba, Ibaraki, Japan.,PhD Program of Neurosciences, Degree Program of Comprehensive Human Sciences, Graduate School of Comprehensive Human Sciences,, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Rei Nagata
- Faculty of Medicine, Department of Anatomy and Neuroscience, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Satoru Takahashi
- Faculty of Medicine, Department of Anatomy and Embryology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yosuke Takei
- Faculty of Medicine, Department of Anatomy and Neuroscience, University of Tsukuba, Tsukuba, Ibaraki, Japan.,PhD Program of Neurosciences, Degree Program of Comprehensive Human Sciences, Graduate School of Comprehensive Human Sciences,, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
46
|
Wang C, Zhang R, Zhang Q, Jin H, Wei C, Wu C, Mei L, Liu Y, Zhang P. Cytokine Profiles and the Effect of Intravitreal Aflibercept Treatment on Experimental Choroidal Neovascularization. Ophthalmic Res 2020; 65:68-76. [PMID: 33279910 DOI: 10.1159/000513588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/28/2020] [Indexed: 11/19/2022]
Abstract
PURPOSE The purpose of our study was to investigate the profiles of inflammatory cytokines and the macrophage polarization gene in a choroidal neovascularization (CNV) mouse model before and after intravitreal aflibercept treatment. METHODS The CNV mouse model was conducted by laser photocoagulation. A total of 58 cytokines were measured by the multiplex mouse cytokine antibody array. The macrophage polarization genes were tested by reverse transcription polymerase chain reaction. The relationship between the cytokines and the CNV lesion area was analyzed by correlation. RESULTS MIP-1a on day 3 after laser photocoagulation, MCP-5 and Fas-L on day 7, and IL-15 and IL-7 on day 14 were significantly upregulated (p < 0.001, fold change >10.0). After the intravitreal aflibercept treatment, GM-CSF and MCP-1 on day 3 and TIMP-1 on days 7 and 14 were the most significantly upregulated cytokines (p < 0.001, fold change >10.0). MIP-1 on day 3, IL-13 and Fas-L on day 7, and Fas-L on day 14 were the most significantly downregulated cytokines after intravitreal aflibercept treatment (p < 0.001, fold change >5.0). M2 polarization and VEGFA genes were significantly increased in the CNV formation, whereas aflibercept suppressed M2 polarization and VEGFA genes. IL-7 was negatively related to the CNV lesion area on day 14 after intravitreal aflibercept treatment (r = -0.938, p = 0.006). CONCLUSION The inflammatory cytokines and the M1/M2 macrophage genes significantly changed in the CNV mouse model. This result suggests that inflammatory cytokines and macrophages play a critical role in the physiopathology of CNV.
Collapse
Affiliation(s)
- Chen Wang
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Rongrong Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Qi Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Huixiang Jin
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Chenghua Wei
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Changfan Wu
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Lixin Mei
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Yinping Liu
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Pengfei Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China.,Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, China
| |
Collapse
|
47
|
Pavlenko D, Funahashi H, Sakai K, Hashimoto T, Lozada T, Yosipovitch G, Akiyama T. IL-23 modulates histamine-evoked itch and responses of pruriceptors in mice. Exp Dermatol 2020; 29:1209-1215. [PMID: 33010057 DOI: 10.1111/exd.14206] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/14/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2023]
Abstract
Accumulating evidence has highlighted the essential roles of cytokines in itch processing. Although IL-23 and Th17 cytokines are elevated in inflammatory skin disorders, their role in itch is unknown. Here, we investigated the role of IL-23 and IL-17A in itch response using an in vitro calcium imaging of mouse dorsal root ganglion (DRG) neurons and an in vivo behaviour test. Calcium imaging studies revealed that a few DRG neurons (~5%) responded to either IL-23 or IL-17A. Pretreatment cells with IL-23 significantly reduced calcium responses to histamine and capsaicin but not chloroquine. Behaviour experiments showed neither IL-23 nor IL-17A evoked scratching. IL-23 significantly decreased histamine-evoked scratching without affecting chloroquine-evoked scratching. There was no difference in scratching between IL-17A- and vehicle-treated groups. These results indicate that IL-23 might play a role in regulating histaminergic itch via modulation of TRPV1 activity.
Collapse
Affiliation(s)
- Darya Pavlenko
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hideki Funahashi
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Psychiatry, University of Miyazaki, Kiyotake, Japan
| | - Kent Sakai
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Takashi Hashimoto
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Taisa Lozada
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Gil Yosipovitch
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Tasuku Akiyama
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Miami Itch Center, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
48
|
Ronaldson PT, Davis TP. Regulation of blood-brain barrier integrity by microglia in health and disease: A therapeutic opportunity. J Cereb Blood Flow Metab 2020; 40:S6-S24. [PMID: 32928017 PMCID: PMC7687032 DOI: 10.1177/0271678x20951995] [Citation(s) in RCA: 230] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The blood-brain barrier (BBB) is a critical regulator of CNS homeostasis. It possesses physical and biochemical characteristics (i.e. tight junction protein complexes, transporters) that are necessary for the BBB to perform this physiological role. Microvascular endothelial cells require support from astrocytes, pericytes, microglia, neurons, and constituents of the extracellular matrix. This intricate relationship implies the existence of a neurovascular unit (NVU). NVU cellular components can be activated in disease and contribute to dynamic remodeling of the BBB. This is especially true of microglia, the resident immune cells of the brain, which polarize into distinct proinflammatory (M1) or anti-inflammatory (M2) phenotypes. Current data indicate that M1 pro-inflammatory microglia contribute to BBB dysfunction and vascular "leak", while M2 anti-inflammatory microglia play a protective role at the BBB. Understanding biological mechanisms involved in microglia activation provides a unique opportunity to develop novel treatment approaches for neurological diseases. In this review, we highlight characteristics of M1 proinflammatory and M2 anti-inflammatory microglia and describe how these distinct phenotypes modulate BBB physiology. Additionally, we outline the role of other NVU cell types in regulating microglial activation and highlight how microglia can be targeted for treatment of disease with a focus on ischemic stroke and Alzheimer's disease.
