1
|
Dietz S, Hebel J, Rühle J, Huff A, Eltzschig HK, Lajqi T, Poets CF, Gille C, Köstlin‐Gille N. Impact of the adenosine receptor A2BR expressed on myeloid cells on immune regulation during pregnancy. Eur J Immunol 2024; 54:e2451149. [PMID: 39460389 PMCID: PMC11628929 DOI: 10.1002/eji.202451149] [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: 03/18/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024]
Abstract
During pregnancy, the maternal immune system must carefully balance protection against pathogens with tolerance toward the semiallogeneic fetus. Dysfunctions of the immune system can lead to severe complications such as preeclampsia, fetal growth restriction, or pregnancy loss. Adenosine plays a role in physiological processes and plasma-level increase during pregnancy. The adenosine receptor A2B (A2BR), which is expressed on both, immune and nonimmune cells, is activated by high adenosine concentrations, achieved during pregnancy. We investigated the impact of A2BR expressed on myeloid cells on immune regulation during pregnancy using a mouse model with myeloid deficiency for A2BR. We demonstrate systemic changes in myeloid and lymphoid cell populations during pregnancy in A2BR-KO (Adora2B923f/f-LysMCre) mice with increased monocytes, neutrophils, and T cells but decreased B cells as well as altered T-cell subpopulations with decreased Th1 cells and Tregs and increased Th17 cells. Lack of A2BR on myeloid cells caused an increased systemic expression of IL-6 but decreased systemic accumulation and function of MDSC and reduced numbers of uterine natural killer cells. The pregnancy outcome was only marginally affected. Our results demonstrate that A2BR on myeloid cells plays a role in immune regulation during pregnancy, but the clinical impact on pregnancy remains unclear.
Collapse
Affiliation(s)
- Stefanie Dietz
- Department of NeonatologyTuebingen University Children's HospitalTuebingenGermany
- Department of NeonatologyHeidelberg University, Medical FacultyHeidelbergGermany
| | - Janine Hebel
- Department of NeonatologyTuebingen University Children's HospitalTuebingenGermany
| | - Jessica Rühle
- Department of NeonatologyTuebingen University Children's HospitalTuebingenGermany
| | - Alisha Huff
- Department of NeonatologyTuebingen University Children's HospitalTuebingenGermany
| | | | - Trim Lajqi
- Department of NeonatologyHeidelberg University, Medical FacultyHeidelbergGermany
| | - Christian F. Poets
- Department of NeonatologyTuebingen University Children's HospitalTuebingenGermany
| | - Christian Gille
- Department of NeonatologyHeidelberg University, Medical FacultyHeidelbergGermany
| | - Natascha Köstlin‐Gille
- Department of NeonatologyTuebingen University Children's HospitalTuebingenGermany
- Department of NeonatologyHeidelberg University, Medical FacultyHeidelbergGermany
| |
Collapse
|
2
|
Han J, Cherry C, Mejías JC, Krishnan K, Ruta A, Maestas DR, Peña AN, Nguyen HH, Nagaraj S, Yang B, Gray-Gaillard EF, Rutkowski N, Browne M, Tam AJ, Fertig EJ, Housseau F, Ganguly S, Moore EM, Pardoll DM, Elisseeff JH. Age-associated Senescent - T Cell Signaling Promotes Type 3 Immunity that Inhibits the Biomaterial Regenerative Response. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310476. [PMID: 38087458 DOI: 10.1002/adma.202310476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/20/2023] [Indexed: 12/30/2023]
Abstract
Aging is associated with immunological changes that compromise response to infections and vaccines, exacerbate inflammatory diseases and can potentially mitigate tissue repair. Even so, age-related changes to the immune response to tissue damage and regenerative medicine therapies remain unknown. Here, it is characterized how aging induces changes in immunological signatures that inhibit tissue repair and therapeutic response to a clinical regenerative biological scaffold derived from extracellular matrix. Signatures of inflammation and interleukin (IL)-17 signaling increased with injury and treatment both locally and regionally in aged animals, and computational analysis uncovered age-associated senescent-T cell communication that promotes type 3 immunity in T cells. Local inhibition of type 3 immune activation using IL17-neutralizing antibodies improves healing and restores therapeutic response to the regenerative biomaterial, promoting muscle repair in older animals. These results provide insights into tissue immune dysregulation that occurs with aging that can be targeted to rejuvenate repair.
Collapse
Affiliation(s)
- Jin Han
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Christopher Cherry
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Joscelyn C Mejías
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Kavita Krishnan
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Anna Ruta
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - David R Maestas
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Alexis N Peña
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Helen Hieu Nguyen
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Sushma Nagaraj
- Department of Neurology, Brain Science Institute, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Brenda Yang
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Elise F Gray-Gaillard
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Natalie Rutkowski
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Maria Browne
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Ada J Tam
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Elana J Fertig
- Department of Biomedical Engineering and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21218, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Franck Housseau
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Sudipto Ganguly
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Erika M Moore
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Drew M Pardoll
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Jennifer H Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21231, USA
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| |
Collapse
|
3
|
Kido-Nakahara M, Chiba T, Mizusawa Y, Higashi Y, Ibusuki A, Igawa S, Murakami Y, Matsunaka H, Kuba-Fuyuno Y, Tsuji G, Nakahara T. Cytokine profile of the stratum corneum in atopic dermatitis lesions differs between the face and the trunk. Allergol Int 2024:S1323-8930(24)00111-4. [PMID: 39332980 DOI: 10.1016/j.alit.2024.08.010] [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: 03/31/2024] [Revised: 08/13/2024] [Accepted: 08/31/2024] [Indexed: 09/29/2024] Open
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic inflammatory skin disease with intense pruritus. Dupilumab, an anti-IL-4 receptor alpha antibody, has been revealed to be highly effective against the symptoms of AD; however, dupilumab takes longer to improve facial dermatitis in some patients. We thus examined whether the cytokine profiles in AD lesions differ between different anatomical locations. METHODS Stratum corneum was collected by tape stripping from lesions of the forehead and abdomen of 24 patients with moderate to severe AD and at the same anatomical locations of 14 control subjects. These samples were then used to determine the expression profiles of Th1, Th2, and Th17 cytokines/chemokines by multiplex assay and immunocytochemistry. RESULTS We found that cytokines/chemokines in the stratum corneum differed in their expression between different anatomical areas in AD patients and also in healthy control subjects. The expression of Th1 and Th17 cytokines/chemokines such as IP-10, MIG, and IL-17 tended to be higher in the forehead than in the abdomen in the AD group. Regarding Th2 cytokines/chemokines, some (e.g., IL-13 and IL-33) were highly expressed in the abdomen, others (e.g., IL-4 and IL-31) were highly expressed in the forehead, and a third group (e.g., TARC and TSLP) did not differ significantly in their expression between the forehead and abdomen. These patterns of Th2 cytokines were almost identical in the stratum corneum of healthy individuals. CONCLUSIONS Differences in cytokine/chemokine profiles in the stratum corneum between different anatomical areas might affect the responsiveness to AD treatment.
Collapse
Affiliation(s)
- Makiko Kido-Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Takahito Chiba
- Department of Dermatology and Plastic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Yuta Mizusawa
- Department of Dermatology and Plastic Surgery, Akita University Graduate School of Medicine, Akita, Japan
| | - Yuko Higashi
- Department of Dermatology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Atsuko Ibusuki
- Department of Dermatology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Satomi Igawa
- Department of Dermatology, Asahikawa Medical University, Asahikawa, Japan
| | - Yumi Murakami
- NOV Academic Research, Tokiwa Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Hiroshi Matsunaka
- NOV Academic Research, Tokiwa Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Yoko Kuba-Fuyuno
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Gaku Tsuji
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
4
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [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: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
5
|
Li Y, Stewart CA, Finer Y. Advanced Antimicrobial and Anti-Infective Strategies to Manage Peri-Implant Infection: A Narrative Review. Dent J (Basel) 2024; 12:125. [PMID: 38786523 PMCID: PMC11120417 DOI: 10.3390/dj12050125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/21/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
Despite reductions in bacterial infection and enhanced success rate, the widespread use of systemic antibiotic prophylaxis in implant dentistry is controversial. This use has contributed to the growing problem of antimicrobial resistance, along with creating significant health and economic burdens. The basic mechanisms that cause implant infection can be targeted by new prevention and treatment methods which can also lead to the reduction of systemic antibiotic exposure and its associated adverse effects. This review aims to summarize advanced biomaterial strategies applied to implant components based on anti-pathogenic mechanisms and immune balance mechanisms. It emphasizes that modifying the dental implant surface and regulating the early immune response are promising strategies, which may further prevent or slow the development of peri-implant infection, and subsequent failure.
Collapse
Affiliation(s)
- Yihan Li
- Faculty of Dentistry, University of Toronto, 124 Edward St., Toronto, ON M5G 1G6, Canada; (Y.L.); (C.A.S.)
| | - Cameron A. Stewart
- Faculty of Dentistry, University of Toronto, 124 Edward St., Toronto, ON M5G 1G6, Canada; (Y.L.); (C.A.S.)
- Institute of Biomedical Engineering, University of Toronto, 164 College St., Toronto, ON M5S 3E2, Canada
| | - Yoav Finer
- Faculty of Dentistry, University of Toronto, 124 Edward St., Toronto, ON M5G 1G6, Canada; (Y.L.); (C.A.S.)
- Institute of Biomedical Engineering, University of Toronto, 164 College St., Toronto, ON M5S 3E2, Canada
| |
Collapse
|
6
|
Lantz BJ, Moriwaki M, Oyebamiji OM, Guo Y, Gonzalez Bosc L. Chronic hypoxia disrupts T regulatory cell phenotype contributing to the emergence of exTreg-T H17 cells. Front Physiol 2024; 14:1304732. [PMID: 38347920 PMCID: PMC10859758 DOI: 10.3389/fphys.2023.1304732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 12/28/2023] [Indexed: 02/15/2024] Open
Abstract
The imbalance between pro-inflammatory T helper 17 (TH17) cells and anti-inflammatory regulatory T cells (Tregs) has been implicated in multiple inflammatory and autoimmune conditions, but the effects of chronic hypoxia (CH) on this balance have yet to be explored. CH-exposed mice have an increased prevalence of TH17 cells in the lungs with no change in Tregs. This imbalance is significant because it precedes the development of pulmonary hypertension (PH), and TH17 cells are a major contributor to CH-induced PH. While Tregs have been shown to attenuate or prevent the development of certain types of PH through activation and adoptive transfer experiments, why Tregs remain unable to prevent disease progression naturally, specifically in CH-induced PH, remains unclear. Our study aimed to test the hypothesis that increased TH17 cells observed following CH are caused by decreased circulating levels of Tregs and switching of Tregs to exTreg-TH17 cells, following CH. We compared gene expression profiles of Tregs from normoxia or 5-day CH splenocytes harvested from Foxp3tm9(EGFP/cre/ERT2)Ayr/J x Ai14-tdTomato mice, which allowed for Treg lineage tracing through the presence or absence of EGFP and/or tdTomato expression. We found Tregs in CH exposed mice contained gene profiles consistent with decreased suppressive ability. We determined cell prevalence and expression of CD25 and OX40, proteins critical for Treg function, in splenocytes from Foxp3tm9(EGFP/cre/ERT2)Ayr/J x Ai14-tdTomato mice under the same conditions. We found TH17 cells to be increased and Tregs to be decreased, following CH, with protein expression of CD25 and OX40 in Tregs matching the gene expression data. Finally, using the lineage tracing ability of this mouse model, we were able to demonstrate the emergence of exTreg-TH17 cells, following CH. These findings suggest that CH causes a decrease in Treg suppressive capacity, and exTregs respond to CH by transitioning to TH17 cells, both of which tilt the Treg-TH17 cell balance toward TH17 cells, creating a pro-inflammatory environment.
Collapse
Affiliation(s)
- Benjamin J. Lantz
- Gonzalez Bosc Laboratory, Health Sciences Center, Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Mika Moriwaki
- Gonzalez Bosc Laboratory, Health Sciences Center, Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| | - Olufunmilola M. Oyebamiji
- Division of Molecular Medicine, Health Sciences Center, Internal Medicine, University of New Mexico, Albuquerque, NM, United States
| | - Yan Guo
- Department of Public Health and Sciences, University of Miami, Miami, FL, United States
| | - Laura Gonzalez Bosc
- Gonzalez Bosc Laboratory, Health Sciences Center, Cell Biology and Physiology, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
7
|
Najar M, Rahmani S, Faour WH, Alsabri SG, Lombard CA, Fayyad-Kazan H, Sokal EM, Merimi M, Fahmi H. Umbilical Cord Mesenchymal Stromal/Stem Cells and Their Interplay with Th-17 Cell Response Pathway. Cells 2024; 13:169. [PMID: 38247860 PMCID: PMC10814115 DOI: 10.3390/cells13020169] [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: 11/14/2023] [Revised: 12/30/2023] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
As a form of immunomodulatory therapeutics, mesenchymal stromal/stem cells (MSCs) from umbilical cord (UC) tissue were assessed for their dynamic interplay with the Th-17 immune response pathway. UC-MSCs were able to modulate lymphocyte response by promoting a Th-17-like profile. Such modulation depended on the cell ratio of the cocultures as well as the presence of an inflammatory setting underlying their plasticity. UC-MSCs significantly increased the expression of IL-17A and RORγt but differentially modulated T cell expression of IL-23R. In parallel, the secretion profile of the fifteen factors (IL1β, IL-4, IL-6, IL-10, IL-17A, IL-17F, IL-22, IL-21, IL-23, IL-25, IL-31, IL-33, INF-γ, sCD40, and TNF-α) involved in the Th-17 immune response pathway was substantially altered during these cocultures. The modulation of these factors demonstrates the capacity of UC-MSCs to sense and actively respond to tissue challenges. Protein network and functional enrichment analysis indicated that several biological processes, molecular functions, and cellular components linked to distinct Th-17 signaling interactions are involved in several trophic, inflammatory, and immune network responses. These immunological changes and interactions with the Th-17 pathway are likely critical to tissue healing and may help to identify molecular targets that will improve therapeutic strategies involving UC-MSCs.
