1
|
Li L, Wu H, Yao L, Yan J, Tao A. Skin inflammation and lipid disorder induced by topical application of Mal f 1. Int Immunopharmacol 2025; 148:114051. [PMID: 39823796 DOI: 10.1016/j.intimp.2025.114051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/20/2025]
Abstract
OBJECTED Mal f 1, the first allergen cloned from Malassezia furfur, has been found to have positive IgE reactivity in sera from patients with skin inflammation. In vitro, it has also been shown to induce maturation of dendritic cells and release inflammatory factors. However, its role in skin lipid homeostasis remains largely unexplored. METHODS A mouse model of skin inflammation was established by intradermal injection of Mal f 1 into the ear. Metabolomic analyses were used to study the potential mechanisms of Mal f 1-induced skin inflammation. RESULTS Subcutaneous injection of Mal f 1 can successfully induce skin inflammation, including skin thickening, inflammatory cell infiltration and the increase of various inflammatory factors. Mal f 1 caused significant changes in 102 lipid metabolites compared with the control group, which includes metabolites in arachidonic acid metabolism, glycerophospholipid metabolism, linoleic acid metabolism, and cholesterol biosynthesis. CONCLUSIONS The present study analyze the characteristics of Mal f 1-induced cutaneous inflammation and the associated disorders in skin lipid metabolism, thereby offering valuable insights for future investigations into the pathophysiology, prevention, and treatment strategies for these conditions.
Collapse
Affiliation(s)
- Linmei Li
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Hanxi Wu
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Li Yao
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Jie Yan
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China.
| | - Ailin Tao
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Immunology, The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Agger AE, Samara A, Geng T, Olstad OK, Reseland JE. Mimicking and in vitro validating chronic inflammation in human gingival fibroblasts. Arch Oral Biol 2025; 169:106113. [PMID: 39447377 DOI: 10.1016/j.archoralbio.2024.106113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
OBJECTIVE The aim of this study was to identify and validate in vitro conditions that may mimic the translational, cytokine and chemokine profiles observed in human inflamed gingiva in vivo. DESIGN Primary human gingiva fibroblast cells (HFIB-G) were cultured under serum starvation conditions (0 - 10 %), supplemented with increasing lipopolysaccharide (LPS) concentrations (0.1, 1, or 10 µg/ml) from two bacterial strains E. coli and P. gingivalis and 0.1, 1, or 10 ng/ml recombinant interleukin 1β (IL-1β), alone or in combinations. The levels of cytokines/chemokines were measured in the cell culture medium by Luminex, and gene expression was quantified by Affymetrix microarrays at 24, 48 and 72 h. RESULTS Inflammation markers were not elevated after stimulation with P. gingivalis LPS, while E. coli LPS and IL-1β individually increased the secretion of interleukin 6 (IL-6) and monocyte chemoattractant protein-1 (MCP-1) to the cell culture medium. IL-1β administration also increased the secretion of several factors, including tumor necrosis factor (TNFα). However, the combination of 1 µg/ml E. coli LPS, 1 ng/ml IL-1β and serum starvation led to increased secretion of IL-6, TNFα, in addition to other factors found in inflamed tissue. Gene expression analyses revealed that this combination not only enhanced the expression interleukins/chemokines genes but also T helper cell signaling and matrix metalloproteinases. CONCLUSION Serum reduction in cell culture medium together with the administration of E. coli LPS and IL-1β resulted in gene expression and secreted cytokine/chemokine profiles similar to that found in vivo during chronic inflammation.
Collapse
Affiliation(s)
- Anne Eriksson Agger
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway; FUTURE, Center for Functional Tissue Reconstruction, University of Oslo, Oslo, Norway.
| | - Athina Samara
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway; FUTURE, Center for Functional Tissue Reconstruction, University of Oslo, Oslo, Norway.
| | - Tianxiang Geng
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway.
| | | | - Janne Elin Reseland
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway; FUTURE, Center for Functional Tissue Reconstruction, University of Oslo, Oslo, Norway.
| |
Collapse
|
3
|
Nomura T, Muramatsu K, Yaguchi H, Uwatoko H, Kawashima A, Izumi K, Ujiie H, Fukazawa T, Yabe I. Three cases of multiple sclerosis presenting with palmoplantar pustulosis while receiving ofatumumab. J Neurol Sci 2024; 467:123315. [PMID: 39579684 DOI: 10.1016/j.jns.2024.123315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/26/2024] [Accepted: 11/16/2024] [Indexed: 11/25/2024]
Affiliation(s)
- Taichi Nomura
- Sapporo Neurology Hospital, Japan; Department of Neurology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Ken Muramatsu
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroaki Yaguchi
- Sapporo Neurology Hospital, Japan; Department of Neurology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
| | - Hisashi Uwatoko
- Sapporo Neurology Hospital, Japan; Department of Neurology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | | | - Kentaro Izumi
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hideyuki Ujiie
- Department of Dermatology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | | | - Ichiro Yabe
- Department of Neurology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
4
|
Hao L, Yang J, Lian B, Yin C, Xiao Y, Liu Y. Study on serum TL1A levels and their correlation with Th17 cells, IL-17 and IL-21 in children with Graves' disease. Front Immunol 2024; 15:1455025. [PMID: 39735549 PMCID: PMC11671471 DOI: 10.3389/fimmu.2024.1455025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/26/2024] [Indexed: 12/31/2024] Open
Abstract
Objective To investigate serum TL1A levels and their correlation with Th17 cells, IL-17, and IL-21 in children with Graves' disease (GD). Methods Thirty-seven children (12 males and 25 females) aged 9-14 years with newly diagnosed and untreated GD were enrolled in this study. Serum TL1A, IL-17, and IL-21 levels were measured using enzyme-linked immunosorbent assay (ELISA). The percentage of Th17 cells in peripheral blood was determined by flow cytometry. The correlation between serum TL1A levels and Th17 cells, IL-17, and IL-21 was analyzed using Pearson's correlation coefficient. Results Serum TL1A levels and the percentage of Th17 cells were significantly higher in children with GD compared to healthy controls (P<0.05). Serum IL-17 and IL-21 levels were also significantly elevated in GD patients (P<0.05). Serum TL1A levels positively correlated with the percentage of Th17 cells (r=0.625, P<0.05), IL-17 (r=0.573, P<0.05), and IL-21 (r=0.542, P<0.05) in children with GD. Conclusion Serum TL1A levels are increased in children with GD and positively correlate with Th17 cells, IL-17, and IL-21, suggesting that TL1A may play a role in the pathogenesis of GD by regulating Th17 cell differentiation and the production of IL-17 and IL-21.
Collapse
Affiliation(s)
- Lijun Hao
- Neonatal Department, Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| | - Jiong Yang
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Biyao Lian
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yanfeng Xiao
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yuesheng Liu
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
5
|
Chandwaskar R, Dalal R, Gupta S, Sharma A, Parashar D, Kashyap VK, Sohal JS, Tripathi SK. Dysregulation of T cell response in the pathogenesis of inflammatory bowel disease. Scand J Immunol 2024; 100:e13412. [PMID: 39394898 DOI: 10.1111/sji.13412] [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: 11/20/2023] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 10/14/2024]
Abstract
Inflammatory bowel disease (IBD), comprised of Crohn's disease (CD) and ulcerative colitis (UC), are gut inflammatory diseases that were earlier prevalent in the Western Hemisphere but now are on the rise in the East, with India standing second highest in the incidence rate in the world. Inflammation in IBD is a cause of dysregulated immune response, wherein helper T (Th) cell subsets and their cytokines play a major role in the pathogenesis of IBD. In addition, gut microbiota, environmental factors such as dietary factors and host genetics influence the outcome and severity of IBD. Dysregulation between effector and regulatory T cells drives gut inflammation, as effector T cells like Th1, Th17 and Th9 subsets Th cell lineages were found to be increased in IBD patients. In this review, we attempted to discuss the role of different Th cell subsets together with other T cells like CD8+ T cells, NKT and γδT cells in the outcome of gut inflammation in IBD. We also highlighted the potential therapeutic candidates for IBD.