Collapse
Affiliation(s)
- Patrick T Ronaldson
- Department of Pharmacology, College of Medicine University of Arizona, Tucson, AZ, USA
| | - Thomas P Davis
- Department of Pharmacology, College of Medicine University of Arizona, Tucson, AZ, USA
| |
Collapse
|
49
|
Sant'Anna MB, Giardini AC, Ribeiro MAC, Lopes FSR, Teixeira NB, Kimura LF, Bufalo MC, Ribeiro OG, Borrego A, Cabrera WHK, Ferreira JCB, Zambelli VO, Sant'Anna OA, Picolo G. The Crotoxin:SBA-15 Complex Down-Regulates the Incidence and Intensity of Experimental Autoimmune Encephalomyelitis Through Peripheral and Central Actions. Front Immunol 2020; 11:591563. [PMID: 33193433 PMCID: PMC7655790 DOI: 10.3389/fimmu.2020.591563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/05/2020] [Indexed: 01/18/2023] Open
Abstract
Crotoxin (CTX), the main neurotoxin from Crotalus durissus terrificus snake venom, has anti-inflammatory, immunomodulatory and antinociceptive activities. However, the CTX-induced toxicity may compromise its use. Under this scenario, the use of nanoparticle such as nanostructured mesoporous silica (SBA-15) as a carrier might become a feasible approach to improve CTX safety. Here, we determined the benefits of SBA-15 on CTX-related neuroinflammatory and immunomodulatory properties during experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis that replicates several histopathological and immunological features observed in humans. We showed that a single administration of CTX:SBA-15 (54 μg/kg) was more effective in reducing pain and ameliorated the clinical score (motor impairment) in EAE animals compared to the CTX-treated EAE group; therefore, improving the disease outcome. Of interest, CTX:SBA-15, but not unconjugated CTX, prevented EAE-induced atrophy and loss of muscle function. Further supporting an immune mechanism, CTX:SBA-15 treatment reduced both recruitment and proliferation of peripheral Th17 cells as well as diminished IL-17 expression and glial cells activation in the spinal cord in EAE animals when compared with CTX-treated EAE group. Finally, CTX:SBA-15, but not unconjugated CTX, prevented the EAE-induced cell infiltration in the CNS. These results provide evidence that SBA-15 maximizes the immunomodulatory and anti-inflammatory effects of CTX in an EAE model; therefore, suggesting that SBA-15 has the potential to improve CTX effectiveness in the treatment of MS.
Collapse
Affiliation(s)
| | - Aline C Giardini
- Laboratory of Pain and Signaling, Butantan Institute, Sao Paulo, Brazil
| | - Marcio A C Ribeiro
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Flavia S R Lopes
- Laboratory of Pain and Signaling, Butantan Institute, Sao Paulo, Brazil
| | | | - Louise F Kimura
- Laboratory of Pain and Signaling, Butantan Institute, Sao Paulo, Brazil
| | - Michelle C Bufalo
- Laboratory of Pain and Signaling, Butantan Institute, Sao Paulo, Brazil
| | | | - Andrea Borrego
- Laboratory of Immunogenetics, Butantan Institute, Sao Paulo, Brazil
| | - Wafa H K Cabrera
- Laboratory of Immunogenetics, Butantan Institute, Sao Paulo, Brazil
| | - Julio C B Ferreira
- Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil.,Department of Chemical and Systems Biology, School of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Vanessa O Zambelli
- Laboratory of Pain and Signaling, Butantan Institute, Sao Paulo, Brazil.,Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | | | - Gisele Picolo
- Laboratory of Pain and Signaling, Butantan Institute, Sao Paulo, Brazil
| |
Collapse
|
50
|
Chen J, Liu X, Zhong Y. Interleukin-17A: The Key Cytokine in Neurodegenerative Diseases. Front Aging Neurosci 2020; 12:566922. [PMID: 33132897 PMCID: PMC7550684 DOI: 10.3389/fnagi.2020.566922] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases are characterized by the loss of neurons and/or myelin sheath, which deteriorate over time and cause dysfunction. Interleukin 17A is the signature cytokine of a subset of CD4+ helper T cells known as Th17 cells, and the IL-17 cytokine family contains six cytokines and five receptors. Recently, several studies have suggested a pivotal role for the interleukin-17A (IL-17A) cytokine family in human inflammatory or autoimmune diseases and neurodegenerative diseases, including psoriasis, rheumatoid arthritis (RA), Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and glaucoma. Studies in recent years have shown that the mechanism of action of IL-17A is more subtle than simply causing inflammation. Although the specific mechanism of IL-17A in neurodegenerative diseases is still controversial, it is generally accepted now that IL-17A causes diseases by activating glial cells. In this review article, we will focus on the function of IL-17A, in particular the proposed roles of IL-17A, in the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Junjue Chen
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohong Liu
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yisheng Zhong
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|