Collapse
Affiliation(s)
- Mehdi Najar
- Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada
- Faculty of Medicine, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Saida Rahmani
- LBBES Laboratory, Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco
| | - Wissam H. Faour
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos 5053, Lebanon
| | - Sami G. Alsabri
- Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Catherine A. Lombard
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, P.O. Box 6573/14, Beirut 1103, Lebanon
| | - Etienne M. Sokal
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Makram Merimi
- LBBES Laboratory, Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco
| | - Hassan Fahmi
- Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada
| |
Collapse
|
8
|
Peña-Juárez MC, Guadarrama-Escobar OR, Serrano-Castañeda P, Méndez-Albores A, Vázquez-Durán A, Vera-Graziano R, Rodríguez-Pérez B, Salgado-Machuca M, Anguiano-Almazán E, Morales-Florido MI, Rodríguez-Cruz IM, Escobar-Chávez JJ. Synergistic Effect of Retinoic Acid and Lactoferrin in the Maintenance of Gut Homeostasis. Biomolecules 2024; 14:78. [PMID: 38254678 PMCID: PMC10813542 DOI: 10.3390/biom14010078] [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: 11/22/2023] [Revised: 12/26/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Lactoferrin (LF) is a glycoprotein that binds to iron ions (Fe2+) and other metallic ions, such as Mg2+, Zn2+, and Cu2+, and has antibacterial and immunomodulatory properties. The antibacterial properties of LF are due to its ability to sequester iron. The immunomodulatory capability of LF promotes homeostasis in the enteric environment, acting directly on the beneficial microbiota. LF can modulate antigen-presenting cell (APC) biology, including migration and cell activation. Nonetheless, some gut microbiota strains produce toxic metabolites, and APCs are responsible for initiating the process that inhibits the inflammatory response against them. Thus, eliminating harmful strains lowers the risk of inducing chronic inflammation, and consequently, metabolic disease, which can progress to type 2 diabetes mellitus (T2DM). LF and retinoic acid (RA) exhibit immunomodulatory properties such as decreasing cytokine production, thus modifying the inflammatory response. Their activities have been observed both in vitro and in vivo. The combined, simultaneous effect of these molecules has not been studied; however, the synergistic effect of LF and RA may be employed for enhancing the secretion of humoral factors, such as IgA. We speculate that the combination of LF and RA could be a potential prophylactic alternative for the treatment of metabolic dysregulations such as T2DM. The present review focuses on the importance of a healthy diet for a balanced gut and describes how probiotics and prebiotics with immunomodulatory activity as well as inductors of differentiation and cell proliferation could be acquired directly from the diet or indirectly through the oral administration of formulations aimed to maintain gut health or restore a eubiotic state in an intestinal environment that has been dysregulated by external factors such as stress and a high-fat diet.
Collapse
Affiliation(s)
- Ma. Concepción Peña-Juárez
- Unidad de Investigación Multidisciplinaria Lab-12 (Sistemas Transdérmicos y Materiales Nanoestructurados), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (M.C.P.-J.); (O.R.G.-E.); (P.S.-C.); (M.S.-M.); (E.A.-A.); (M.I.M.-F.)
| | - Omar Rodrigo Guadarrama-Escobar
- Unidad de Investigación Multidisciplinaria Lab-12 (Sistemas Transdérmicos y Materiales Nanoestructurados), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (M.C.P.-J.); (O.R.G.-E.); (P.S.-C.); (M.S.-M.); (E.A.-A.); (M.I.M.-F.)
| | - Pablo Serrano-Castañeda
- Unidad de Investigación Multidisciplinaria Lab-12 (Sistemas Transdérmicos y Materiales Nanoestructurados), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (M.C.P.-J.); (O.R.G.-E.); (P.S.-C.); (M.S.-M.); (E.A.-A.); (M.I.M.-F.)
| | - Abraham Méndez-Albores
- Unidad de Investigación Multidisciplinaria Lab-14 (Ciencia y Tecnología de los Materiales), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (A.M.-A.); (A.V.-D.)
| | - Alma Vázquez-Durán
- Unidad de Investigación Multidisciplinaria Lab-14 (Ciencia y Tecnología de los Materiales), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (A.M.-A.); (A.V.-D.)
| | - Ricardo Vera-Graziano
- Instituto de Investigaciones en Materiales, Universidad Nacional Autónoma de México, Apartado Postal 70-360, Ciudad Universitaria, Coyoacán, Ciudad de México 04510, Mexico;
| | - Betsabé Rodríguez-Pérez
- Laboratorio de Servicio de Análisis de Propóleos (LASAP), Unidad de Investigación Multidisciplinaria (UIM), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Cuautitlán Izcalli 54714, Mexico;
| | - Mariana Salgado-Machuca
- Unidad de Investigación Multidisciplinaria Lab-12 (Sistemas Transdérmicos y Materiales Nanoestructurados), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (M.C.P.-J.); (O.R.G.-E.); (P.S.-C.); (M.S.-M.); (E.A.-A.); (M.I.M.-F.)
| | - Ericka Anguiano-Almazán
- Unidad de Investigación Multidisciplinaria Lab-12 (Sistemas Transdérmicos y Materiales Nanoestructurados), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (M.C.P.-J.); (O.R.G.-E.); (P.S.-C.); (M.S.-M.); (E.A.-A.); (M.I.M.-F.)
| | - Miriam Isabel Morales-Florido
- Unidad de Investigación Multidisciplinaria Lab-12 (Sistemas Transdérmicos y Materiales Nanoestructurados), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (M.C.P.-J.); (O.R.G.-E.); (P.S.-C.); (M.S.-M.); (E.A.-A.); (M.I.M.-F.)
- Laboratorio de Farmacia Molecular y Liberación Controlada, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Mexico City 04960, Mexico
| | - Isabel Marlene Rodríguez-Cruz
- Unidad de Enseñanza e Investigación, Hospital Regional e Alta Especialidad de Sumpango, Carretera Zumpango-Jilotzingo #400, Barrio de Santiago, 2ª Sección, Zumpango 55600, Mexico;
| | - José Juan Escobar-Chávez
- Unidad de Investigación Multidisciplinaria Lab-12 (Sistemas Transdérmicos y Materiales Nanoestructurados), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Carretera Cuautitlán Teoloyucan, Km 2.5, San Sebastián Xhala, Cuautitlán Izcalli 54714, Mexico; (M.C.P.-J.); (O.R.G.-E.); (P.S.-C.); (M.S.-M.); (E.A.-A.); (M.I.M.-F.)
| |
Collapse
|
9
|
Gao F, Litchfield B, Wu H. Adipose tissue lymphocytes and obesity. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:5. [PMID: 38455510 PMCID: PMC10919906 DOI: 10.20517/jca.2023.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Obesity is associated with chronic inflammation in adipose tissue (AT), mainly evidenced by infiltration and phenotypic changes of various types of immune cells. Macrophages are the major innate immune cells and represent the predominant immune cell population within AT. Lymphocytes, including T cells and B cells, are adaptive immune cells and constitute another important immune cell population in AT. In obesity, CD8+ effector memory T cells, CD4+ Th1 cells, and B2 cells are increased in AT and promote AT inflammation, while regulatory T cells and Th2 cells, which usually function as immune regulatory or type 2 inflammatory cells, are reduced in AT. Immune cells may regulate the metabolism of adipocytes and other cells through various mechanisms, contributing to the development of metabolic diseases, including insulin resistance and type 2 diabetes. Efforts targeting immune cells and inflammation to prevent and treat obesity-linked metabolic disease have been explored, but have not yielded significant success in clinical studies. This review provides a concise overview of the changes in lymphocyte populations within AT and their potential role in AT inflammation and the regulation of metabolic functions in the context of obesity.
Collapse
Affiliation(s)
- Feng Gao
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Huaizhu Wu
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
10
|
Zogorean R, Wirtz S. The yin and yang of B cells in a constant state of battle: intestinal inflammation and inflammatory bowel disease. Front Immunol 2023; 14:1260266. [PMID: 37849749 PMCID: PMC10577428 DOI: 10.3389/fimmu.2023.1260266] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/18/2023] [Indexed: 10/19/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease of the gastrointestinal tract, defined by a clinical relapse-remitting course. Affecting people worldwide, the origin of IBD is still undefined, arising as a consequence of the interaction between genes, environment, and microbiota. Although the root cause is difficult to identify, data clearly indicate that dysbiosis and pathogenic microbial taxa are connected with the establishment and clinical course of IBD. The composition of the microbiota is shaped by plasma cell IgA secretion and binding, while cytokines such as IL10 or IFN-γ are important fine-tuners of the immune response in the gastrointestinal environment. B cells may also influence the course of inflammation by promoting either an anti-inflammatory or a pro-inflammatory milieu. Here, we discuss IgA-producing B regulatory cells as an anti-inflammatory factor in intestinal inflammation. Moreover, we specify the context of IgA and IgG as players that can potentially participate in mucosal inflammation. Finally, we discuss the role of B cells in mouse infection models where IL10, IgA, or IgG contribute to the outcome of the infection.
Collapse
Affiliation(s)
- Roxana Zogorean
- Medizinische Klinik 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Stefan Wirtz
- Medizinische Klinik 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Medical Immunology Campus Erlangen, FAU Erlangen-Nürnberg, Erlangen, Bavaria, Germany
| |
Collapse
|
11
|
Gautam S, Kumar R, Kumar U, Kumar S, Luthra K, Dada R. Yoga maintains Th17/Treg cell homeostasis and reduces the rate of T cell aging in rheumatoid arthritis: a randomized controlled trial. Sci Rep 2023; 13:14924. [PMID: 37696876 PMCID: PMC10495372 DOI: 10.1038/s41598-023-42231-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 09/07/2023] [Indexed: 09/13/2023] Open
Abstract
The pathogenesis of rheumatoid arthritis (RA) is characterized by a Th17/Treg cell imbalance. A pro-inflammatory cytokine milieu that promotes the continued proliferation of Th17 cells is related to the development of autoinflammation. In RA, T cells have several hallmarks of cellular aging, and they accumulate DNA damage, predisposing to the occurrence of mutations and epigenetic alterations. Since the onset, progression, and treatment response are influenced by a variety of external stressors and environmental factors, this study aimed to evaluate the impact of 8-week yoga practice on disease severity, T cell subsets, markers of T cell ageing and inflammation, epigenetic alterations and gene expression patterns in active RA patients on standard disease-modifying anti-rheumatic drugs (DMARDs). A total of 64 participants with active RA were randomized into 2 groups, yoga group (n = 32) or non-yoga group (n = 32); that were assessed for disease severity, at baseline and after 8 week duration, for Disease Activity Score (DAS28-ESR), T cell subsets [Th17 (CD3+ CD4+ IL17+ RORγt+) cells and Treg (CD3+ CD4+ CD25+ CD127-Foxp3+) cells], markers of T cell aging [aged Th17 cells (CD3+ CD4+ IL17+ RORγt+ CD28-) and aged Treg cells (CD3+ CD4+ CD25+ CD127-Foxp3+ CD28-)], pro-inflammatory markers [IL-6, and IL-17], anti-inflammatory markers [TGF-β, and IL-10], epigenetic alterations [5-methyl cytosine, 5-hydroxymethyl cytosine, and HDAC1] and gene expression patterns [RORγt, FoxP3, IL-17, IL-6, TGF-β, CXCL2, CXCR2, and JUN]. In yoga group, there was a significant improvement in DAS28-ESR scores at the end of 8-weeks of yoga program. The Th17 cells and aged T cell subsets showed a significant decline whereas Treg cell population showed a significant elevation in yoga group. There were significant improvements observed in epigenetic markers as well as inflammatory markers post 8-weeks of yoga practice. The yoga group showed downregulation of RORγt, IL-17, IL-6, CXCL2, CXCR2, and upregulation of FoxP3 and TGF-β transcripts. Yoga enables the maintenance of immune-homeostasis as evident by increased Treg cell population and reduced Th17 cell population. Yoga reduces the rate of immunological aging in T cells, as seen by the reduction in population of aged Th17 cells and aged Treg cells. Yoga positively modifies transcriptome and epigenome by normalization of various inflammatory markers, gene expression patterns and epigenetic alterations. Taken together, yoga reduces RA severity, and aids in immune-modulation and hence can be beneficial as an adjunct therapy.