Collapse
Affiliation(s)
- Rucha Chandwaskar
- Amity Institute of Microbial Technology (AIMT), Amity University Jaipur, Rajasthan, India
| | - Rajdeep Dalal
- Infection and Immunology Lab, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, India
| | - Saurabh Gupta
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Aishwarya Sharma
- Sri Siddhartha Medical College and Research Center, Tumkur, Karnataka, India
| | - Deepak Parashar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Vivek K Kashyap
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
| | - Jagdip Singh Sohal
- Centre for Vaccines and Diagnostic Research, GLA University, Mathura, Uttar Pradesh, India
| | - Subhash K Tripathi
- Center for Immunity and Immunotherapies and Program for Cell and Gene Therapy, Seattle Children's Research Institute, Seattle, Washington, USA
| |
Collapse
|
6
|
Hijal N, Fouani M, Awada B. Unveiling the fate and potential neuroprotective role of neural stem/progenitor cells in multiple sclerosis. Front Neurol 2024; 15:1438404. [PMID: 39634777 PMCID: PMC11614735 DOI: 10.3389/fneur.2024.1438404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Chronic pathological conditions often induce persistent systemic inflammation, contributing to neuroinflammatory diseases like Multiple Sclerosis (MS). MS is known for its autoimmune-mediated damage to myelin, axonal injury, and neuronal loss which drive disability accumulation and disease progression, often manifesting as cognitive impairments. Understanding the involvement of neural stem cells (NSCs) and neural progenitor cells (NPCs) in the remediation of MS through adult neurogenesis (ANG) and gliogenesis-the generation of new neurons and glial cells, respectively is of great importance. Hence, these phenomena, respectively, termed ANG and gliogenesis, involve significant structural and functional changes in neural networks. Thus, the proper integration of these newly generated cells into existing circuits is not only key to understanding the CNS's development but also its remodeling in adulthood and recovery from diseases such as MS. Understanding how MS influences the fate of NSCs/NPCs and their possible neuroprotective role, provides insights into potential therapeutic interventions to alleviate the impact of MS on cognitive function and disease progression. This review explores MS, its pathogenesis, clinical manifestations, and its association with ANG and gliogenesis. It highlights the impact of altered NSCs and NPCs' fate during MS and delves into the potential benefits of its modifications. It also evaluates treatment regimens that influence the fate of NSCS/NPCs to counteract the pathology subsequently.
Collapse
Affiliation(s)
- Nora Hijal
- Department of Nursing, American University of Beirut Medical Center, Beirut, Lebanon
| | - Malak Fouani
- Department of Neurology, Duke University Medical Center, Durham, NC, United States
| | - Bassel Awada
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
7
|
Huo MH, Adeerjiang Y, Abulitipu A, Khan U, Li XX, Zhang L, Tian Y, Jiang S, Xu CC, Chao XZ, Yang YF, Zhang JX, Du GL. Th17/Treg cell balance in patients with papillary thyroid carcinoma: a new potential biomarker and therapeutic target. Front Oncol 2024; 14:1325575. [PMID: 39534095 PMCID: PMC11554530 DOI: 10.3389/fonc.2024.1325575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Papillary thyroid carcinoma (PTC) is the most common subtype of thyroid carcinoma. The most effective treatment for PTC is surgical resection, and patients who undergo surgery have good survival outcomes, but some patients have distant metastasis or even multiorgan metastases at the time of initial diagnosis. Distant metastasis is associated with poorer prognosis and a higher mortality rate. Helper T lymphocyte 17 (Th17) cells and regulatory T lymphocytes (Tregs) play different roles in PTC, and the Th17/Treg balance is closely related to the progression of PTC. Th17 cells play anticancer roles, whereas Tregs play cancer-promoting roles. A Th17/Treg imbalance promotes tumor progression and accelerates invasive behaviors such as tumor metastasis. Th17/Treg homeostasis can be regulated by the TGF-β/IL-2 and IL-6 cytokine axes. Immune checkpoint inhibitors contribute to Treg/Th17 cell homeostasis. For PTC, monoclonal antibodies against CTLA-4, PD-1 and PD-L1 inhibit the activation of Tregs, reversing the Th17/Treg cell imbalance and providing a new option for the prevention and treatment of PTC. This article reviews the role of Tregs and Th17 cells in PTC and their potential targets, aiming to provide better treatment options for PTC.
Collapse
Affiliation(s)
- Meng-Han Huo
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Gastroenterology and Endocrinology, Tianjin Haihe Hospital, Tianjin, China
| | - Yilinuer Adeerjiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ayiguzhali Abulitipu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Umair Khan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xin-Xi Li
- Department of Endocrine Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Lei Zhang
- Department of Endocrine Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Ye Tian
- Department of Endocrine Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Sheng Jiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Can-Can Xu
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xian-Zhen Chao
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Ye-Fan Yang
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Jin-Xia Zhang
- First Clinical Medical College of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Guo-Li Du
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Endocrinology, Bayingolin Mongolian Autonomous Prefecture People's Hospital, Kuerle, China
| |
Collapse
|
8
|
Donald K, Serapio-Palacios A, Gerbec Z, Bozorgmehr T, Holani R, Cruz AR, Schnupf P, Finlay BB. Secretory IgA in breast milk protects against asthma through modulation of the gut microbiota. Cell Rep 2024; 43:114835. [PMID: 39368092 DOI: 10.1016/j.celrep.2024.114835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 07/22/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024] Open
Abstract
Asthma susceptibility is linked to dysbiosis in early-life gut microbiota, and the antibody secretory immunoglobulin (Ig)A (SIgA) is a key determinant of gut microbiota composition. SIgA is obtained through breast milk during the critical early-life window. We use a mouse model of SIgA deficiency and the house dust mite (HDM) model of asthma to elucidate the role of maternal SIgA in modulating the early-life gut microbiota and asthma protection. Mice that do not receive maternal SIgA display a transient bloom of segmented filamentous bacteria (SFB) in the small intestine during the early post-weaning period. Mice that do not receive maternal SIgA also display elevated T helper type 17 (Th17) cell activation in the intestine, which persists into adulthood and is associated with more severe inflammation in response to the HDM model of asthma. This study demonstrates a mechanism by which breast-milk-derived SIgA influences immune development and asthma susceptibility by modulating the early-life gut microbiota.
Collapse
Affiliation(s)
- Katherine Donald
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Antonio Serapio-Palacios
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Zachary Gerbec
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; British Columbia Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | - Tahereh Bozorgmehr
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Ravi Holani
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Ana Raquel Cruz
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Laboratory of Host-Microbiota Interaction, 75015 Paris, France
| | - Pamela Schnupf
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Laboratory of Host-Microbiota Interaction, 75015 Paris, France
| | - B Brett Finlay
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
9
|
Ruiz García Y, Marrazzo J, Martinón-Torres F, Workowski K, Giordano G, Pizza M, Sohn WY. Urgent Need to Understand and Prevent Gonococcal Infection: From the Laboratory to Real-World Context. J Infect Dis 2024; 230:e758-e767. [PMID: 38819303 PMCID: PMC11481298 DOI: 10.1093/infdis/jiae289] [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: 04/26/2023] [Revised: 09/19/2023] [Accepted: 05/30/2024] [Indexed: 06/01/2024] Open
Abstract
Neisseria gonorrhoeae is widespread globally. Primary prevention is unsuccessful and antimicrobial resistance threatens optimal management. There is no specific vaccine and natural infection studies show that N gonorrhoeae can avoid and suppress immune responses. In addition to extensive variation in expression and specificity of many gonococcal surface antigens, it induces a robust inflammatory response through the Th17 pathway with a large influx of neutrophils and inflammatory cytokines but evades macrophages. The Th1- and Th2-mediated response is suppressed, resulting in low, short-lived antibody titers. Real-world evidence suggests that gonorrhea cases are reduced among recipients of Neisseria meningitidis group B vaccines containing outer membrane vesicles (OMVs). Although the first randomized trial of an OMV-containing MenB vaccine against N gonorrhoeae infection did not show statistically significant vaccine efficacy, ongoing trials might shed further light. Several candidate vaccine antigens for a gonococcal-specific vaccine are being evaluated preclinically but only one has reached clinical trials.
Collapse
Affiliation(s)
| | - Jeanne Marrazzo
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Federico Martinón-Torres
- Translational Pediatrics and Infectious Diseases, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
- Genetics, Vaccines and Infections Research Group, Instituto de Investigación Sanitaria de Santiago, University of Santiago de Compostela, Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | | | | | | | | |
Collapse
|
10
|
Mukherjee S, Ghosh P, Ghosh S, Sengupta A, Sarkar S, Chatterjee R, Saha A, Bawali S, Choudhury A, Daptary AH, Gangopadhyay A, Keswani T, Bhattacharyya A. Administration of rIL-33 Restores Altered mDC/pDC Ratio, MDSC Frequency, and Th-17/Treg Ratio during Experimental Cerebral Malaria. Pathogens 2024; 13:877. [PMID: 39452748 PMCID: PMC11509898 DOI: 10.3390/pathogens13100877] [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: 06/24/2024] [Revised: 09/28/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024] Open
Abstract
The onset of malaria causes the induction of various inflammatory markers in the host's body, which in turn affect the body's homeostasis and create several cerebral complications. Polarization of myeloid-derived suppressor cells (MDSCs) from the classically activated M1 to alternatively activated M2 phenotype increases the secretion of pro-inflammatory molecules. Treatment with recombinant IL-33 (rIL-33) not only alters this MDSC's polarization but also targets the glycolysis pathway of the metabolism in MDSCs, rendering them less immunosuppressive. Along with that, the Helper T-cells subset 17 (Th17)/T regulatory cells (Tregs) ratio is skewed towards Th17, which increases inflammation by producing more IL-17. However, treating with rIL-33 also helps to restore this ratio, which brings back homeostasis. During malaria infection, there is an upregulation of IL-12 production from dendritic cells along with a distorted myeloid dendritic cells (mDC)/plasmacytoid dendritic cells (pDC) ratio towards mDCs promoting inflammation. Administering rIL-33 will also subvert this IL-12 production and increase the population of pDC in the host's immune system during malaria infection, thus restoring mDC/pDC to homeostasis. Therefore, treatment with rIL-33 to reduce the pro-inflammatory signatures and maintenance of immune homeostasis along with the increase in survivability could be a potential therapeutic approach for cerebral malaria.
Collapse
Affiliation(s)
- Saikat Mukherjee
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Pronabesh Ghosh
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Soubhik Ghosh
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Anirban Sengupta
- Centre for Infectious Medicine, Department of Medicine Huddinge, Karolinksa Institutet, 14152 Stockholm, Sweden;
| | - Samrat Sarkar
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Rimbik Chatterjee
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Atreyee Saha
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Sriparna Bawali
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Abhishek Choudhury
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Altamas Hossain Daptary
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Anwesha Gangopadhyay
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| | - Tarun Keswani
- Center for Immunological and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Arindam Bhattacharyya
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India; (S.M.); (P.G.); (S.G.); (S.S.); (R.C.); (A.S.); (S.B.); (A.C.); (A.H.D.); (A.G.)
| |
Collapse
|
11
|
Abreu H, Lallukka M, Raineri D, Leigheb M, Ronga M, Cappellano G, Spriano S, Chiocchetti A. Evaluation of the immune response of peripheral blood mononuclear cells cultured on Ti6Al4V-ELI polished or etched surfaces. Front Bioeng Biotechnol 2024; 12:1458091. [PMID: 39439551 PMCID: PMC11493608 DOI: 10.3389/fbioe.2024.1458091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction While titanium and its alloys exhibit excellent biocompatibility and corrosion resistance, their polished surfaces can hinder fast and effective osseointegration and other biological processes, such as angiogenesis, due to their inert and hydrophobic properties. Despite being commonly used for orthopedic implants, research focuses on developing surface treatments to improve osseointegration, promoting cell adhesion and proliferation, as well as increasing protein adsorption capacity. This study explores a chemical treatment intended for titanium-based implants that enhances tissue integration without compromising the mechanical properties of the Ti6Al4V substrate. However, recognizing that inflammation contributes to nearly half of early implant failures, we assessed the impact of this treatment on T-cell viability, cytokine production, and phenotype. Methods Ti6Al4V with extra low interstitial (ELI) content discs were treated with hydrofluoric acid followed by a controlled oxidation step in hydrogen peroxide that creates a complex surface topography with micro- and nano-texture and modifies the chemistry of the surface oxide layer. The acid etched surface contains an abundance of hydroxyl groups, crucial for promoting bone growth and apatite precipitation, while also enabling further functionalization with biomolecules. Results While cell viability remained high in both groups, untreated discs triggered an increase in Th2 cells and a decrease of the Th17 subset. Furthermore, peripheral blood mononuclear cells exposed to untreated discs displayed a rise in various pro-inflammatory and anti-inflammatory cytokines compared to the control and treated groups. Conversely, the treated discs showed a similar profile to the control, both in terms of immune cell subset frequencies and cytokine secretion. Discussion The dysregulation of the cytokine profile upon contact with untreated Ti6Al4V-ELI discs, namely upregulation of IL-2 could be responsible for the decrease in Th17 frequency, and thus might contribute to implant-associated bacterial infection. Interestingly, the chemical treatment restores the immune response to levels comparable to the control condition, suggesting the treatment's potential to mitigate inflammation by enhancing biocompatibility.
Collapse
Affiliation(s)
- Hugo Abreu
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale, Novara, Italy
| | - Mari Lallukka
- Applied Science and Technology Department, Politecnico di Torino, Torino, Italy
| | - Davide Raineri
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale, Novara, Italy
| | - Massimiliano Leigheb
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Orthopaedics and Traumatology Unit, “Maggiore della Carità” Hospital, Novara, Italy
| | - Mario Ronga
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Orthopaedics and Traumatology Unit, “Maggiore della Carità” Hospital, Novara, Italy
| | - Giuseppe Cappellano
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale, Novara, Italy
| | - Silvia Spriano
- Applied Science and Technology Department, Politecnico di Torino, Torino, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases-IRCAD, Università del Piemonte Orientale, Novara, Italy
- Center for Translational Research on Autoimmune and Allergic Diseases-CAAD, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
12
|
Rodríguez-Fernández K, Zarzoso-Foj J, Saez-Bello M, Mateu-Puchades A, Martorell-Calatayud A, Merino-Sanjuan M, Gras-Colomer E, Climente-Martí M, Mangas-Sanjuan V. Model-Informed Precision Dosing for Personalized Ustekinumab Treatment in Plaque Psoriasis. Pharmaceutics 2024; 16:1295. [PMID: 39458624 PMCID: PMC11510411 DOI: 10.3390/pharmaceutics16101295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/29/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Implementing model-informed precision dosing (MIPD) strategies guided by population pharmacokinetic/pharmacodynamic (PK/PD) models could enhance the management of inflammatory diseases such as psoriasis. However, the extent of individual experimental data gathered during MIPD significantly influences the uncertainty in estimating individual PK/PD parameters, affecting clinical dose selection decisions. METHODS This study proposes a methodology to individualize ustekinumab (UTK) dosing strategies for 23 Spanish patients with moderate to severe chronic plaque psoriasis., considering the uncertainty of individual parameters within a population PK/PD model. RESULTS An indirect response model from previous research was used to describe the PK/PD relationship between UTK serum concentrations and the Psoriasis Area and Severity Index (PASI) score. A maximum inhibition drug effect (Imax) model was selected, and a first-order remission constant rate of psoriatic skin lesion (kout = 0.016 d-1) was estimated. CONCLUSIONS The MIPD approach predicted that 35% and 26% of the patients would need an optimized and intensified dosage regimen, respectively, compared to the regimen typically used in clinical practice. This analysis demonstrated its utility as a tool for selecting personalized UTK dosing regimens in clinical practice in order to optimize the probability of achieving targeted clinical outcomes in patients with psoriasis.