Collapse
Affiliation(s)
- Surabhi Gautam
- Department of Anatomy, Molecular Reproduction and Genetics Facility, All India Institute of Medical Sciences (AIIMS), New Delhi, India
- Department of Orthopaedics, Emory Musculoskeletal Institute, Emory University School of Medicine, Atlanta, USA
| | - Romsha Kumar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Uma Kumar
- Department of Rheumatology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Sanjeev Kumar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Rima Dada
- Department of Anatomy, Molecular Reproduction and Genetics Facility, All India Institute of Medical Sciences (AIIMS), New Delhi, India.
| |
Collapse
|
12
|
Wu D, Zhang X, Zimmerly KM, Wang R, Livingston A, Iwawaki T, Kumar A, Wu X, Mandell MA, Liu M, Yang XO. Unconventional Activation of IRE1 Enhances TH17 Responses and Promotes Neutrophilic Airway Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.30.547286. [PMID: 37461622 PMCID: PMC10349957 DOI: 10.1101/2023.06.30.547286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Treatment-refractory severe asthma manifests a neutrophilic phenotype associated with TH17 responses. Heightened unfolded protein responses (UPRs) are associated with the risk of asthma, including severe asthma. However, how UPRs participate in the deregulation of TH17 cells leading to this type of asthma remains elusive. In this study, we investigated the role of the UPR sensor IRE1 in TH17 cell function and neutrophilic airway inflammation. We found that IRE1 is induced in fungal asthma and is highly expressed in TH17 cells relative to naïve CD4+ T cells. Cytokine (e.g. IL-23) signals induce the IRE1-XBP1s axis in a JAK2-dependent manner. This noncanonical activation of the IRE1-XBP1s pathway promotes UPRs and cytokine secretion by TH17 cells. Ern1 (encoding IRE1)-deficiency decreases the expression of ER stress factors and impairs the differentiation and cytokine secretion of TH17 cells. Genetic ablation of Ern1 leads to alleviated TH17 responses and airway neutrophilia in a Candida albicans asthma model. Consistently, IL-23 activates the JAK2-IRE1-XBP1s pathway in vivo and enhances TH17 responses and neutrophilic infiltration into the airway. Taken together, our data indicate that IRE1, noncanonically activated by cytokine signals, promotes neutrophilic airway inflammation through the UPRmediated secretory function of TH17 cells. The findings provide a novel insight into the fundamental understanding of IRE1 in TH17-biased TH2-low asthma.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Xing Zhang
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Kourtney M. Zimmerly
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Ruoning Wang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Amanda Livingston
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Takao Iwawaki
- Division of Cell Medicine, Medical Research Institute, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204, USA
| | - Xiang Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
- Department of Parasitology, School of Basic Medical Sciences, Xiangya School of Medicine, Central South University, Changsha, China
| | - Michael A. Mandell
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Xuexian O. Yang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| |
Collapse
|
13
|
Li Y, Guo D, Wang Q, Li A, Yin S, Li S, Li Y, Wang B, Guo T, Feng S. Benzoylaconitine Alleviates Progression of Psoriasis via Suppressing STAT3 Phosphorylation in Keratinocytes. Molecules 2023; 28:molecules28114473. [PMID: 37298949 DOI: 10.3390/molecules28114473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/27/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Psoriasis is a chronic and multifactorial skin disease which is caused by inflammatory infiltrates, keratinocyte hyperproliferation, and accumulation of immune cells. As part of the Aconitum species, Benzoylaconitine (BAC) shows potential antiviral, anti-tumor, and anti-inflammatory effects. In this study, we investigated the effects and mechanisms of BAC on tumor necrosis factor-alpha (TNF-α)/LPS-induced HaCaT keratinocytes in a imiquimod(IMQ)-induced mice model. The results showed that BAC could relieve the symptoms of psoriasis by inhibiting cell proliferation, the release of inflammatory factors, and the accumulation of Th17 cells, while no obvious effect on cell viability and safety was observed both in vitro and in vivo. Additionally, BAC can markedly inhibit the protein and mRNA levels of inflammatory cytokines in TNF-α/LPS-induced HaCaT keratinocytes by inhibiting the phosphorylation of STAT3. In brief, our data indicated that BAC could alleviate the progression of psoriasis and may be a potential therapeutic agent for treating psoriasis in clinical practice.
Collapse
Affiliation(s)
- Yuanbo Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Department of Neurobiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Dandan Guo
- Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Qianqian Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Aifang Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Sugai Yin
- Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Shuxuan Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yalan Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Baiyan Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Tao Guo
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Shuying Feng
- Medical College, Henan University of Chinese Medicine, Zhengzhou 450046, China
| |
Collapse
|
14
|
Dar HY, Perrien DS, Pal S, Stoica A, Uppuganti S, Nyman JS, Jones RM, Weitzmann MN, Pacifici R. Callus γδ T cells and microbe-induced intestinal Th17 cells improve fracture healing in mice. J Clin Invest 2023; 133:e166577. [PMID: 36881482 PMCID: PMC10104897 DOI: 10.1172/jci166577] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
IL-17A (IL-17), a driver of the inflammatory phase of fracture repair, is produced locally by several cell lineages including γδ T cells and Th17 cells. However, the origin of these T cells and their relevance for fracture repair are unknown. Here, we show that fractures rapidly expanded callus γδ T cells, which led to increased gut permeability by promoting systemic inflammation. When the microbiota contained the Th17 cell-inducing taxon segmented filamentous bacteria (SFB), activation of γδ T cells was followed by expansion of intestinal Th17 cells, their migration to the callus, and improved fracture repair. Mechanistically, fractures increased the S1P receptor 1-mediated (S1PR1-mediated) egress of Th17 cells from the intestine and enhanced their homing to the callus through a CCL20-mediated mechanism. Fracture repair was impaired by deletion of γδ T cells, depletion of the microbiome by antibiotics (Abx), blockade of Th17 cell egress from the gut, or Ab neutralization of Th17 cell influx into the callus. These findings demonstrate the relevance of the microbiome and T cell trafficking for fracture repair. Modifications of microbiome composition via Th17 cell-inducing bacteriotherapy and avoidance of broad-spectrum Abx may represent novel therapeutic strategies to optimize fracture healing.
Collapse
Affiliation(s)
- Hamid Y. Dar
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
| | - Daniel S. Perrien
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
| | - Subhashis Pal
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
| | - Andreea Stoica
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
| | - Sasidhar Uppuganti
- Department of Orthopedic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jeffry S. Nyman
- Department of Orthopedic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Rheinallt M. Jones
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University, Atlanta, Georgia, USA
| | - M. Neale Weitzmann
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
- Atlanta VA Health Care System, Department of Veterans Affairs, Decatur, Georgia, USA
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
- Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
15
|
Notch Signaling Pathway Promotes Th17 Cell Differentiation and Participates in Thyroid Autoimmune Injury in Experimental Autoimmune Thyroiditis Mice. Mediators Inflamm 2023; 2023:1195149. [PMID: 36643586 PMCID: PMC9839414 DOI: 10.1155/2023/1195149] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 08/17/2022] [Accepted: 12/23/2022] [Indexed: 01/09/2023] Open
Abstract
Purpose To investigate whether the Notch signaling pathway participates in the occurrence and development of experimental autoimmune thyroiditis (EAT) by affecting the differentiation and function of Th17 cells. Materials and Methods Experimental mice were randomly divided into a control group, an EAT-A group (porcine thyroid immunoglobulin- (pTg-) treated mice) and an EAT-B group (treated with the DAPT γ-secretase inhibitor before pTg). HE staining, IHC staining, flow cytometry, RT-qPCR, and ELISA were used to evaluate the degrees of thyroiditis, detect the percentage of Th17 cells and measure the expression of retinoic acid-related orphan receptor gamma t (RORγt), interleukin-17A (IL-17A), and the main components of the Notch signaling pathway. Results The degrees of thyroiditis, the proportions of Th17 cells, and the expression of RORγt and IL-17A were significantly decreased in the EAT-B group after blocking the Notch signaling pathway by DAPT, and these parameters were significantly increased in the EAT-A group compared to the control group (all P < 0.05). Additionally, the Th17 cell percentages and IL-17A concentrations in spleen mononuclear cells (SMCs) from EAT-A mice decreased in a dose-dependent manner after DAPT treatment in vitro (all P < 0.01). Correlation analyses revealed that the Th17 cell percentages were positively correlated with the serum TgAb titers, Notch pathway-related mRNA expression levels, and IL-17A concentrations in EAT mice (all P < 0.05). Conclusions The expression of Notch signaling pathway components was upregulated in EAT mice, but blockade of the Notch signaling pathway alleviated the degree of thyroiditis, decreased the Th17 cell proportions, and downregulated the IL-17A effector cytokine both in vivo and in vitro. These findings suggested that the Notch signaling pathway may be involved in the pathogenesis of thyroid autoimmune injury in EAT mice by promoting the differentiation of Th17 cells.
Collapse
|
16
|
Puranik N, Yadav D, Song M. Insight into Early Diagnosis of Multiple Sclerosis by Targeting Prognostic Biomarkers. Curr Pharm Des 2023; 29:2534-2544. [PMID: 37921136 DOI: 10.2174/0113816128247471231018053737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 08/04/2023] [Accepted: 09/06/2023] [Indexed: 11/04/2023]
Abstract
Multiple sclerosis (MS) is a central nervous system (CNS) immune-mediated disease that mainly strikes young adults and leaves them disabled. MS is an autoimmune illness that causes the immune system to attack the brain and spinal cord. The myelin sheaths, which insulate the nerve fibers, are harmed by our own immune cells, and this interferes with brain signal transmission. Numbness, tingling, mood swings, memory problems, exhaustion, agony, vision problems, and/or paralysis are just a few of the symptoms. Despite technological advancements and significant research efforts in recent years, diagnosing MS can still be difficult. Each patient's MS is distinct due to a heterogeneous and complex pathophysiology with diverse types of disease courses. There is a pressing need to identify markers that will allow for more rapid and accurate diagnosis and prognosis assessments to choose the best course of treatment for each MS patient. The cerebrospinal fluid (CSF) is an excellent source of particular indicators associated with MS pathology. CSF contains molecules that represent pathological processes such as inflammation, cellular damage, and loss of blood-brain barrier integrity. Oligoclonal bands, neurofilaments, MS-specific miRNA, lncRNA, IgG-index, and anti-aquaporin 4 antibodies are all clinically utilised indicators for CSF in MS diagnosis. In recent years, a slew of new possible biomarkers have been presented. In this review, we look at what we know about CSF molecular markers and how they can aid in the diagnosis and differentiation of different MS forms and treatment options, and monitoring and predicting disease progression, therapy response, and consequences during such opportunistic infections.
Collapse
Affiliation(s)
- Nidhi Puranik
- Biological Sciences Department, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Dhananjay Yadav
- Department of Life Science, Yeungnam University, Gyeongsan 38541, Korea
| | - Minseok Song
- Department of Life Science, Yeungnam University, Gyeongsan 38541, Korea
| |
Collapse
|
17
|
Iraji D, Oftedal BE, Wolff ASB. Th17 Cells: Orchestrators of Mucosal Inflammation and Potential Therapeutic Targets. Crit Rev Immunol 2023; 43:25-52. [PMID: 37831521 DOI: 10.1615/critrevimmunol.2023050360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
T helper 17 (Th17) cells represent a specialized subgroup of effector CD4+ T cells known for their role in provoking neutrophil-driven tissue inflammation, particularly within mucosal tissues. Although they are pivotal for defending the host against extracellular bacteria and fungi, they have also been associated with development of various T cell-mediated inflammatory conditions, autoimmune diseases, and even cancer. Notably, Th17 cells exhibit a dual nature, with different Th17 cell subtypes showcasing distinct effector functions and varying capacities to incite autoimmune tissue inflammation. Furthermore, Th17 cells exhibit significant plasticity, which carries important functional implications, both in terms of their expression of cytokines typically associated with other effector T cell subsets and in their interactions with regulatory CD4+ T cells. The intricate balance of Th17 cytokines can also be a double-edged sword in inflammation, autoimmunity, and cancer. Within this article, we delve into the mechanisms that govern the differentiation, function, and adaptability of Th17 cells. We culminate with an exploration of therapeutic potentials in harnessing the power of Th17 cells and their cytokines. Targeted interventions to modulate Th17 responses are emerging as promising strategies for autoimmunity, inflammation, and cancer treatment. By precisely fine-tuning Th17-related pathways, we may unlock new avenues for personalized therapeutic approaches, aiming to restore immune balance, alleviate the challenges of these disorders, and ultimately enhance the quality of life for individuals affected by them.
Collapse
Affiliation(s)
- Dorsa Iraji
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bergithe E Oftedal
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette S B Wolff
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
18
|
Kim Y, Kim GT. Positive Effects of Biologics on Osteoporosis in Rheumatoid Arthritis. JOURNAL OF RHEUMATIC DISEASES 2023; 30:3-17. [PMID: 37476528 PMCID: PMC10351356 DOI: 10.4078/jrd.22.0046] [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: 10/11/2022] [Revised: 12/10/2022] [Accepted: 12/10/2022] [Indexed: 07/22/2023]
Abstract
Osteoporosis is a systemic skeletal disorder that causes vulnerability of bones to fracture owing to reduction in bone density and deterioration of the bone tissue microstructure. The prevalence of osteoporosis is higher in patients with autoimmune inflammatory rheumatic diseases, including rheumatoid arthritis (RA), than in those of the general population. In this autoimmune inflammatory rheumatic disease, in addition to known risk factors for osteoporosis, various factors such as chronic inflammation, autoantibodies, metabolic disorders, drugs, and decreased physical activity contribute to additional risk. In RA, disease-related inflammation plays an important role in local or systemic bone loss, and active treatment for inflammation can help prevent osteoporosis. In addition to conventional synthetic disease-modifying anti-rheumatic drugs that have been traditionally used for treatment of RA, biologic DMARDs and targeted synthetic DMARDs have been widely used. These agents can be employed more selectively and precisely based on disease pathogenesis. It has been reported that these drugs can inhibit bone loss by not only reducing inflammation in RA, but also by inhibiting bone resorption and promoting bone formation. In this review, the pathogenesis and research results of the increase in osteoporosis in RA are reviewed, and the effects of biological agents on osteoporosis are discussed.
Collapse
Affiliation(s)
- Yunkyung Kim
- Division of Rheumatology, Department of Internal Medicine, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Korea
| | - Geun-Tae Kim
- Division of Rheumatology, Department of Internal Medicine, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Korea
| |
Collapse
|
19
|
Effect of Probiotic E. coli Nissle 1917 Supplementation on the Growth Performance, Immune Responses, Intestinal Morphology, and Gut Microbes of Campylobacter jejuni Infected Chickens. Infect Immun 2022; 90:e0033722. [PMID: 36135600 PMCID: PMC9584303 DOI: 10.1128/iai.00337-22] [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] [Indexed: 11/24/2022] Open
Abstract
Campylobacter jejuni is the most common cause of bacterial foodborne gastroenteritis and holds significant public health importance. The continuing increase of antibiotic-resistant Campylobacter necessitates the development of antibiotic-alternative approaches to control infections in poultry and in humans. Here, we assessed the ability of E. coli Nissle 1917 (EcN; free and chitosan-alginate microencapsulated) to reduce C. jejuni colonization in chickens and measured the effect of EcN on the immune responses, intestinal morphology, and gut microbes of chickens. Our results showed that the supplementation of 3-week-old chickens daily with free EcN in drinking water resulted in a 2.0 log reduction of C. jejuni colonization in the cecum, whereas supplementing EcN orally three times a week, either free or microencapsulated, resulted in 2.0 and 2.5 log reductions of C. jejuni colonization, respectively. Gavaged free and microencapsulated EcN did not have an impact on the evenness or the richness of the cecal microbiota, but it did increase the villous height (VH), crypt depth (CD), and VH:CD ratio in the jejunum and ileum of chickens. Further, the supplementation of EcN (all types) increased C. jejuni-specific and total IgA and IgY antibodies in chicken’s serum. Microencapsulated EcN induced the expression of several cytokines and chemokines (1.6 to 4.3-fold), which activate the Th1, Th2, and Th17 pathways. Overall, microencapsulated EcN displayed promising effects as a potential nonantibiotic strategy to control C. jejuni colonization in chickens. Future studies on testing microencapsulated EcN in the feed and water of chickens raised on built-up floor litter would facilitate the development of EcN for industrial applications to control Campylobacter infections in poultry.