Collapse
Affiliation(s)
- Karine Rodríguez-Fernández
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, 46100 Valencia, Spain; (K.R.-F.); (J.Z.-F.); (M.M.-S.)
- Interuniversity Research Institute for Molecular Recognition and Technological Development, Polytechnic University of Valencia-University of Valencia, 46100 Valencia, Spain
| | - Javier Zarzoso-Foj
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, 46100 Valencia, Spain; (K.R.-F.); (J.Z.-F.); (M.M.-S.)
- Interuniversity Research Institute for Molecular Recognition and Technological Development, Polytechnic University of Valencia-University of Valencia, 46100 Valencia, Spain
| | - Marina Saez-Bello
- Pharmacy Service, Doctor Peset University Hospital, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (M.S.-B.); (M.C.-M.)
| | - Almudena Mateu-Puchades
- Dermatology Service, Doctor Peset University Hospital, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain;
| | | | - Matilde Merino-Sanjuan
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, 46100 Valencia, Spain; (K.R.-F.); (J.Z.-F.); (M.M.-S.)
- Interuniversity Research Institute for Molecular Recognition and Technological Development, Polytechnic University of Valencia-University of Valencia, 46100 Valencia, Spain
| | | | - Monica Climente-Martí
- Pharmacy Service, Doctor Peset University Hospital, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (M.S.-B.); (M.C.-M.)
| | - Victor Mangas-Sanjuan
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, 46100 Valencia, Spain; (K.R.-F.); (J.Z.-F.); (M.M.-S.)
- Interuniversity Research Institute for Molecular Recognition and Technological Development, Polytechnic University of Valencia-University of Valencia, 46100 Valencia, Spain
| |
Collapse
|
13
|
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
|
14
|
Engesser J, Wang H, Kapffer S, Kaffke A, Peters A, Paust HJ, Geissen M, Krebs CF, Panzer U, Asada N. S1PR1 mediates Th17 cell migration from the thymus to the skin in health and disease. Front Immunol 2024; 15:1473130. [PMID: 39380990 PMCID: PMC11459589 DOI: 10.3389/fimmu.2024.1473130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/23/2024] [Indexed: 10/10/2024] Open
Abstract
Th17 cells play crucial roles in host defense and the pathogenesis of autoimmune diseases in the skin. While their differentiation mechanisms have been extensively studied, the origin of skin Th17 cells remains unclear. In this study, we analyzed single-cell RNA-sequencing data and identify the presence of Th17 cells in the human thymus. Thymic Th17 cells were characterized by high expression levels of Sphingosine-1-Phosphate Receptor 1 (S1PR1), a receptor crucial for T cell egress from lymphoid tissues. In mice, Th17 cell-specific knockout of S1pr1 resulted in the accumulation of Th17 cells in the thymus and a corresponding decrease in their numbers in the skin. Th17 cells that accumulated in the thymus exhibited a lower IL-17A production capacity compared to those in the skin, indicating that the local environment in the skin is important for maintaining the Th17 cell phenotype. Additionally, using a murine psoriasis model, we demonstrated that Th17 cell-specific knockout of S1pr1 reduced their migration to the inflamed skin, thereby ameliorating disease progression. Collectively, our data suggest that S1PR1 mediates Th17 cell migration from the thymus to the skin, thereby modulating their functional engagement in both homeostatic and inflammatory conditions.
Collapse
Affiliation(s)
- Jonas Engesser
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Huiying Wang
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Sonja Kapffer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Kaffke
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Anett Peters
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Hans-Joachim Paust
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Markus Geissen
- Clinic and Polyclinic for Vascular Medicine, University Heart and Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian F. Krebs
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Ulf Panzer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Nariaki Asada
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg Eppendorf, Hamburg, Germany
| |
Collapse
|
15
|
Zhang H, Felthaus O, Eigenberger A, Klein S, Prantl L. Treg Cell Therapeutic Strategies for Breast Cancer: Holistic to Local Aspects. Cells 2024; 13:1526. [PMID: 39329710 PMCID: PMC11429654 DOI: 10.3390/cells13181526] [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: 07/30/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Regulatory T cells (Tregs) play a key role in maintaining immune homeostasis and preventing autoimmunity through their immunosuppressive function. There have been numerous reports confirming that high levels of Tregs in the tumor microenvironment (TME) are associated with a poor prognosis, highlighting their role in promoting an immunosuppressive environment. In breast cancer (BC), Tregs interact with cancer cells, ultimately leading to the suppression of immune surveillance and promoting tumor progression. This review discusses the dual role of Tregs in breast cancer, and explores the controversies and therapeutic potential associated with targeting these cells. Researchers are investigating various strategies to deplete or inhibit Tregs, such as immune checkpoint inhibitors, cytokine antagonists, and metabolic inhibition. However, the heterogeneity of Tregs and the variable precision of treatments pose significant challenges. Understanding the functional diversity of Tregs and the latest advances in targeted therapies is critical for the development of effective therapies. This review highlights the latest approaches to Tregs for BC treatment that both attenuate Treg-mediated immunosuppression in tumors and maintain immune tolerance, and advocates precise combination therapy strategies to optimize breast cancer outcomes.
Collapse
Affiliation(s)
- Hanwen Zhang
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauss Allee 11, 93053 Regensburg, Germany (L.P.)
| | | | | | | | | |
Collapse
|
16
|
Murayama M, Chow SK, Lee ML, Young B, Ergul YS, Shinohara I, Susuki Y, Toya M, Gao Q, Goodman SB. The interactions of macrophages, lymphocytes, and mesenchymal stem cells during bone regeneration. Bone Joint Res 2024; 13:462-473. [PMID: 39237112 PMCID: PMC11377107 DOI: 10.1302/2046-3758.139.bjr-2024-0122.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/07/2024] Open
Abstract
Bone regeneration and repair are crucial to ambulation and quality of life. Factors such as poor general health, serious medical comorbidities, chronic inflammation, and ageing can lead to delayed healing and nonunion of fractures, and persistent bone defects. Bioengineering strategies to heal bone often involve grafting of autologous bone marrow aspirate concentrate (BMAC) or mesenchymal stem cells (MSCs) with biocompatible scaffolds. While BMAC shows promise, variability in its efficacy exists due to discrepancies in MSC concentration and robustness, and immune cell composition. Understanding the mechanisms by which macrophages and lymphocytes - the main cellular components in BMAC - interact with MSCs could suggest novel strategies to enhance bone healing. Macrophages are polarized into pro-inflammatory (M1) or anti-inflammatory (M2) phenotypes, and influence cell metabolism and tissue regeneration via the secretion of cytokines and other factors. T cells, especially helper T1 (Th1) and Th17, promote inflammation and osteoclastogenesis, whereas Th2 and regulatory T (Treg) cells have anti-inflammatory pro-reconstructive effects, thereby supporting osteogenesis. Crosstalk among macrophages, T cells, and MSCs affects the bone microenvironment and regulates the local immune response. Manipulating the proportion and interactions of these cells presents an opportunity to alter the local regenerative capacity of bone, which potentially could enhance clinical outcomes.