Collapse
|
20
|
Li YJ, Chen CY, Yang JH, Chiu YF. Modulating cholesterol-rich lipid rafts to disrupt influenza A virus infection. Front Immunol 2022; 13:982264. [PMID: 36177026 PMCID: PMC9513517 DOI: 10.3389/fimmu.2022.982264] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Influenza A virus (IAV) is widely disseminated across different species and can cause recurrent epidemics and severe pandemics in humans. During infection, IAV attaches to receptors that are predominantly located in cell membrane regions known as lipid rafts, which are highly enriched in cholesterol and sphingolipids. Following IAV entry into the host cell, uncoating, transcription, and replication of the viral genome occur, after which newly synthesized viral proteins and genomes are delivered to lipid rafts for assembly prior to viral budding from the cell. Moreover, during budding, IAV acquires an envelope with embedded cholesterol from the host cell membrane, and it is known that decreased cholesterol levels on IAV virions reduce infectivity. Statins are commonly used to inhibit cholesterol synthesis for preventing cardiovascular diseases, and several studies have investigated whether such inhibition can block IAV infection and propagation, as well as modulate the host immune response to IAV. Taken together, current research suggests that there may be a role for statins in countering IAV infections and modulating the host immune response to prevent or mitigate cytokine storms, and further investigation into this is warranted.
Collapse
Affiliation(s)
- Yu-Jyun Li
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Yuan Chen
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan
| | - Jeng-How Yang
- Division of Infectious Diseases, Department of Medicine, Chang Gung Memorial Hospital, New Taipei, Taiwan
| | - Ya-Fang Chiu
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan, Taiwan
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
21
|
Cunha C, Koike T, Seki Y, Yamamoto M, Iwashima M. Schnurri 3 promotes Th2 cytokine production during the late phase of T-cell antigen stimulation. Eur J Immunol 2022; 52:1077-1094. [PMID: 35490426 PMCID: PMC9276650 DOI: 10.1002/eji.202149633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 03/22/2022] [Accepted: 04/28/2022] [Indexed: 11/29/2022]
Abstract
Th1 and Th2 polarization is determined by the coordination of numerous factors including the affinity and strength of the antigen-receptor interaction, predominant cytokine environment, and costimulatory molecules present. Here, we show that Schnurri (SHN) proteins have distinct roles in Th1 and Th2 polarization. SHN2 was previously found to block the induction of GATA3 and Th2 differentiation. We found that, in contrast to SHN2, SHN3 is critical for IL-4 production and Th2 polarization. Strength of stimulation controls SHN2 and SHN3 expression patterns, where higher doses of antigen receptor stimulation promoted SHN3 expression and IL-4 production, along with repression of SHN2 expression. SHN3-deficient T cells showed a substantial defect in IL-4 production and expression of AP-1 components, particularly c-Jun and Jun B. This loss of early IL-4 production led to reduced GATA3 expression and impaired Th2 differentiation. Together, these findings uncover SHN3 as a novel, critical regulator of Th2 development.
Collapse
Affiliation(s)
- Christina Cunha
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
| | - Toru Koike
- Department of Biology, Faculty of ScienceShizuoka UniversityShizuokaJapan
| | - Yoichi Seki
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
- Van Kampen Cardiovascular Research Laboratory, Department of Thoracic and Cardiovascular Surgery, Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| | - Mutsumi Yamamoto
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
- Van Kampen Cardiovascular Research Laboratory, Department of Thoracic and Cardiovascular Surgery, Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| | - Makio Iwashima
- Department of Microbiology and ImmunologyLoyola UniversityChicagoIllinoisUSA
- Van Kampen Cardiovascular Research Laboratory, Department of Thoracic and Cardiovascular Surgery, Stritch School of MedicineLoyola UniversityChicagoIllinoisUSA
| |
Collapse
|
22
|
Higgins BW, Shuparski AG, Miller KB, Robinson AM, McHeyzer-Williams LJ, McHeyzer-Williams MG. Isotype-specific plasma cells express divergent transcriptional programs. Proc Natl Acad Sci U S A 2022; 119:e2121260119. [PMID: 35704755 PMCID: PMC9231473 DOI: 10.1073/pnas.2121260119] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 04/29/2022] [Indexed: 11/18/2022] Open
Abstract
Antibodies are produced across multiple isotypes with distinct properties that coordinate initial antigen clearance and confer long-term antigen-specific immune protection. Here, we interrogate the molecular programs of isotype-specific murine plasma cells (PC) following helper T cell-dependent immunization and within established steady-state immunity. We developed a single-cell-indexed and targeted molecular strategy to dissect conserved and divergent components of the rapid effector phase of antigen-specific IgM+ versus inflammation-modulating programs dictated by type 1 IgG2a/b+ PC differentiation. During antibody affinity maturation, the germinal center (GC) cycle imparts separable programs for post-GC type 2 inhibitory IgG1+ and type 1 inflammatory IgG2a/b+ PC to direct long-term cellular function. In the steady state, two subsets of IgM+ and separate IgG2b+ PC programs clearly segregate from splenic type 3 IgA+ PC programs that emphasize mucosal barrier protection. These diverse isotype-specific molecular pathways of PC differentiation control complementary modules of antigen clearance and immune protection that could be selectively targeted for immunotherapeutic applications and vaccine design.
Collapse
Affiliation(s)
- Brett W. Higgins
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | - Andrew G. Shuparski
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | - Karen B. Miller
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | - Amanda M. Robinson
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037
| | | | | |
Collapse
|
23
|
Gupta S, Gautam S, Kumar U, Arora T, Dada R. Potential Role of Yoga Intervention in the Management of Chronic Non-malignant Pain. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:5448671. [PMID: 35668780 PMCID: PMC9167073 DOI: 10.1155/2022/5448671] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/18/2022] [Accepted: 05/13/2022] [Indexed: 12/22/2022]
Abstract
Pain is an unpleasant and upsetting experience. Persistent pain has an impact on an individual's quality of life which causes stress and mood disorders. There are currently no pain-relieving techniques available that can eliminate pain and offer relief without causing any adverse effects. These factors draw attention to traditional treatments like yoga and meditation, which can reduce biological stress and hence increase immunity, as well as alleviate the psychological and emotional suffering produced by pain. Yoga reduces the stress response and the pain cascade via the downregulation of the hypothalamus-pituitary-adrenal (HPA) axis and vagal stimulation. Yoga is a cost-effective growing health practice that, unlike pharmaceuticals, has no side effects and can help patients stay in remission for longer periods of time with fewer relapses. Yoga not only reduces stress and depression severity but also improves functional status and reduces pain perception. This article highlights the impact of yoga on pain management and on a malfunctioning immune system, which leads to improved health, pain reduction, disease management, and improvement in overall quality of life.
Collapse
Affiliation(s)
- Shivani Gupta
- Laboratory for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Surabhi Gautam
- Laboratory for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Uma Kumar
- Department of Rheumatology, All India Institute of Medical Sciences, New Delhi, India
| | - Taruna Arora
- Division of Reproductive Biology, Maternal & Child Health, Indian Council of Medical Research, New Delhi, India
| | - Rima Dada
- Laboratory for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
24
|
Kim ST, Chu Y, Misoi M, Suarez-Almazor ME, Tayar JH, Lu H, Buni M, Kramer J, Rodriguez E, Hussain Z, Neelapu SS, Wang J, Shah AY, Tannir NM, Campbell MT, Gibbons DL, Cascone T, Lu C, Blumenschein GR, Altan M, Lim B, Valero V, Loghin ME, Tu J, Westin SN, Naing A, Garcia-Manero G, Abdel-Wahab N, Tawbi HA, Hwu P, Oliva ICG, Davies MA, Patel SP, Zou J, Futreal A, Diab A, Wang L, Nurieva R. Distinct molecular and immune hallmarks of inflammatory arthritis induced by immune checkpoint inhibitors for cancer therapy. Nat Commun 2022; 13:1970. [PMID: 35413951 PMCID: PMC9005525 DOI: 10.1038/s41467-022-29539-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 03/17/2022] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors are associated with immune-related adverse events (irAEs), including arthritis (arthritis-irAE). Management of arthritis-irAE is challenging because immunomodulatory therapy for arthritis should not impede antitumor immunity. Understanding of the mechanisms of arthritis-irAE is critical to overcome this challenge, but the pathophysiology remains unknown. Here, we comprehensively analyze peripheral blood and/or synovial fluid samples from 20 patients with arthritis-irAE, and unmask a prominent Th1-CD8+ T cell axis in both blood and inflamed joints. CX3CR1hi CD8+ T cells in blood and CXCR3hi CD8+ T cells in synovial fluid, the most clonally expanded T cells, significantly share TCR repertoires. The migration of blood CX3CR1hi CD8+ T cells into joints is possibly mediated by CXCL9/10/11/16 expressed by myeloid cells. Furthermore, arthritis after combined CTLA-4 and PD-1 inhibitor therapy preferentially has enhanced Th17 and transient Th1/Th17 cell signatures. Our data provide insights into the mechanisms, predictive biomarkers, and therapeutic targets for arthritis-irAE.
Collapse
Affiliation(s)
- Sang T Kim
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yanshuo Chu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mercy Misoi
- Department of General Internal Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Maria E Suarez-Almazor
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jean H Tayar
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Huifang Lu
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Maryam Buni
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jordan Kramer
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Biology, Georgetown University, Washington, DC, 20057, USA
| | - Emma Rodriguez
- Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Zulekha Hussain
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jennifer Wang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Amishi Y Shah
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Matthew T Campbell
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Don L Gibbons
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Tina Cascone
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Charles Lu
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - George R Blumenschein
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mehmet Altan
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Vincente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Monica E Loghin
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Janet Tu
- Department of General Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Noha Abdel-Wahab
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Rheumatology and Rehabilitation, Assiut University Hospitals, Faculty of Medicine, Assiut University, El Fateh, Egypt
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Isabella C Glitza Oliva
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Sapna P Patel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jun Zou
- Department of Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Adi Diab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX, 77030, USA.
| | - Roza Nurieva
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX, 77030, USA.
| |
Collapse
|
25
|
Karimi E, Azari H, Tahmasebi A, Nikpoor AR, Negahi AA, Sanadgol N, Shekari M, Mousavi P. LncRNA-miRNA network analysis across the Th17 cell line reveals biomarker potency of lncRNA NEAT1 and KCNQ1OT1 in multiple sclerosis. J Cell Mol Med 2022; 26:2351-2362. [PMID: 35266286 PMCID: PMC8995444 DOI: 10.1111/jcmm.17256] [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] [Received: 10/24/2021] [Revised: 02/08/2022] [Accepted: 02/12/2022] [Indexed: 12/11/2022] Open
Abstract
Differentiation of CD4+ T cells into Th17 cells is an important factor in the onset and progression of multiple sclerosis (MS) and Th17/Treg imbalance. Little is known about the role of lncRNAs in the differentiation of CD4+ cells from Th17 cells. This study aimed to analyse the lncRNA‐miRNAs network involved in MS disease and its role in the differentiation of Th17 cells. The lncRNAs in Th17 differentiation were obtained from GSE66261 using the GEO datasets. Differential expression of lncRNAs in Th17 primary cells compared to Th17 effector cells was investigated by RNA‐seq analysis. Next, the most highlighted lncRNAs in autoimmune diseases were downloaded from the lncRNAs disease database, and the most critical miRNA was extracted by literature search. Then, the lncRNA‐miRNA interaction was achieved by the Starbase database, and the ceRNA network was designed by Cytoscape. Finally, using the CytoHubba application, two hub lncRNAs with the most interactions with miRNAs were identified by the MCODE plug‐in. The expression level of genes was measured by qPCR, and the plasma level of cytokines was analysed by ELISA kits. The results showed an increase in the expression of NEAT1, KCNQ1OT1 and RORC and a decrease in the expression of FOXP3. In plasma, an upregulation of IL17 and a downregulation of TGFB inflammatory cytokines were detected. The dysregulated expression of these genes could be attributed to relapsing‐remitting MS (RR‐MS) patients and help us understand MS pathogenesis better.
Collapse
Affiliation(s)
- Elham Karimi
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Hanieh Azari
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | - Amin Reza Nikpoor
- Sciences Research Center for Molecular Medicine, Hormozgan University of Medical, Hormozgan, Iran
| | - Ahmad Agha Negahi
- Department of Internal Medicine, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Nima Sanadgol
- Institute of Neuroanatomy, RWTH University Hospital Aachen, Aachen, Germany
| | - Mohammad Shekari
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.,Sciences Research Center for Molecular Medicine, Hormozgan University of Medical, Hormozgan, Iran
| | - Pegah Mousavi
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.,Sciences Research Center for Molecular Medicine, Hormozgan University of Medical, Hormozgan, Iran
| |
Collapse
|
26
|
Wang G, Su Z, Li H, Xiao L, Li C, Lian G. The role of metabolism in Th17 cell differentiation and autoimmune diseases. Int Immunopharmacol 2021; 103:108450. [PMID: 34954561 DOI: 10.1016/j.intimp.2021.108450] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/26/2021] [Accepted: 12/05/2021] [Indexed: 12/24/2022]
Abstract
T helper 17 cells (Th17) have been associated with the pathogenesis of autoimmune and inflammatory diseases, which makes them become a sharp focus when the researchers are seeking therapeutic target for these diseases. A growing body of evidence has suggested that cellular metabolism dictates Th17 cell differentiation and effector function. Moreover, various studies have disclosed that metabolism is linked to the occurrence of autoimmune diseases. In this article, we reviewed the most recent findings regarding the importance of metabolism in Th17 cell differentiation and autoimmune diseases and also discussed the modulation mechanisms of glycolysis, fatty acid and cholesterol synthesis, and amino acids metabolism for Th17 cell differentiation. This review summarized the potential therapeutic or preventing strategies for Th17 cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Guang Wang
- Department of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Zehong Su
- Department of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China; Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province Department of Education.
| | - Hui Li
- Department of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China.
| | - Li Xiao
- Department of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China.
| | - Chengyue Li
- Department of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China.
| | - Gaojian Lian
- Department of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
27
|
Logotheti M, Agioutantis P, Katsaounou P, Loutrari H. Microbiome Research and Multi-Omics Integration for Personalized Medicine in Asthma. J Pers Med 2021; 11:jpm11121299. [PMID: 34945771 PMCID: PMC8707330 DOI: 10.3390/jpm11121299] [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: 10/15/2021] [Revised: 11/13/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Asthma is a multifactorial inflammatory disorder of the respiratory system characterized by high diversity in clinical manifestations, underlying pathological mechanisms and response to treatment. It is generally established that human microbiota plays an essential role in shaping a healthy immune response, while its perturbation can cause chronic inflammation related to a wide range of diseases, including asthma. Systems biology approaches encompassing microbiome analysis can offer valuable platforms towards a global understanding of asthma complexity and improving patients' classification, status monitoring and therapeutic choices. In the present review, we summarize recent studies exploring the contribution of microbiota dysbiosis to asthma pathogenesis and heterogeneity in the context of asthma phenotypes-endotypes and administered medication. We subsequently focus on emerging efforts to gain deeper insights into microbiota-host interactions driving asthma complexity by integrating microbiome and host multi-omics data. One of the most prominent achievements of these research efforts is the association of refractory neutrophilic asthma with certain microbial signatures, including predominant pathogenic bacterial taxa (such as Proteobacteria phyla, Gammaproteobacteria class, especially species from Haemophilus and Moraxella genera). Overall, despite existing challenges, large-scale multi-omics endeavors may provide promising biomarkers and therapeutic targets for future development of novel microbe-based personalized strategies for diagnosis, prevention and/or treatment of uncontrollable asthma.
Collapse
Affiliation(s)
- Marianthi Logotheti
- G.P. Livanos and M. Simou Laboratories, 1st Department of Critical Care Medicine & Pulmonary Services, Evangelismos Hospital, Medical School, National Kapodistrian University of Athens, 3 Ploutarchou Str., 10675 Athens, Greece; (M.L.); (P.A.)
- Biotechnology Laboratory, School of Chemical Engineering, National Technical University of Athens, 5 Iroon Polytechniou Str., Zografou Campus, 15780 Athens, Greece
| | - Panagiotis Agioutantis
- G.P. Livanos and M. Simou Laboratories, 1st Department of Critical Care Medicine & Pulmonary Services, Evangelismos Hospital, Medical School, National Kapodistrian University of Athens, 3 Ploutarchou Str., 10675 Athens, Greece; (M.L.); (P.A.)
| | - Paraskevi Katsaounou
- Pulmonary Dept First ICU, Evangelismos Hospital, Medical School, National Kapodistrian University of Athens, Ipsilantou 45-7, 10675 Athens, Greece;
| | - Heleni Loutrari
- G.P. Livanos and M. Simou Laboratories, 1st Department of Critical Care Medicine & Pulmonary Services, Evangelismos Hospital, Medical School, National Kapodistrian University of Athens, 3 Ploutarchou Str., 10675 Athens, Greece; (M.L.); (P.A.)
- Correspondence:
| |
Collapse
|
28
|
Salazar-Viedma M, Vergaño-Salazar JG, Pastenes L, D’Afonseca V. Simulation Model for Hashimoto Autoimmune Thyroiditis Disease. Endocrinology 2021; 162:bqab190. [PMID: 34496027 PMCID: PMC8477452 DOI: 10.1210/endocr/bqab190] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Indexed: 02/07/2023]
Abstract
Hashimoto thyroiditis (HT) is a pathology that often causes a gradual thyroid insufficiency in affected patients due to the autoimmune destruction of this gland. The cellular immune response mediated by T helper lymphocytes TH1 and TH17 can induce the HT disease. In this pathologic condition, there is an imbalance between the TH17 and Treg lymphocytes as well as a gut microbiota dysfunction. The objective of this work was to describe the interactions of the cell subpopulations that participate in HT. To achieve this goal, we generated a mathematical model that allowed the simulation of different scenarios for the dynamic interaction between thyroid cells, the immune system, and the gut microbiota. We used a hypothetical-deductive design of mathematical modeling based on a system of ordinary differential equations, where the state variables are the TH1, TH17, and Treg lymphocytes, the thyrocytes, and the bacteria from gut microbiota. This work generated a compartmental model of the cellular immune response occurring in the thyroid gland. It was observed that TH1 and TH17 lymphocytes could increase the immune cells' activity, as well as activate effector cells directly and trigger the apoptosis and inflammation processes of healthy thyrocytes indirectly. Likewise, the model showed that a reduction in Treg lymphocytes could increase the activity of TH17 lymphocytes when an imbalance of the gut microbiota composition occurred. The numerical results highlight the TH1, TH17, and bacterial balance of the gut microbiota activities as important factors for the development of HT disease.
Collapse
Affiliation(s)
- Marcela Salazar-Viedma
- Laboratorio de Genética y Microevolución, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca 3466706, Chile
| | - Juan Gabriel Vergaño-Salazar
- Doctorado en Modelamiento Matemático Aplicado, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca 3466706, Chile
| | - Luis Pastenes
- Laboratorio de Genética y Microevolución, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca 3466706, Chile
- Departamento de Biología y Química, Facultad de Ciencias Básicas, Universidad Católica del Maule, Talca 3466706, Chile
| | - Vivian D’Afonseca
- Centro de Investigación y Estudios Avanzados del Maule (CIEAM), Vicerrectoría de Investigación y Posgrado, Universidad Católica del Maule, Talca 3466706, Chile
| |
Collapse
|
29
|
Najar M, Merimi M, Faour WH, Lombard CA, Moussa Agha D, Ouhaddi Y, Sokal EM, Lagneaux L, Fahmi H. In Vitro Cellular and Molecular Interplay between Human Foreskin-Derived Mesenchymal Stromal/Stem Cells and the Th17 Cell Pathway. Pharmaceutics 2021; 13:1736. [PMID: 34684029 PMCID: PMC8537928 DOI: 10.3390/pharmaceutics13101736] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 10/06/2021] [Accepted: 10/13/2021] [Indexed: 12/13/2022] Open
Abstract
Foreskin, considered a biological waste material, has been shown to be a reservoir of therapeutic cells. The immunomodulatory properties of mesenchymal stromal/stem cells (MSCs) from the foreskin (FSK-MSCs) are being evaluated in cell-based therapy for degenerative, inflammatory and autoimmune disorders. Within the injured/inflamed tissue, proinflammatory lymphocytes such as IL-17-producing T helper cells (Th17) may interact with the stromal microenvironment, including MSCs. In this context, MSCs may encounter different levels of T cells as well as specific inflammatory signals. Uncovering the cellular and molecular changes during this interplay is central for developing an efficient and safe immunotherapeutic tool. To this end, an in vitro human model of cocultures of FSK-MSCs and T cells was established. These cocultures were performed at different cell ratios in the presence of an inflammatory setting. After confirming that FSK-MSCs respond to ISCT criteria by showing a typical phenotype and multilineage potential, we evaluated by flow cytometry the expression of Th17 cell markers IL-17A, IL23 receptor and RORγt within the lymphocyte population. We also measured 15 human Th17 pathway-related cytokines. Regardless of the T cell/MSC ratio, we observed a significant increase in IL-17A expression associated with an increase in IL-23 receptor expression. Furthermore, we observed substantial modulation of IL-1β, IL-4, IL-6, IL-10, IL-17A, IL-17F, IL-21, IL-22, IL-23, IL-25, IL-31, IL-33, INF-γ, sCD40, and TNF-α secretion. These findings suggest that FSK-MSCs are receptive to their environment and modulate the T cell response accordingly. The changes within the secretome of the stromal and immune environment are likely relevant for the therapeutic effect of MSCs. FSK-MSCs represent a valuable cellular product for immunotherapeutic purposes that needs to be further clarified and developed.
Collapse
Affiliation(s)
- Mehdi Najar
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, 1070 Brussels, Belgium; (M.N.); (L.L.)
- Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Makram Merimi
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (M.M.); (D.M.A.)
- LBBES Laboratory, Genetics and Immune Cell Therapy Unit, Faculty of Sciences, University Mohammed Premier, Oujda 60000, Morocco
| | - Wissam H. Faour
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos 5053, Lebanon;
| | - Catherine A. Lombard
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (C.A.L.); (E.M.S.)
| | - Douâa Moussa Agha
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, 1000 Brussels, Belgium; (M.M.); (D.M.A.)
| | - Yassine Ouhaddi
- Orthopaedics Division, Department of Surgery, Faculty of Medicine, McGill University, Montreal General Hospital (MGH), The Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, QC H3G 1A4, Canada;
| | - Etienne M. Sokal
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (C.A.L.); (E.M.S.)
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Jules Bordet Institute, Université Libre de Bruxelles, 1070 Brussels, Belgium; (M.N.); (L.L.)
| | - Hassan Fahmi
- Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), Montreal, QC H2X 0A9, Canada
| |
Collapse
|
30
|
Maguire PT, Loughran ST, Harvey R, Johnson PA. A TLR5 mono-agonist restores inhibited immune responses to Streptococcus pneumoniae during influenza virus infection in human monocytes. PLoS One 2021; 16:e0258261. [PMID: 34644311 PMCID: PMC8513880 DOI: 10.1371/journal.pone.0258261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 09/22/2021] [Indexed: 11/20/2022] Open
Abstract
Influenza A virus (IAV) predisposes individuals to often more severe secondary bacterial infections with Streptococcus pneumonia (S. pneumoniae). The outcomes of these infections may be made worse with the increase in antimicrobial resistance and a lack of new treatments to combat this. Th17 responses are crucial in clearing S. pneumoniae from the lung. We previously demonstrated that early IAV infection of human monocytes significantly reduced levels of S. pneumoniae-driven cytokines involved in the Th17 response. Here, we have further identified that IAV targets specific TLRs (TLR2, TLR4, TLR9) involved in sensing S. pneumoniae infection resulting, in a reduction in TLR agonist-induced IL-23 and TGF-β. The effect of IAV is more profound on the TLR2 and TLR9 pathways. We have established that IAV-mediated inhibition of TLR9-induction is related to a downregulation of RORC, a Th17 specific transcription factor. Other studies using mouse models demonstrated that TLR5 agonism improved the efficacy of antibiotics in the treatment of IAV/S. pneumoniae co-infections. Therefore, we investigated if TLR5 agonism could restore inhibited Th17 responses in human monocytes. Levels of pneumococcus-driven cytokines, which had previously been inhibited by IAV were not reduced in the presence of the TLR5 mono-agonist, suggesting that such treatment may overcome IAV inhibition of Th17 responses. The importance of our research is in demonstrating the IAV directly targets S. pneumoniae-associated TLR pathways. Additionally, the IAV-inhibition of Th17 responses can be restored by TLR5 agonism, which indicates that there may be a different Th17 signalling pathway which is not affected by IAV infection.
Collapse
Affiliation(s)
- Paula T Maguire
- Viral Immunology Laboratory, School of Nursing, Psychotherapy and Community Health, Dublin City University, Dublin, Ireland
| | - Sinéad T Loughran
- Department of Applied Science, Dundalk Institute of Technology, County Louth, Ireland
| | - Ruth Harvey
- National Institute for Biological Standards and Controls, Potters Bar, Herts, United Kingdom
| | - Patricia A Johnson
- Viral Immunology Laboratory, School of Nursing, Psychotherapy and Community Health, Dublin City University, Dublin, Ireland
| |
Collapse
|
31
|
Mitrovic S, Hassold N, Kamissoko A, Rosine N, Mathian A, Mercy G, Pertuiset E, Nocturne G, Fautrel B, Koné-Paut I. Adult-onset Still's disease or systemic-onset juvenile idiopathic arthritis and spondyloarthritis: overlapping syndrome or phenotype shift? Rheumatology (Oxford) 2021; 61:2535-2547. [PMID: 34559214 DOI: 10.1093/rheumatology/keab726] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/16/2021] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVES Systemic-onset juvenile idiopathic arthritis (SJIA) and adult-onset Still's disease (AOSD) are the same sporadic systemic auto-inflammatory disease. Spondyloarthritis (SpA) is a group of inflammatory non-autoimmune disorders. We report the observations of eight patients with SJIA/AOSD who also presented features of SpA during their disease evolution and estimate the prevalence of SpA in SJIA/AOSD. METHODS This was a retrospective national survey of the departments of paediatric and adult rheumatology and internal medicine. To be included, SJIA patients had to fulfil the ILAR criteria, AOSD patients the Yamaguchi or Fautrel criteria, and all patients the ASAS classification criteria for axial or peripheral SpA, ESSG criteria for spondyloarthropathy or CASPAR criteria for psoriatic arthritis. The data were collected with a standardized form. RESULTS Eight patients (five adults) were identified in one paediatric and two adult departments. In all but one patient, SpA manifestations occurred several years after SJIA/AOSD onset (mean delay 6.2 ± 3.8 years). Two patients had peripheral and three axial SpA, and four later exhibited psoriatic arthritis and one SAPHO syndrome. The prevalence of SpA in an adult cohort of 76 patients with AOSD was 6.58% (95% CI [2.17-14.69]), greater than the prevalence of SpA in the French general population (0.3%, 95%CI [0.17-0.46]). The prevalence of SpA in an SJIA cohort of 30 patients was 10% (95%CI [2.11-26.53]), more than that reported in the general population of industrialized countries, estimated at 0.016% to 0.15%. CONCLUSION Whilst the temporal disassociation between SpA and AOSD in most cases might suggest a coincidental finding, our work raises the possibility of an SpA AOSD spectrum overlap that needs further study.