Collapse
Affiliation(s)
- Masatoshi Murayama
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Simon K Chow
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Max L Lee
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Bill Young
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Yasemin S Ergul
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Issei Shinohara
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Yosuke Susuki
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Masakazu Toya
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Bioengineering, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
17
|
Xu X, Niu M, Lamberty BG, Emanuel K, Ramachandran S, Trease AJ, Tabassum M, Lifson JD, Fox HS. Microglia and macrophages alterations in the CNS during acute SIV infection: A single-cell analysis in rhesus macaques. PLoS Pathog 2024; 20:e1012168. [PMID: 39283947 PMCID: PMC11426456 DOI: 10.1371/journal.ppat.1012168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 09/26/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
Human Immunodeficiency Virus (HIV) is widely acknowledged for its profound impact on the immune system. Although HIV primarily affects peripheral CD4 T cells, its influence on the central nervous system (CNS) cannot be overlooked. Within the brain, microglia and CNS-associated macrophages (CAMs) serve as the primary targets for HIV and the simian immunodeficiency virus (SIV) in nonhuman primates. This infection can lead to neurological effects and establish a viral reservoir. Given the gaps in our understanding of how these cells respond in vivo to acute CNS infection, we conducted single-cell RNA sequencing (scRNA-seq) on myeloid cells from the brains of three rhesus macaques 12 days after SIV infection, along with three uninfected controls. Our analysis revealed six distinct microglial clusters including homeostatic microglia, preactivated microglia, and activated microglia expressing high levels of inflammatory and disease-related molecules. In response to acute SIV infection, the homeostatic and preactivated microglia population decreased, while the activated and disease-related microglia increased. All microglial clusters exhibited upregulation of MHC class I molecules and interferon-related genes, indicating their crucial roles in defending against SIV during the acute phase. All microglia clusters also upregulated genes linked to cellular senescence. Additionally, we identified two distinct CAM populations: CD14lowCD16hi and CD14hiCD16low CAMs. Interestingly, during acute SIV infection, the dominant CAM population changed to one with an inflammatory phenotype. Specific upregulated genes within one microglia and one macrophage cluster were associated with neurodegenerative pathways, suggesting potential links to neurocognitive disorders. This research sheds light on the intricate interactions between viral infection, innate immune responses, and the CNS, providing valuable insights for future investigations.
Collapse
Affiliation(s)
- Xiaoke Xu
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Meng Niu
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Benjamin G Lamberty
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Katy Emanuel
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Shawn Ramachandran
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Andrew J Trease
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Mehnaz Tabassum
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, Maryland, United States of America
| | - Howard S Fox
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
18
|
Alanazi H, Zhang Y, Fatunbi J, Luu T, Kwak-Kim J. The impact of reproductive hormones on T cell immunity; normal and assisted reproductive cycles. J Reprod Immunol 2024; 165:104295. [PMID: 39053203 DOI: 10.1016/j.jri.2024.104295] [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: 04/24/2024] [Revised: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 07/27/2024]
Abstract
During pregnancy, a unique immune milieu is established systemically and locally at the maternal-fetal interface. While preparing for embryonic implantation, endometrial effectors significantly change their proportions and function, which are synchronized with hormonal changes. During assisted reproductive technology cycles, various cytokines, chemokines, and immune factors dynamically change with the altered receptor expressions on the immune effectors. Thus, the hormonal regulation of immune effectors is critical to maintaining the immune milieu. In this review, hormonal effects on T cell subsets are reviewed. Sex hormones affect T cell ontogeny and development, consequently affecting their functions. Like other T cell subsets, CD4+ T helper (Th) cells are modulated by estrogen, where low estrogen concentration promotes Th1-driven cell-mediated immunity in the uterus and in vitro by enhancing IFN-γ production, while a high estrogen level decreases it. The abundance and differentiation of T regulatory (Treg) cells are controlled by estrogen, inducing Treg expansion. Conversely, progesterone maintains immune homeostasis by balancing Th1/Th2 and Th17/Treg immunity, leading to maternal-fetal tolerance. Therefore, the understanding of the hormonal impact on various T cell subsets during the reproductive cycles is critical to improving reproductive outcomes in women with recurrent pregnancy losses, repeated implantation failures, and undergoing assisted reproductive cycles.
Collapse
Affiliation(s)
- Hallah Alanazi
- Reproductive Medicine and Immunology, Obstetrics and Gynaecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, USA; IVF and Reproductive Endocrinology Department, Women's Health Hospital, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Yuan Zhang
- Reproductive Medicine and Immunology, Obstetrics and Gynaecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, USA; Department of Reproductive Medicine, Jiangsu Province Hospital, Guangzhou Road 300, Nanjing, Jiangsu 210029, China
| | - Joy Fatunbi
- Reproductive Medicine and Immunology, Obstetrics and Gynaecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, USA
| | - Than Luu
- Reproductive Medicine and Immunology, Obstetrics and Gynaecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, USA
| | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Obstetrics and Gynaecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, USA.
| |
Collapse
|
19
|
Hung L, Zientara B, Berin MC. Contribution of T cell subsets to different food allergic diseases. Immunol Rev 2024; 326:35-47. [PMID: 39054597 DOI: 10.1111/imr.13368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Food allergies occur due to a lack of tolerance to the proteins found in foods. While IgE- and non-IgE-mediated food allergies have different clinical manifestations, epidemiology, pathophysiology, and management, they share dysregulated T cell responses. Recent studies have shed light on the contributions of different T cell subsets to the development and persistence of different food allergic diseases. This review discusses the role of T cells in both IgE- and non-IgE-mediated food allergies and considers the potential future investigations in this context.
Collapse
Affiliation(s)
- Lisa Hung
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Brianna Zientara
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - M Cecilia Berin
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
20
|
Kim HH, Jeong SH, Park MY, Bhosale PB, Abusaliya A, Heo JD, Kim HW, Seong JK, Kim TY, Park JW, Kim BS, Kim GS. The Skin Histopathology of Pro- and Parabiotics in a Mouse Model of Atopic Dermatitis. Nutrients 2024; 16:2903. [PMID: 39275219 PMCID: PMC11397434 DOI: 10.3390/nu16172903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/16/2024] Open
Abstract
As it has been revealed that the activation of human immune cells through the activity of intestinal microorganisms such as pro- and prebiotics plays a vital role, controlling the proliferation of beneficial bacteria and suppressing harmful bacteria in the intestine has become essential. The importance of probiotics, especially for skin health and the immune system, has led to the emergence of products in various forms, including probiotics, prebiotics, and parabiotics. In particular, atopic dermatitis (AD) produces hypersensitive immunosuppressive substances by promoting the differentiation and activity of immune regulatory T cells. As a result, it has been in the Th1 and Th2 immune balance through a mechanism that suppresses skin inflammation or allergic immune responses caused by bacteria. Furthermore, an immune mechanism has recently emerged that simultaneously controls the expression of IL-17 produced by Th17. Therefore, the anti-atopic effect was investigated by administering doses of anti-atopic candidate substances (Lactobacilus sakei CVL-001, Lactobacilus casei MCL, and Lactobacilus sakei CVL-001 Lactobacilus casei MCL mixed at a ratio of 4:3) in an atopy model using 2,4-dinitrochlorobenzene and observing symptom changes for 2 weeks to confirm the effect of pro-, para-, and mixed biotics on AD. First, the body weight and feed intake of the experimental animals were investigated, and total IgG and IgM were confirmed through blood biochemical tests. Afterward, histopathological staining was performed using H&E staining, Toluidine blue staining, Filaggrin staining, and CD8 antibody staining. In the treatment group, the hyperproliferation of the epidermal layer, the inflammatory cell infiltration of the dermal layer, the expression of CD8, the expression of filaggrin, and the secretion of mast cells were confirmed to be significantly reduced. Lastly, small intestine villi were observed through a scanning microscope, and scoring evaluation was performed through skin damage. Through these results, it was confirmed that AD was reduced when treated with pro-, para-, and mixed biotics containing probiotics and parabiotics.