Collapse
Affiliation(s)
- Stéphane Mitrovic
- Service de Rhumatologie, Hôpital Pitié-Salpêtrière, Paris, FranceSorbonne Université - APHP.,Centre d'Etude et de Référence sur les Maladies AutoInflammatoires et les Amyloses (CEREMAIA), FAI2R network, Paris, France.,Département de Médecine Interne, Unité de Rhumatologie, Institut Mutualiste Montsouris, Paris, France
| | - Nolan Hassold
- Center for Immunology of Viral Infections and Autoimmune Diseases, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Le Kremlin-Bicêtre, Université Paris Saclay, INSERM, Paris, France.,Service de Rhumatologie, Hôpital de Bicêtre, APHP, université de Paris sud-Saclay, Le Kremlin-Bicêtre, France.,Service de rhumatologie pédiatrique, and CEREMAIA, Hôpital de Bicêtre, APHP, université de Paris sud-Saclay, Le Kremlin-Bicêtre, France
| | - Aly Kamissoko
- Service de Rhumatologie, Hôpital Pitié-Salpêtrière, Paris, FranceSorbonne Université - APHP.,Service de Rhumatologie, Hôpital National Ignace Deen, Conakry, Guinée
| | - Nicolas Rosine
- Service de Rhumatologie, Hôpital Pitié-Salpêtrière, Paris, FranceSorbonne Université - APHP
| | - Alexis Mathian
- Service de Médecine Interne 2, Hôpital Universitaire Pitié-Salpêtrière, AP-HP, Paris, France
| | - Guillaume Mercy
- Service de Radiologie, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Edouard Pertuiset
- Centre hospitalier René Dubos, Service de rhumatologie, Pontoise, France
| | - Gaëtane Nocturne
- Center for Immunology of Viral Infections and Autoimmune Diseases, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Le Kremlin-Bicêtre, Université Paris Saclay, INSERM, Paris, France.,Service de Rhumatologie, Hôpital de Bicêtre, APHP, université de Paris sud-Saclay, Le Kremlin-Bicêtre, France
| | - Bruno Fautrel
- Service de Rhumatologie, Hôpital Pitié-Salpêtrière, Paris, FranceSorbonne Université - APHP.,Centre d'Etude et de Référence sur les Maladies AutoInflammatoires et les Amyloses (CEREMAIA), FAI2R network, Paris, France.,Institut d'Epidémiologie, et de Santé Publique Pierre Louis, UMR S 1136, Equipe PEPITES, Paris, France
| | - Isabelle Koné-Paut
- Centre d'Etude et de Référence sur les Maladies AutoInflammatoires et les Amyloses (CEREMAIA), FAI2R network, Paris, France.,Service de rhumatologie pédiatrique, and CEREMAIA, Hôpital de Bicêtre, APHP, université de Paris sud-Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
32
|
Pandiyan P, McCormick TS. Regulation of IL-17A-Producing Cells in Skin Inflammatory Disorders. J Invest Dermatol 2021; 142:867-875. [PMID: 34561088 DOI: 10.1016/j.jid.2021.06.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/09/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022]
Abstract
This review focuses on the IL-17A family of cytokines produced by T lymphocytes and other immune cells and how they are involved in cutaneous pathogenic responses. It will also discuss cutaneous dysbiosis and FOXP3+ regulatory T cells in the context of inflammatory conditions linked to IL-17 responses in the skin. Specifically, it will review key literature on chronic mucocutaneous candidiasis and psoriasis.
Collapse
Affiliation(s)
- Pushpa Pandiyan
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.
| | - Thomas S McCormick
- Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
33
|
Youssef ME, Abd El-Fattah EE, Abdelhamid AM, Eissa H, El-Ahwany E, Amin NA, Hetta HF, Mahmoud MH, Batiha GES, Gobba N, Ahmed Gaafar AG, Saber S. Interference With the AMPKα/mTOR/NLRP3 Signaling and the IL-23/IL-17 Axis Effectively Protects Against the Dextran Sulfate Sodium Intoxication in Rats: A New Paradigm in Empagliflozin and Metformin Reprofiling for the Management of Ulcerative Colitis. Front Pharmacol 2021; 12:719984. [PMID: 34489707 PMCID: PMC8417441 DOI: 10.3389/fphar.2021.719984] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 07/26/2021] [Indexed: 12/20/2022] Open
Abstract
Empagliflozin and metformin are widely used for the treatment of type 2 diabetes. These drugs showed marked anti-inflammatory effects in different animal models via enhancing AMPK activity. Yet, the protective anti-inflammatory effects of their combination against ulcerative colitis have not been previously investigated. The current study aimed to explore the potential of empagliflozin/metformin combination to mitigate the DSS-induced rat colitis model. The modulating effects of empagliflozin and metformin on the AMPK/mTOR/NLRP3 axis and T cell polarization were delineated. In this study, distal colons were examined for macroscopic and microscopic pathological alterations. ELISA, qRT-PCR, and immunohistochemistry techniques were applied to detect proteins and cytokines involved in AMPK/mTOR/NLRP3 axis and T Cell polarization. Oral administration of empagliflozin (10 mg/kg/day) and metformin (200 mg/kg/day) combination alleviated colitis as revealed by the reduced disease activity index, macroscopic damage index, colon weight/length ratio, and histopathologic scoring values. Interestingly, empagliflozin/metformin combination significantly enhanced AMPK phosphorylation and depressed mTOR and NLRP3 expression leading to a subsequent reduction in caspase-1 cleavage and inhibition of several inflammatory cytokines, including IL-1β, and IL-18. Reduced mTOR expression and reduced IL-6 levels led to a reduction in Th17 cell polarization and maintenance. Together, the current study reveals that the protective effects of empagliflozin and metformin against DSS-induced colitis are fundamentally mediated via enhancing AMPK phosphorylation. Since adult humans with diabetes mellitus are at greater risk for developing inflammatory bowel diseases, clinical application of empagliflozin/metformin combination represents a novel therapeutic approach for treating diabetic patients with ulcerative colitis.
Collapse
Affiliation(s)
- Mahmoud E Youssef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Eslam E Abd El-Fattah
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Amir M Abdelhamid
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Hanan Eissa
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Eman El-Ahwany
- Department of Immunology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Noha A Amin
- Department of Hematology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Helal F Hetta
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt.,Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Mohamed H Mahmoud
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Naglaa Gobba
- Department of Pharmacology and Toxicology, College of Pharmacy, Misr University for Science and Technology, 6th of October City, Egypt
| | - Ahmed Gaafar Ahmed Gaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Port-Said University, Port-Said, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| |
Collapse
|
34
|
Mirdamadi K, Kwok J, Nevo O, Berger H, Piquette-Miller M. Impact of Th-17 Cytokines on the Regulation of Transporters in Human Placental Explants. Pharmaceutics 2021; 13:881. [PMID: 34203644 PMCID: PMC8232183 DOI: 10.3390/pharmaceutics13060881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 11/16/2022] Open
Abstract
Activated T helper 17 (Th-17) cytokines play a role in the pathophysiology of autoimmune and infectious diseases. While these diseases affect many women of childbearing age, little is known about the effect of these cytokines on placental transporters. As several pro-inflammatory cytokines impact the expression of ABC and SLC placental transporters, we hypothesized that these transporters may be similarly altered by elevated levels of circulating Th-17 cytokines. Cultured term human villous explants were treated with IL-17A, IL-22, or IL-23, alone or in combination. Samples were analyzed using qRT-PCR and Western blotting. The mRNA expression of OATP2B1 was significantly downregulated in explants by all individual cytokines and combination treatments, while decreased protein expression was seen with IL-23 and combination (p < 0.01). Combination treatment decreased the mRNA expression of BCRP and OAT4 but increased that of OCT3 (p < 0.01). Decreased accumulation of the OATP substrate, cascade blue, was seen in IL-23-treated choriocarcinoma JAr cells (p < 0.01). Elevated Th-17 cytokines, which are seen in infectious and autoimmune diseases, affect the expression and activity of OATP2B1, as well as mRNA expression of placental BCRP, OAT4, and OCT3. This dysregulation could impact the fetal exposure to endogenous and exogenous substrates.
Collapse
Affiliation(s)
- Kamelia Mirdamadi
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; (K.M.); (J.K.); (O.N.)
| | - Jacinda Kwok
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; (K.M.); (J.K.); (O.N.)
| | - Ori Nevo
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; (K.M.); (J.K.); (O.N.)
- Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
| | - Howard Berger
- Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada;
| | - Micheline Piquette-Miller
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; (K.M.); (J.K.); (O.N.)
| |
Collapse
|
35
|
Mitrovic S, Fautrel B. Clinical Phenotypes of Adult-Onset Still's Disease: New Insights from Pathophysiology and Literature Findings. J Clin Med 2021; 10:jcm10122633. [PMID: 34203779 PMCID: PMC8232697 DOI: 10.3390/jcm10122633] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 06/10/2021] [Indexed: 01/02/2023] Open
Abstract
Adult-onset Still's disease (AOSD) is a non-familial, polygenic systemic autoinflammatory disorder. It is traditionally characterized by four cardinal manifestations-spiking fever, an evanescent salmon-pink maculopapular rash, arthralgia or arthritis and a white-blood-cell count (WBC) ≥ 10,000/mm3, mainly neutrophilic polymorphonuclear cells (PMNs)-but many other manifestations and complications can be associated, making clinical expression very heterogeneous and diagnosis sometimes difficult. The AOSD course can be diverse and is currently impossible to predict. Several clinical phenotypes have been described, either on the basis of the evolution of symptoms over time (monocyclic, polycyclic and chronic evolution) or according to dominant clinical evolution (systemic and arthritis subtypes). However, these patterns are mainly based on case series and not on robust epidemiological studies. Furthermore, they have mainly been established a long time ago, before the era of the biological treatments. Thus, based on our personal experience and on recent advances in the understanding of disease pathogenesis, it appears interesting to reshuffle AOSD phenotypes, emphasizing the continuum between AOSD profiles and other systemic autoinflammatory disorders, eventually proposing a research agenda.
Collapse
Affiliation(s)
- Stéphane Mitrovic
- Service de Rhumatologie, Hôpital Pitié-Salpêtrière, Sorbonne Université—APHP, 75013 Paris, France;
- Centre d’Etude et de Référence sur les Maladies AutoInflammatoires et les Amyloses (CEREMAIA), FAI2R Network, 75013 Paris, France
- Département de Médecine Interne, Institut Mutualiste Montsouris, 75014 Paris, France
| | - Bruno Fautrel
- Service de Rhumatologie, Hôpital Pitié-Salpêtrière, Sorbonne Université—APHP, 75013 Paris, France;
- Centre d’Etude et de Référence sur les Maladies AutoInflammatoires et les Amyloses (CEREMAIA), FAI2R Network, 75013 Paris, France
- Institut d’Epidémiologie et de Santé Publique Pierre Louis, UMR S 1136, Equipe PEPITES, 75013 Paris, France
- Correspondence:
| |
Collapse
|
36
|
Wang X, Chen H, Jiang R, Hong X, Peng J, Chen W, Jiang J, Li J, Huang D, Dai H, Wang W, Lu J, Zhao Y, Wu W. Interleukin-17 activates and synergizes with the notch signaling pathway in the progression of pancreatic ductal adenocarcinoma. Cancer Lett 2021; 508:1-12. [PMID: 33713738 DOI: 10.1016/j.canlet.2021.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 02/27/2021] [Accepted: 03/03/2021] [Indexed: 12/28/2022]
Abstract
Interleukin (IL)-17 is a prominent cytokine that promotes pancreatic intraepithelial neoplasia (PanIN) and pancreatic ductal adenocarcinoma (PDAC) and is associated with the oncogenic pathways in tumor progression. However, the mechanism and therapeutic value of the IL-17 axis remain unclear. In this study, we verified the activation of the IL-17 and Notch pathways in PanIN/PDAC via complementary approaches and validated their pro-tumor effects on tumor progression. Additionally, we found a positive correlation between IL-17 and Notch; the IL-17 axis can upregulate Notch activity via the canonical NF-κB pathway in vitro, thus synergistically promoting PanIN/PDAC. Furthermore, we observed that the co-inhibition of IL-17 and the Notch pathway can enhance the therapeutic effect by restricting tumor growth in vivo. Our study highlights the synergistic effect of the IL-17 axis and Notch pathway in promoting PanIN/PDAC and further suggests that IL-17-Notch co-inhibition is a novel therapeutic strategy with superior potential in treating PDAC.
Collapse
Affiliation(s)
- Xianze Wang
- Department of Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Hao Chen
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Rui Jiang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Xiafei Hong
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Junya Peng
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Wenyan Chen
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Jialin Jiang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Jie Li
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Dan Huang
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Hongmei Dai
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Wenze Wang
- Department of Pathology, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Junliang Lu
- Department of Pathology, Molecular Pathology Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Yupei Zhao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China.
| | - Wenming Wu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
37
|
Kim ST, Pundole X, Dadu R, Lambotte O, Ramos-Casals M, Suarez-Almazor ME. Use of immune checkpoint inhibitors in cancer patients with pre-existing sarcoidosis. Immunotherapy 2021; 13:465-475. [PMID: 33641345 DOI: 10.2217/imt-2020-0272] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Aim: To evaluate adverse events in cancer patients with pre-existing sarcoidosis receiving immune checkpoint inhibitors (ICIs). Patients & methods: We retrospectively reviewed cancer patients with sarcoidosis who underwent treatment with ICI to determine frequency of sarcoidosis flares. Results: 32 patients with sarcoidosis received ICIs The median time to ICI initiation was 7 years (range: 1 month to 51 years). One patient (3%) with a 20-year remote history of sarcoidosis developed a clinically symptomatic exacerbation after three doses of atezolizumab, with hilar lymphadenopathy, subcutaneous nodules, arthritis and uveitis. Atezolizumab was discontinued and prednisone initiated. She had a fluctuating course with two additional flares. Conclusion: Frequency of flares in patients with a remote history of sarcoidosis who receive ICIs is low.
Collapse
Affiliation(s)
- Sang T Kim
- Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xerxes Pundole
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ramona Dadu
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Olivier Lambotte
- AP-HP. Université Paris-Saclay, Hôpital Bicêtre Department of Internal Medicine & Clinical Immunology, Le Kremlin Bicêtre, France.,Université Paris-Saclay; INSERM; CEA, Centre Immunology of Viral Infections & Autoimmune Diseases, IDMIT Department, IBFJ, Le Kremlin-Bicêtre, France
| | - Manuel Ramos-Casals
- Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain.,Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Barcelona, Spain.,Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Maria E Suarez-Almazor
- Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
38
|
Yu M, Pal S, Paterson CW, Li JY, Tyagi AM, Adams J, Coopersmith CM, Weitzmann MN, Pacifici R. Ovariectomy induces bone loss via microbial-dependent trafficking of intestinal TNF+ T cells and Th17 cells. J Clin Invest 2021; 131:143137. [PMID: 33586672 PMCID: PMC7880410 DOI: 10.1172/jci143137] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023] Open
Abstract
Estrogen deficiency causes a gut microbiome-dependent expansion of BM Th17 cells and TNF-α-producing T cells. The resulting increased BM levels of IL-17a (IL-17) and TNF stimulate RANKL expression and activity, causing bone loss. However, the origin of BM Th17 cells and TNF+ T cells is unknown. Here, we show that ovariectomy (ovx) expanded intestinal Th17 cells and TNF+ T cells, increased their S1P receptor 1-mediated (S1PR1-mediated) egress from the intestine, and enhanced their subsequent influx into the BM through CXCR3- and CCL20-mediated mechanisms. Demonstrating the functional relevance of T cell trafficking, blockade of Th17 cell and TNF+ T cell egress from the gut or their influx into the BM prevented ovx-induced bone loss. Therefore, intestinal T cells are a proximal target of sex steroid deficiency relevant for bone loss. Blockade of intestinal T cell migration may represent a therapeutic strategy for the treatment of postmenopausal bone loss.
Collapse
Affiliation(s)
- Mingcan Yu
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
| | - Subhashis Pal
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
| | - Cameron W. Paterson
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
- Medical Corps, United States Navy, NROTC, Atlanta, Georgia, USA
| | - Jau-Yi Li
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
| | - Abdul Malik Tyagi
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
| | - Jonathan Adams
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
| | - Craig M. Coopersmith
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - M. Neale Weitzmann
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
- Atlanta VA Medical Center, Decatur, Georgia, USA
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine and
- Emory Microbiome Research Center, Emory University, Atlanta, Georgia, USA
- Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
39
|
Tian YM, He HL, Cheng YT, Yi L, Yang Y, Yang P. A Combined Phytochemical and Network Pharmacology Approach to Reveal the Effective Substances and Mechanism of Eomecon chionantha Hance for the Treatment of Ulcerative Colitis. Nat Prod Commun 2021; 16. [DOI: 10.1177/1934578x21992966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
Eomecon chionantha Hance (ECH), a traditional Chinese herbal medicine, has been reported for the treatment of traumatic injuries and colitis. The current treatments for ulcerative colitis (UC) are unstable and have side effects, so ECH is potentially useful for treating this condition. However, the active ingredients and pharmacological mechanisms of ECH in treating UC remain unclear. In this study, 21 alkaloids were extracted and purified from the roots of ECH, among which 13 were extracted from this herb for the first time. Our results showed that 12 ingredients may have effective pharmacological effects after absorption, distribution, metabolism, and excretion (ADME) screening. Network pharmacological analysis revealed that the active ingredients may have positive effects on 19 significant signaling pathways, such as on small cell lung cancer, serotonergic synapses, the IL-17 signaling pathway, Th17 cell differentiation, the estrogen signaling pathway, transcriptional misregulation in cancer, the PI3K-Akt signaling pathway, and others by targeting 23 proteins, including MAPK14, RXRA, GSK3B, CDK2, RXRB, HSP90AA1, PTGS2, and ESR1. It is of great benefit to use separation, purification, and network pharmacology together to screen active natural products. This study indicated potential anti-UC mechanisms of the active ingredients of ECH and provides theoretical support for the treatment of UC using ECH.
Collapse
Affiliation(s)
- Yu-mei Tian
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, School of Nursing, Hunan University of Medicine, Huaihua 418000, China
| | - Hai-lang He
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, School of Nursing, Hunan University of Medicine, Huaihua 418000, China
| | - Ya-ting Cheng
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, School of Nursing, Hunan University of Medicine, Huaihua 418000, China
| | - Li Yi
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, School of Nursing, Hunan University of Medicine, Huaihua 418000, China
| | - Yuan Yang
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, School of Nursing, Hunan University of Medicine, Huaihua 418000, China
- Dong Medicine Key Laboratory of Hunan Province, Department of Laboratory Medicine, Hunan University of Medicine, Huaihua 418000, China
- Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, Hunan University of Medicine, Huaihua 418000, China
| | - Peng Yang
- Hunan Province Key Laboratory for Antibody-based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, School of Nursing, Hunan University of Medicine, Huaihua 418000, China
- Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, Hunan University of Medicine, Huaihua 418000, China
| |
Collapse
|
40
|
Zhang H, Zhang F, Li W. Function of intestinal barrier protected by regulating the miR-199a-3p in ulcerative colitis: Modulation of IL-23/IL-17A axis. Fundam Clin Pharmacol 2021; 35:852-860. [PMID: 33475196 DOI: 10.1111/fcp.12650] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/11/2021] [Indexed: 12/19/2022]
Abstract
Ulcerative colitis is a chronic inflammatory bowel disorder, which is having higher mortality rate. The present report evaluates the protective effect of miR-199a-3p oligomer for the treatment of Ulcerative colitis (UC). Ulcerative colitis was induced by administration of dextran sulfate sodium [DSS (3%)] with drinking water for the duration of one week in mice and miR-199a-3p oligomer was treated for the same duration. Effect of miR-199a-3p oligomer was determined by estimating the body weight, blood stool and length of colon in UC mice. Inflammatory cytokines, oxidative stress parameters and Treg/Th17 ratio was determined in the serum, intestinal and spleen tissue of UC mice. mRNA expression of TGFβ, Foxp3, RORγt and STAT3 was estimated in the intestinal tissue of UC mice. Moreover, permeability of intestine was determined by estimating the expression of FITC-dextran in the serum and expression of junction protein in the tissue of UC mice. The data of the study suggest that treatment with miR-199a-3p oligomer ameliorates the altered condition in ulcerative colitis mice. There was reduction in the level of cytokines and parameters of oxidative stress in the intestine of miR-199a-3p oligomer than UC group. Moreover, intestinal permeability was enhanced in miR-199a-3p oligomer treated UC mice. The level of Th17 in the serum and mRNA expression of TGFβ, Foxp3, RORγt and STAT3 was attenuated in miR-199a-3p oligomer treated UC mice. In conclusion, the data of the study suggest that treatment with miR-199a-3p oligomer ameliorates intestinal barrier in ulcerative colitis by down regulating the IL-17A/IL-23 axis.
Collapse
Affiliation(s)
- Huixia Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fangbin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Li
- Department of Physiology and Neurobiology, School of Basic Medicine Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
41
|
Immunology Basics of Rheumatic Disease. PHYSICIAN ASSISTANT CLINICS 2021. [DOI: 10.1016/j.cpha.2020.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
42
|
Kim J, Hope CM, Perkins GB, Stead SO, Scaffidi JC, Kette FD, Carroll RP, Barry SC, Coates PT. Rapamycin and abundant TCR stimulation are required for the generation of stable human induced regulatory T cells. Clin Transl Immunology 2020; 9:e1223. [PMID: 33425354 PMCID: PMC7780108 DOI: 10.1002/cti2.1223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 07/07/2020] [Accepted: 11/12/2020] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES Regulatory T cells (Tregs) are a vital sub-population of CD4+ T cells with major roles in immune tolerance and homeostasis. Given such properties, the use of regulatory T cells for immunotherapies has been extensively investigated, with a focus on adoptive transfer of ex vivo expanded natural Tregs (nTregs). For immunotherapies, induced Tregs (iTregs), generated in vitro from naïve CD4+ T cells, provide an attractive alternative, given the ease of generating cell numbers required for clinical dosage. While the combination of TGF-β, ATRA and rapamycin has been shown to generate highly suppressive iTregs, the challenge for therapeutic iTreg generation has been their instability. Here, we investigate the impact of rapamycin concentrations and α-CD3/CD28 bead ratios on human iTreg stability. METHODS We assess iTregs generated with various concentrations of rapamycin and differing ratios of α-CD3/CD28 beads for their differentiation, stability, expression of Treg signature molecules and T helper effector cytokines, and Treg-specific demethylation region (TSDR) status. RESULTS iTregs generated in the presence of TGF-β, ATRA, rapamycin and a higher ratio of α-CD3/CD28 beads were highly suppressive and stable upon in vitro re-stimulation. These iTregs exhibited a similar expression profile of Treg signature molecules and T helper effector cytokines to nTregs, in the absence of TSDR demethylation. CONCLUSION This work establishes a method to generate human iTregs which maintain stable phenotype and function upon in vitro re-stimulation. Further validation in pre-clinical models will be needed to ensure its suitability for applications in adoptive transfer.
Collapse
Affiliation(s)
- Juewan Kim
- The Department of Molecular & Biomedical ScienceThe School of Biological SciencesThe Faculty of SciencesThe University of AdelaideAdelaideSAAustralia
| | - Christopher M Hope
- Department of GastroenterologyWomen’s and Children’s HospitalAdelaideSAAustralia
- Molecular Immunology GroupRobinson Research InstituteSchool of MedicineThe University of AdelaideAdelaideSAAustralia
| | - Griffith B Perkins
- The Department of Molecular & Biomedical ScienceThe School of Biological SciencesThe Faculty of SciencesThe University of AdelaideAdelaideSAAustralia
| | - Sebastian O Stead
- Discipline of MedicineSchool of MedicineThe University of AdelaideAdelaideSAAustralia
- College of Medicine and Public HealthDiscipline of MedicineFlinders UniversityBedford ParkSAAustralia
| | - Jacqueline C Scaffidi
- Discipline of MedicineSchool of MedicineThe University of AdelaideAdelaideSAAustralia
| | - Francis D Kette
- Discipline of MedicineSchool of MedicineThe University of AdelaideAdelaideSAAustralia
- College of Medicine and Public HealthDiscipline of MedicineFlinders UniversityBedford ParkSAAustralia
| | - Robert P Carroll
- Discipline of MedicineSchool of MedicineThe University of AdelaideAdelaideSAAustralia
- Central Northern Adelaide Renal and Transplantation Service (CNARTS)The Royal Adelaide HospitalAdelaideSAAustralia
- Division of Medical SciencesUniversity of South AustraliaAdelaideSAAustralia
| | - Simon C Barry
- Department of GastroenterologyWomen’s and Children’s HospitalAdelaideSAAustralia
- Molecular Immunology GroupRobinson Research InstituteSchool of MedicineThe University of AdelaideAdelaideSAAustralia
| | - Patrick Toby Coates
- Discipline of MedicineSchool of MedicineThe University of AdelaideAdelaideSAAustralia
- Central Northern Adelaide Renal and Transplantation Service (CNARTS)The Royal Adelaide HospitalAdelaideSAAustralia
| |
Collapse
|
43
|
Adefegha SA, Bottari NB, Leal DB, de Andrade CM, Schetinger MR. Interferon gamma/interleukin-4 modulation, anti-inflammatory and antioxidant effects of hesperidin in complete Freund's adjuvant (CFA)-induced arthritis model of rats. Immunopharmacol Immunotoxicol 2020; 42:509-520. [PMID: 32838587 DOI: 10.1080/08923973.2020.1814806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND This study sought to assess the effect of hesperidin on serum inflammatory cytokines and oxidative damage in liver of complete Freund's adjuvant (CFA)-induced arthritic rats. METHOD Fifty-six adult female Wistar rats (220-250 g) were acclimatized for two weeks. Intraplantar injection of CFA was done for the induction of arthritis and confirmed on the 14th day prior to oral administration of 40 and 80 mg/kg of hesperidin or dexamethasone for 45 days. RESULT The result showed that treatment with both doses of hesperidin and dexamethasone in the joint of arthritic rats significantly (p < .05) diminished paw swelling/edema and arthritis score as well as enhanced latency in thermal hyperalgesia test. In addition, hesperidin treatment in arthritis rats showed significant (p < .01) improvement in red blood cells and platelets counts as well as hemoglobin and hematocrit compared to the arthritis control rat group. Furthermore, hesperidin treatment significantly (p < .05) reduced serum interferon gamma (IFN-γ) and interleukin-4 (IL-4) levels in arthritic rat. In addition, treatment with hesperidin significantly (p < .05) decreased the liver of thiobarbituric acid reactive species and reactive oxygen species levels but raised the levels of total and non-protein thiols of rat induced with CFA. The reduced activities of liver δ-aminolevulinate dehydratase, catalase, glutathione-S transferase in arthritic rats were significantly (p < .05) increased with hesperidin treatment in arthritic rats. This study suggests that hesperidin demonstrated an anti-arthritic effect via modulation of serum IFN-γ and IL-4 levels as well as protection against oxidative damage. CONCLUSION Hence, hesperidin could be a potential immune-modulatory, anti-inflammatory and anti-oxidant agent.