Collapse
Affiliation(s)
- Hun Hwan Kim
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Se Hyo Jeong
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Min Yeong Park
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Pritam Bhagwan Bhosale
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Abuyaseer Abusaliya
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Jeong Doo Heo
- Biological Resources Research Group, Gyeongnam Department of Environment Toxicology and Chemistry, Korea Institute of Toxicology, Jinju 52834, Republic of Korea
| | - Hyun Wook Kim
- Division of Animal Bioscience & Intergrated Biotechnology, Gyeongsang National University, Jinju 52725, Republic of Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, BK21 PLUS Program for Creative Veterinary Science Research, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Tae Yang Kim
- R&D Group, Kick the Hurdle, Changwon-si 51139, Republic of Korea
| | - Jeong Woo Park
- R&D Group, Kick the Hurdle, Changwon-si 51139, Republic of Korea
| | - Byeong Soo Kim
- R&D Group, Kick the Hurdle, Changwon-si 51139, Republic of Korea
| | - Gon Sup Kim
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
21
|
Saini C, Sapra L, Puri P, Mishra PK, Chawla R, Srivastava RK. Dysregulated Th17/Treg cell axis is correlated with local and systemic immune response in human intermediate uveitis. Immunol Lett 2024; 268:106888. [PMID: 38925441 DOI: 10.1016/j.imlet.2024.106888] [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: 04/20/2024] [Revised: 06/19/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024]
Abstract
Th17/Treg cell balance is essential for immune homeostasis and when disrupted, is associated with the occurrence and development of inflammation in numerous autoimmune diseases. However, its contribution in pathophysiology of uveitis remains unexplored. In this study, we deciphered the role of Th17/Treg cell balance in autoimmune uveitis subjects. Using flow cytometry, we detected the frequencies and absolute count of both Th17 and Treg cells in the aqueous humor and peripheral blood of patients and healthy controls. Our results for the first time reveal a significant increase (p < 0.01 and p < 0.005) in Th17 population alongside a significant decrease (p < 0.001 and p < 0.003) in Treg cell population in both the aqueous humor and PBMCs of uveitis patients. Further we analyzed the expression of Th17-Treg associated genes and cytokines via qPCR and ELISA respectively. These findings align with our flow cytometry results, as evident by a significant (p < 0.002) up-regulation of IL-17 and a concurrent down regulation of IL-10 at transcriptional levels. Moreover, IL-17A cytokine was found to be substantially high (p < 0.001) and IL-10 (p < 0.02) down regulated in serum. Interestingly, we demonstrated a significant correlation of Th17/Treg cells in aqueous humor with those in peripheral blood. Conclusively, our results suggest the pivotal role of Th17/Treg cell axis in the immuno-pathophysiology of human uveitis. Further we propose the therapeutic potential of targeting this novel axis for ameliorating the disease burden associated with uveitis.
Collapse
Affiliation(s)
- Chaman Saini
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Leena Sapra
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | - Prabhav Puri
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Pradyumna K Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, MP 462001, India
| | - Rohan Chawla
- Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Rupesh K Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India.
| |
Collapse
|
22
|
Gómez I, Egui A, Palacios G, Carrilero B, Benítez C, Simón M, Segovia M, Carmelo E, López MC, Thomas MC. The expression of immune response genes in patients with chronic Chagas disease is shifted toward the levels observed in healthy subjects as a result of treatment with Benznidazole. Front Cell Infect Microbiol 2024; 14:1439714. [PMID: 39119291 PMCID: PMC11307780 DOI: 10.3389/fcimb.2024.1439714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction Chagas disease, caused by the Trypanosoma cruzi parasite infection, is a potentially life-threatening neglected tropical disease with a worldwide distribution. During the chronic phase of the disease, there exists a fragile balance between the host immune response and parasite replication that keeps patients in a clinically-silent asymptomatic stage for years or even decades. However, in 40% of patients, the disease progresses to clinical manifestations mainly affecting and compromising the cardiac system. Treatment is recommended in the chronic phase, although there are no early markers of its effectiveness. The aim of this study is to identify differential expression changes in genes involved in the immune response in antigen-restimulated PBMC from chronic patients with Chagas disease due to benznidazole treatment. Methods Thus, high-throughput real-time qPCR analysis has been performed to simultaneously determine global changes in the expression of 106 genes involved in the immune response in asymptomatic (IND) and early cardiac manifestations (CCC I) Chagas disease patients pre- and post-treatment with benznidazole. Results and discussion The results revealed that 7 out of the 106 analyzed genes were differentially expressed (4 up- and 3 downregulated) after treatment in IND patients and 15 out of 106 (3 up- and 12 downregulated) after treatment of early cardiac Chagas disease patients. Particularly in CCC I patients, regulation of the expression level of some of these genes towards a level similar to that of healthy subjects suggests a beneficial effect of treatment and supports recommendation of benznidazole administration to early cardiac Chagas disease patients. The data obtained also demonstrated that both in asymptomatic patients and in early cardiac chronic patients, after treatment with benznidazole there is a negative regulation of the proinflammatory and cytotoxic responses triggered as a consequence of T. cruzi infection and the persistence of the parasite. This downregulation of the immune response likely prevents marked tissue damage and healing in early cardiac patients, suggesting its positive effect in controlling the pathology.
Collapse
Affiliation(s)
- Inmaculada Gómez
- Departamento de Biología Molecular, Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Adriana Egui
- Departamento de Biología Molecular, Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Génesis Palacios
- Departamento de Biología Molecular, Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, La Laguna, Spain
| | - Bartolomé Carrilero
- Unidad Regional de Medicina Tropical, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Celia Benítez
- Departamento de Biología Molecular, Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Marina Simón
- Unidad Regional de Medicina Tropical, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Manuel Segovia
- Unidad Regional de Medicina Tropical, Hospital Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Emma Carmelo
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna, La Laguna, Spain
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Universidad de La Laguna, La Laguna, Spain
| | - Manuel Carlos López
- Departamento de Biología Molecular, Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - M. Carmen Thomas
- Departamento de Biología Molecular, Instituto de Parasitología y Biomedicina López-Neyra, Consejo Superior de Investigaciones Científicas, Granada, Spain
| |
Collapse
|
23
|
Tolomeo M, Cascio A. The Complex Dysregulations of CD4 T Cell Subtypes in HIV Infection. Int J Mol Sci 2024; 25:7512. [PMID: 39062756 PMCID: PMC11276885 DOI: 10.3390/ijms25147512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/04/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Human immunodeficiency virus (HIV) infection remains an important global public health problem. About 40 million people are infected with HIV, and this infection caused about 630,000 deaths in 2022. The hallmark of HIV infection is the depletion of CD4+ T helper lymphocytes (Th cells). There are at least seven different Th subtypes, and not all are the main targets of HIV. Moreover, the effect of the virus in a specific subtype can be completely different from that of the others. Although the most compromised Th subtype in HIV infection is Th17, HIV can induce important dysregulations in other subtypes, such as follicular Th (Tfh) cells and regulatory Th cells (Treg cells or Tregs). Several studies have shown that HIV can induce an increase in the immunosuppressive activity of Tregs without causing a significant reduction in their numbers, at least in the early phase of infection. The increased activity of this Th subtype seems to play an important role in determining the immunodeficiency status of HIV-infected patients, and Tregs may represent a new target for innovative anti-HIV therapies, including the so-called "Kick and Kill" therapeutic method whose goal is the complete elimination of the virus and the healing of HIV infection. In this review, we report the most important findings on the effects of HIV on different CD4+ T cell subtypes, the molecular mechanisms by which the virus impairs the functions of these cells, and the implications for new anti-HIV therapeutic strategies.
Collapse
Affiliation(s)
- Manlio Tolomeo
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy;
- Department of Infectious Diseases, A.O.U.P. Palermo, 90127 Palermo, Italy
| | - Antonio Cascio
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy;
- Department of Infectious Diseases, A.O.U.P. Palermo, 90127 Palermo, Italy
| |
Collapse
|
24
|
Kirkpatrick C, Lu YCW. Deciphering CD4 + T cell-mediated responses against cancer. Mol Carcinog 2024; 63:1209-1220. [PMID: 38725218 PMCID: PMC11166516 DOI: 10.1002/mc.23730] [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: 03/12/2024] [Accepted: 04/05/2024] [Indexed: 05/15/2024]
Abstract
It's been long thought that CD8+ cytotoxic T cells play a major role in T cell-mediated antitumor responses, whereas CD4+ T cells merely provide some assistance to CD8+ T cells as the "helpers." In recent years, numerous studies support the notion that CD4+ T cells play an indispensable role in antitumor responses. Here, we summarize and discuss the current knowledge regarding the roles of CD4+ T cells in antitumor responses and immunotherapy, with a focus on the molecular and cellular mechanisms behind these observations. These new insights on CD4+ T cells may pave the way to further optimize cancer immunotherapy.
Collapse
Affiliation(s)
- Catherine Kirkpatrick
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Yong-Chen William Lu
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
25
|
Elahi R, Nazari M, Mohammadi V, Esmaeilzadeh K, Esmaeilzadeh A. IL-17 in type II diabetes mellitus (T2DM) immunopathogenesis and complications; molecular approaches. Mol Immunol 2024; 171:66-76. [PMID: 38795686 DOI: 10.1016/j.molimm.2024.03.009] [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: 11/25/2023] [Revised: 03/06/2024] [Accepted: 03/19/2024] [Indexed: 05/28/2024]
Abstract
Chronic inflammation has long been considered the characteristic feature of type II diabetes mellitus (T2DM) Immunopathogenesis. Pro-inflammatory cytokines are considered the central drivers of the inflammatory cascade leading to β-cell dysfunction and insulin resistance (IR), two major pathologic events contributing to T2DM. Analyzing the cytokine profile of T2DM patients has also introduced interleukin-17 (IL-17) as an upstream regulator of inflammation, regarding its role in inducing the nuclear factor-kappa B (NF-κB) pathway. In diabetic tissues, IL-17 induces the expression of inflammatory cytokines and chemokines. Hence, IL-17 can deteriorate insulin signaling and β-cell function by activating the JNK pathway and inducing infiltration of neutrophils into pancreatic islets, respectively. Additionally, higher levels of IL-17 expression in patients with diabetic complications compared to non-complicated individuals have also proposed a role for IL-17 in T2DM complications. Here, we highlight the role of IL-17 in the Immunopathogenesis of T2DM and corresponding pathways, recent advances in preclinical and clinical studies targeting IL-17 in T2DM, and corresponding challenges and possible solutions.
Collapse
Affiliation(s)
- Reza Elahi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mahdis Nazari
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Vahid Mohammadi
- School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Kimia Esmaeilzadeh
- Department of Medical Nanotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, Iran; Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
26
|
Wilcox NC, Taheri G, Halievski K, Talbot S, Silva JR, Ghasemlou N. Interactions between skin-resident dendritic and Langerhans cells and pain-sensing neurons. J Allergy Clin Immunol 2024; 154:11-19. [PMID: 38492673 DOI: 10.1016/j.jaci.2024.03.006] [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: 12/22/2023] [Revised: 02/13/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
Various immune cells in the skin contribute to its function as a first line of defense against infection and disease, and the skin's dense innervation by pain-sensing sensory neurons protects the host against injury or damage signals. Dendritic cells (DCs) are a heterogeneous population of cells that link the innate immune response to the adaptive response by capturing, processing, and presenting antigens to promote T-cell differentiation and activation. DCs are abundant across peripheral tissues, including the skin, where they are found in the dermis and epidermis. Langerhans cells (LCs) are a DC subset located only in the epidermis; both populations of cells can migrate to lymph nodes to contribute to broad immune responses. Dermal DCs and LCs are found in close apposition with sensory nerve fibers in the skin and express neurotransmitter receptors, allowing them to communicate directly with the peripheral nervous system. Thus, neuroimmune signaling between DCs and/or LCs and sensory neurons can modulate physiologic and pathophysiologic pathways, including immune cell regulation, host defense, allergic response, homeostasis, and wound repair. Here, we summarize the latest discoveries on DC- and LC-neuron interaction with neurons while providing an overview of gaps and areas not previously explored. Understanding the interactions between these 2 defence systems may provide key insight into developing therapeutic targets for treating diseases such as psoriasis, neuropathic pain, and lupus.
Collapse
Affiliation(s)
- Natalie C Wilcox
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Golnar Taheri
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Katherine Halievski
- Department of Anesthesiology and Perioperative Medicine, Queen's University, Kingston, Ontario, Canada
| | - Sebastien Talbot
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jaqueline R Silva
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada; Department of Anesthesiology and Perioperative Medicine, Queen's University, Kingston, Ontario, Canada; Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada
| | - Nader Ghasemlou
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada; Department of Anesthesiology and Perioperative Medicine, Queen's University, Kingston, Ontario, Canada; Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
27
|
Plaza-Florido A, Liem RI, Haddad F, Radom-Aizik S. Whole-blood transcriptome analysis reveals distinct gene expression signatures in paediatric patients with sickle cell anaemia before and after exercise. Br J Haematol 2024; 205:320-328. [PMID: 38768976 PMCID: PMC11245363 DOI: 10.1111/bjh.19533] [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: 01/03/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
Sickle cell anaemia (SCA) patients display elevated levels of circulating pro-inflammatory cytokines and endothelial activation markers compared to healthy peers. The impact of exercise on the pro-inflammatory state in SCA remains unclear. This study aimed to characterize the whole-blood transcriptome profile in response to an acute bout of exercise in paediatric SCA patients. Twenty-three SCA participants (13 ± 3 years, 52% girls) and 17 healthy controls (14 ± 3 years, 29% girls) performed eight 2-min bouts of cycle ergometry interspersed with 1-min rest intervals. Whole-blood transcriptome profile (RNA-seq) was performed before and after exercise. At baseline, gene pathways associated with gas transport in erythrocytes were up-regulated in SCA patients compared to controls. Following exercise, gene pathways associated with innate immunity were altered in both groups. Interaction analyses revealed 160 annotated genes (101 up- and 59 down-regulated) that differentially altered by exercise in SCA patients. Moreover, genes that exhibited a blunted response to exercise in SCA patients were enriched in the IL-17 signalling pathway, suggesting an impaired innate immune response to exercise. This data will contribute to the development of evidence-based exercise prescription guidelines for this patient population.