Collapse
Affiliation(s)
- Stephen Adeniyi Adefegha
- Department of Biochemistry, Federal University of Technology, Akure, Nigeria.,Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria-RS, Brazil.,Departamento de Microbiologia, Immunologia e Parasitologia, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria-RS, Brazil
| | - Nathieli Bianchin Bottari
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria-RS, Brazil
| | - Daniela Bitencourt Leal
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria-RS, Brazil.,Departamento de Microbiologia, Immunologia e Parasitologia, Centro de Ciências da Saúde, Universidade Federal de Santa Maria, Santa Maria-RS, Brazil
| | - Cínthia Melazzo de Andrade
- Programa de Pós graduação em Medicina Veterinária, Centro de Ciência Rurais, Departamento de Clínica de Pequenos Animais, Laboratório de Patologia Clínica Veternária, Hospital Veterinário, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Maria Rosa Schetinger
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria-RS, Brazil
| |
Collapse
|
44
|
Gautam S, Kumar M, Kumar U, Dada R. Effect of an 8-Week Yoga-Based Lifestyle Intervention on Psycho-Neuro-Immune Axis, Disease Activity, and Perceived Quality of Life in Rheumatoid Arthritis Patients: A Randomized Controlled Trial. Front Psychol 2020; 11:2259. [PMID: 32982898 PMCID: PMC7492675 DOI: 10.3389/fpsyg.2020.02259] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 08/11/2020] [Indexed: 12/15/2022] Open
Abstract
Various external stressors and environmental challenges lead to the provocation of the immune system in autoimmune diseases like Rheumatoid arthritis (RA). The inappropriate immune response further triggers the cascade of inflammatory changes resulting in precipitation of symptoms and hampers quality of life (QOL). The underlying psycho-somatic component of the disease requires a holistic approach to its treatment dimension rather than the use of pharmacotherapy. The applicability of mind-body interventions has become essential in today’s fast-paced life. Yoga, a mind-body technique, alters the mind’s capacity to facilitate systemic functioning at multiple organ system levels. Hence, we conducted this study to evaluate the impact of 8 weeks of a yoga-based lifestyle intervention (YBLI) on psycho-neuro-immune markers, gene expression patterns, and QOL in RA patients on routine medical therapy. A total of 66 patients were randomized into two groups: yoga group or non-yoga group and were assessed for a panel of inflammatory cytokines (IL-6, IL-17A, TNF-α, and TGF-β), mind-body communicative markers (BDNF, DHEAS, β-endorphin, and sirtuin) and transcript levels of various genes (IL-6, TNF-α, NFKB1, TGF-β, and CTLA4). We assessed disease activity and QOL using the DAS28-ESR and WHOQOL-BREF questionnaire, respectively. Yoga group observed significant improvements in the levels of markers, which influenced the psycho-neuro-immune axis (p < 0.001) with an estimated effect size from small to medium range. In the yoga group, there was a significant reduction in DAS28-ESR (p < 0.001) and improvement seen in the physical health, psychological, social relationships domains (p < 0.001) of QOL, except environmental (p > 0.05). The yoga group showed downregulation of IL-6, TNF-α, and CTLA4 and upregulation of TGF-β. These results suggest that a decrease in disease activity after yoga practice is associated with a significant reduction in inflammatory cytokines, the elevation of mind-body communicative markers, and normalization of various transcript levels, which improved QOL. Thus the adoption of YBLI improves clinical outcome in RA, and decreases systemic inflammation by its beneficial effects on psycho-neuro-immune axis and normalization of dysregulated transcripts. Thus YBLI may be used for RA patients as an adjunctive therapy.
Collapse
Affiliation(s)
- Surabhi Gautam
- Lab for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Manoj Kumar
- Lab for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Uma Kumar
- Department of Rheumatology, All India Institute of Medical Sciences, New Delhi, India
| | - Rima Dada
- Lab for Molecular Reproduction and Genetics, Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
45
|
Jafarzadeh A, Nemati M, Chauhan P, Patidar A, Sarkar A, Sharifi I, Saha B. Interleukin-27 Functional Duality Balances Leishmania Infectivity and Pathogenesis. Front Immunol 2020; 11:1573. [PMID: 32849534 PMCID: PMC7427467 DOI: 10.3389/fimmu.2020.01573] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/15/2020] [Indexed: 01/10/2023] Open
Abstract
IL-27 is a cytokine that exerts diverse effects on the cells of innate and adaptive immune systems. Chiefly expressed in macrophages and dendritic cells during the early phase of Leishmania infection, IL-27 contributes to the protection against L. major infection but suppresses the protective Th1 response against L. donovani, L. infantum, L. amazonensis and L. braziliensis infections, suggesting its functional duality. During the late stage of Leishmania infection, IL-27 limits the immunopathogenic reactions and tissue damages. Herein, we analyze the mechanism of the functional duality of IL-27 in the resistance or susceptibility to Leishmania infection, prompting IL-27 for anti-Leishmanial therapy.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Maryam Nemati
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Haematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | | | | | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar, India
| | - Iraj Sharifi
- Leishmaniasis Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Bhaskar Saha
- National Centre for Cell Science, Pune, India
- Trident Academy of Creative Technology, Bhubaneswar, India
| |
Collapse
|
46
|
Bailey C, Holland JW, Secombes CJ, Tafalla C. A portrait of the immune response to proliferative kidney disease (PKD) in rainbow trout. Parasite Immunol 2020; 42:e12730. [PMID: 32403171 PMCID: PMC7507176 DOI: 10.1111/pim.12730] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/31/2020] [Accepted: 05/06/2020] [Indexed: 12/17/2022]
Abstract
Proliferative kidney disease (PKD), caused by the myxozoan Tetracapsuloides bryosalmonae, is one of the most serious parasitic diseases of salmonids in which outbreaks cause severe economic constraints for the aquaculture industry and declines of wild species throughout Europe and North America. Given that rainbow trout (Oncorhynchus mykiss) is one of the most widely farmed freshwater fish and an important model species for fish immunology, most of the knowledge on how the fish immune response is affected during PKD is from this organism. Once rainbow trout are infected, PKD pathogenesis results in a chronic kidney immunopathology mediated by decreasing myeloid cells and increasing lymphocytes. Transcriptional studies have revealed the regulation of essential genes related to T-helper (Th)-like functions and a dysregulated B-cell antibody type response. Recent reports have discovered unique details of teleost B-cell differentiation and functionality and characterized the differential immunoglobulin (Ig)-mediated response. These studies have solidified the rainbow trout T. bryosalmonae system as a sophisticated disease model capable of feeding key advances into mainstream immunology and have contributed essential information to design novel parasite disease prevention strategies. In our following perspective, we summarize these efforts to evaluate the immune mechanisms of rainbow trout during PKD pathogenesis.
Collapse
Affiliation(s)
- Christyn Bailey
- Centro de Investigación en Sanidad Animal (CISA‐INIA)MadridSpain
| | - Jason W. Holland
- Aberdeen Oomycete LaboratoryInstitute of Medical SciencesUniversity of AberdeenAberdeenUK
| | - Christopher J. Secombes
- Scottish Fish Immunology Research CentreInstitute of Biological and Environmental SciencesUniversity of AberdeenAberdeenUK
| | - Carolina Tafalla
- Centro de Investigación en Sanidad Animal (CISA‐INIA)MadridSpain
| |
Collapse
|
47
|
Al Mamun A, Akter A, Hossain S, Sarker T, Safa SA, Mustafa QG, Muhammad SA, Munir F. Role of NLRP3 inflammasome in liver disease. J Dig Dis 2020; 21:430-436. [PMID: 32585073 DOI: 10.1111/1751-2980.12918] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 05/17/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022]
Abstract
Inflammasomes have become an important natural sensor of host immunity, and can protect various organs against pathogenic infections, metabolic syndromes, cellular stress and cancer metastasis. Inflammasomes are intracellular multi-protein complexes found in both parenchymal and non-parenchymal cells, stimulating the initiation of caspase-1 and interleukin (IL)-1β and IL-18 in response to cell danger signals. Inflammasomes induce cell death mechanisms. The potential role of NOD-like receptor protein 3 (NLRP3) inflammasome in alcoholic and non-alcoholic steatohepatitis, hepatitis, nanoparticle-induced liver injury and other liver diseases has recently attracted widespread attention from clinicians and researchers. In this review we summarize the role played by the NLRP3 inflammasome in molecular and pathophysiological mechanisms in the pathogenesis and progression of liver disease. This article aims to establish that targeting the NLRP3 inflammasome and other inflammasome components may make a significant therapeutic approach to the treatment of liver disease.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Afroza Akter
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Sukria Hossain
- Department of Pharmacy, North South University, Dhaka, Bangladesh
| | - Tamanna Sarker
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | | | - Quazi G Mustafa
- School of International Studies, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Syed A Muhammad
- Institute of Molecular Biology and Biotechnology, Bahaudin Zakariya University, Multan, Pakistan
| | - Fahad Munir
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| |
Collapse
|
48
|
Araujo Flores GV, Sandoval Pacheco CM, Sosa Ochoa WH, Gomes CMC, Zúniga C, Corbett CP, Laurenti MD. Th17 lymphocytes in atypical cutaneous leishmaniasis caused by Leishmania (L.) infantum chagasi in Central America. Parasite Immunol 2020; 42:e12772. [PMID: 32603482 DOI: 10.1111/pim.12772] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/21/2020] [Accepted: 06/22/2020] [Indexed: 12/20/2022]
Abstract
Skin lesions in nonulcerated cutaneous leishmaniasis (NUCL) caused by Leishmania (L.) infantum chagasi are characterized by a mononuclear inflammatory infiltrate in the dermis, which is composed mainly of lymphocytes, followed by macrophages, few plasma cells and epithelioid granulomas with mild tissue parasitism. Previous studies have shown that the main population of lymphocytes present in the dermal infiltrate is CD8+ T cells, followed by CD4+ T cells, which are correlated with IFN-γ+ cells. To improve the knowledge of cellular immune responses in NUCL, skin biopsies were submitted to immunohistochemistry using anti-ROR-γt, anti-IL-17, anti-IL-6, anti-TGF-β, and anti-IL-23 antibodies to characterize the involvement of Th17 cells in the skin lesions of patients affected by NUCL. ROR-γt+ , IL-17+ , IL-6+ , TGF-β+ and IL-23+ cells were observed in the dermal inflammatory infiltrate of NUCL skin lesions. A positive correlation between CD4+ T-lymphocytes and ROR-γt+ and IL-17+ cells suggests that some of the CD4+ T-lymphocytes in NUCL could be Th17 lymphocytes. Moreover, a positive correlation between ROR-γt+ cells and TGF-β+ , IL-6+ , IL-17+ and IL-23+ cells could indicate the role of these cytokines in the differentiation and maintenance of Th17 lymphocytes. Our findings improve knowledge of the pathogenesis of this rare and atypical clinical form of leishmaniasis.
Collapse
Affiliation(s)
| | | | - Wilfredo Humberto Sosa Ochoa
- Laboratory of Pathology of Infectious Diseases, Medical School, São Paulo University, São Paulo, Brazil.,Microbiology Research Institute, National Autonomous University of Honduras, Tegucigalpa, Honduras
| | | | - Concepción Zúniga
- Health Surveillance Department, University School Hospital, Tegucigalpa, Honduras
| | - Carlos P Corbett
- Laboratory of Pathology of Infectious Diseases, Medical School, São Paulo University, São Paulo, Brazil
| | - Marcia Dalastra Laurenti
- Laboratory of Pathology of Infectious Diseases, Medical School, São Paulo University, São Paulo, Brazil
| |
Collapse
|
49
|
Ter Haar NM, Jansen MHA, Frenkel JF, Vastert SJ. How autoinflammation may turn into autoimmune inflammation: Insights from monogenetic and complex IL-1 mediated auto-inflammatory diseases. Clin Immunol 2020; 219:108538. [PMID: 32681980 DOI: 10.1016/j.clim.2020.108538] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 06/14/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022]
Abstract
IL-1 mediated auto-inflammatory diseases are characterised by episodes of unexplained fever, generalized and localized inflammation. The characteristic symptoms predominantly result from exaggerated activation of innate immune pathways. However, in some patients with typical IL-1 mediated diseases, chronic disease manifestations develop in the absence of acute inflammation, suggesting the involvement of adaptive immune pathways. We discuss clinical observations as well as novel insights in how chronic activation of innate immune pathways can lead to auto-immune disease features in patients with auto-inflammatory diseases and how we need to better understand these sequelae in order to improve treatment strategies.
Collapse
Affiliation(s)
- N M Ter Haar
- Department of Pediatric Rheumatology and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands; Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, the Netherlands
| | - M H A Jansen
- Department of Pediatric Rheumatology and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - J F Frenkel
- Department of Pediatric Rheumatology and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - S J Vastert
- Department of Pediatric Rheumatology and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands; Center for Translational Immunology (CTI), University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
50
|
Vitiello GA, Miller G. Targeting the interleukin-17 immune axis for cancer immunotherapy. J Exp Med 2020; 217:jem.20190456. [PMID: 31727783 PMCID: PMC7037254 DOI: 10.1084/jem.20190456] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/23/2019] [Accepted: 10/08/2019] [Indexed: 12/12/2022] Open
Abstract
IL-17 plays versatile roles during tumorigenesis. Here, Vitiello and Miller summarize current knowledge in harnessing IL-17–producing γδ and Th17 cells for successful cancer immunotherapy. The role of IL-17 in cancer remains controversial. Emerging evidence suggests that during early oncogenesis IL-17 supports tumor growth, whereas in established tumors IL-17 production by γδ and Th17 cells potentiates antitumor immunity. Consequently, γδ and Th17 cells are attractive targets for immunotherapy in the IL-17 immune axis. To optimize IL-17–based immunotherapy, a deeper understanding of the cytokines dictating IL-17 production and the polarity of γδ and Th17 cells is critical. Here, we delve into the dichotomous roles of IL-17 in cancer and provide insight into the tumor microenvironment conducive for successful IL-17–based γδ and Th17 cell immunotherapy.
Collapse
Affiliation(s)
- Gerardo A Vitiello
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY
| | - George Miller
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, NY
| |
Collapse
|