Collapse
Affiliation(s)
- Abel Plaza-Florido
- Department of Pediatrics, School of Medicine, Pediatric Exercise and Genomics Research Center, University of California Irvine, Irvine, California, USA
| | - Robert I Liem
- Division of Hematology, Oncology & Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Fadia Haddad
- Department of Pediatrics, School of Medicine, Pediatric Exercise and Genomics Research Center, University of California Irvine, Irvine, California, USA
| | - Shlomit Radom-Aizik
- Department of Pediatrics, School of Medicine, Pediatric Exercise and Genomics Research Center, University of California Irvine, Irvine, California, USA
| |
Collapse
|
28
|
Lim JJ, Goedken M, Jin Y, Gu H, Cui JY. Single-cell transcriptomics unveiled that early life BDE-99 exposure reprogrammed the gut-liver axis to promote a proinflammatory metabolic signature in male mice at late adulthood. Toxicol Sci 2024; 200:114-136. [PMID: 38648751 PMCID: PMC11199921 DOI: 10.1093/toxsci/kfae047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are legacy flame retardants that bioaccumulate in the environment. The gut microbiome is an important regulator of liver functions including xenobiotic biotransformation and immune regulation. We recently showed that neonatal exposure to polybrominated diphenyl ether-99 (BDE-99), a human breast milk-enriched PBDE congener, up-regulated proinflammation-related and down-regulated drug metabolism-related genes predominantly in males in young adulthood. However, the persistence of this dysregulation into late adulthood, differential impact among hepatic cell types, and the involvement of the gut microbiome from neonatal BDE-99 exposure remain unknown. To address these knowledge gaps, male C57BL/6 mouse pups were orally exposed to corn oil (10 ml/kg) or BDE-99 (57 mg/kg) once daily from postnatal days 2-4. At 15 months of age, neonatal BDE-99 exposure down-regulated xenobiotic and lipid-metabolizing enzymes and up-regulated genes involved in microbial influx in hepatocytes. Neonatal BDE-99 exposure also increased the hepatic proportion of neutrophils and led to a predicted increase of macrophage migration inhibitory factor signaling. This was associated with decreased intestinal tight junction protein (Tjp) transcripts, altered gut environment, and dysregulation of inflammation-related metabolites. ScRNA-seq using germ-free (GF) mice demonstrated the necessity of a normal gut microbiome in maintaining hepatic immune tolerance. Microbiota transplant to GF mice using large intestinal microbiome from adults neonatally exposed to BDE-99 down-regulated Tjp transcripts and up-regulated several cytokines in large intestine. In conclusion, neonatal BDE-99 exposure reprogrammed cell type-specific gene expression and cell-cell communication in liver towards proinflammation, and this may be partly due to the dysregulated gut environment.
Collapse
Affiliation(s)
- Joe Jongpyo Lim
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105, USA
- Environmental Health and Microbiome Research Center (EHMBRACE), Seattle, Washington 98105, USA
| | - Michael Goedken
- Rutgers Research Pathology Services, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Yan Jin
- Center for Translational Science, Florida International University, Port St Lucie, Florida 34987, USA
| | - Haiwei Gu
- Center for Translational Science, Florida International University, Port St Lucie, Florida 34987, USA
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington 98105, USA
- Environmental Health and Microbiome Research Center (EHMBRACE), Seattle, Washington 98105, USA
| |
Collapse
|
29
|
Tang J, Zhao S, Shi H, Li X, Ran L, Cao J, He Y. Effects on peripheral and central nervous system of key inflammatory intercellular signalling peptides and proteins in psoriasis. Exp Dermatol 2024; 33:e15104. [PMID: 38794817 DOI: 10.1111/exd.15104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/25/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024]
Abstract
Psoriasis is a chronic systemic inflammatory cutaneous disease. Where the immune system plays an important role in its pathogenesis, with key inflammatory intercellular signalling peptides and proteins including IL-17 and IL-23. The psychoneurological system also figures prominently in development of psoriasis. There is a high prevalence of comorbidity between psoriasis and mental health disorders such as depression, anxiety and mania. Patients with psoriasis often suffer from pathological pain in the lesions, and their neurological accidents could improve the lesions in innervated areas. The immune system and the psychoneurological system interact closely in the pathogenesis of psoriasis. Patients with psoriasis exhibit abnormal levels of neuropeptides both in circulating and localized lesion, acting as immunomodulators involved in the inflammatory response. Moreover, receptors for inflammatory factors are expressed in both peripheral and central nervous systems (CNSs), suggesting that nervous system can receive and be influenced by signals from immune system. Key inflammatory intercellular signalling peptides and proteins in psoriasis, such as IL-17 and IL-23, can be involved in sensory signalling and may affect synaptic plasticity and the blood-brain barrier of CNS through the circulation. This review provides an overview of the multiple effects on the peripheral and CNS under conditions of systemic inflammation in psoriasis, providing a framework and inspiration for in-depth studies of neuroimmunomodulation in psoriasis.
Collapse
Affiliation(s)
- Jue Tang
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Siqi Zhao
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Huijuan Shi
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xuan Li
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Liwei Ran
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Jiali Cao
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yanling He
- Department of Dermatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center for Skin and Immune Diseases, Branch in Beijing Chaoyang Hospital, Beijing, China
| |
Collapse
|
30
|
Khan Z, Mehan S, Gupta GD, Narula AS. Immune System Dysregulation in the Progression of Multiple Sclerosis: Molecular Insights and Therapeutic Implications. Neuroscience 2024; 548:9-26. [PMID: 38692349 DOI: 10.1016/j.neuroscience.2024.04.004] [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: 11/22/2023] [Revised: 03/28/2024] [Accepted: 04/09/2024] [Indexed: 05/03/2024]
Abstract
Multiple sclerosis (MS), a prevalent neurological disorder, predominantly affects young adults and is characterized by chronic autoimmune activity. The study explores the immune system dysregulation in MS, highlighting the crucial roles of immune and non-neuronal cells in the disease's progression. This review examines the dual role of cytokines, with some like IL-6, TNF-α, and interferon-gamma (IFN-γ) promoting inflammation and CNS tissue injury, and others such as IL-4, IL-10, IL-37, and TGF-β fostering remyelination and protecting against MS. Elevated chemokine levels in the cerebrospinal fluid (CSF), including CCL2, CCL5, CXCL10, CXCL13, and fractalkine, are analyzed for their role in facilitating immune cell migration across the blood-brain barrier (BBB), worsening inflammation and neurodegeneration. The study also delves into the impact of auto-antibodies targeting myelin components like MOG and AQP4, which activate complement cascades leading to further myelin destruction. The article discusses how compromised BBB integrity allows immune cells and inflammatory mediators to infiltrate the CNS, intensifying MS symptoms. It also examines the involvement of astrocytes, microglia, and oligodendrocytes in the disease's progression. Additionally, the effectiveness of immunomodulatory drugs such as IFN-β and CD20-targeting monoclonal antibodies (e.g., rituximab) in modulating immune responses is reviewed, highlighting their potential to reduce relapse rates and delaying MS progression. These insights emphasize the importance of immune system dysfunction in MS development and progression, guiding the development of new therapeutic strategies. The study underscores recent advancements in understanding MS's molecular pathways, opening avenues for more targeted and effective treatments.
Collapse
Affiliation(s)
- Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India), Moga 142001, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India), Moga 142001, Punjab, India.
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India), Moga, Punjab, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
31
|
Abimannan T, Parthibane V, Le SH, Vijaykrishna N, Fox SD, Karim B, Kunduri G, Blankenberg D, Andresson T, Bamba T, Acharya U, Acharya JK. Sphingolipid biosynthesis is essential for metabolic rewiring during T H17 cell differentiation. SCIENCE ADVANCES 2024; 10:eadk1045. [PMID: 38657065 PMCID: PMC11042737 DOI: 10.1126/sciadv.adk1045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024]
Abstract
T helper 17 (TH17) cells are implicated in autoimmune diseases, and several metabolic processes are shown to be important for their development and function. In this study, we report an essential role for sphingolipids synthesized through the de novo pathway in TH17 cell development. Deficiency of SPTLC1, a major subunit of serine palmitoyl transferase enzyme complex that catalyzes the first and rate-limiting step of de novo sphingolipid synthesis, impaired glycolysis in differentiating TH17 cells by increasing intracellular reactive oxygen species (ROS) through enhancement of nicotinamide adenine dinucleotide phosphate oxidase 2 activity. Increased ROS leads to impaired activation of mammalian target of rapamycin C1 and reduced expression of hypoxia-inducible factor 1-alpha and c-Myc-induced glycolytic genes. SPTLCI deficiency protected mice from developing experimental autoimmune encephalomyelitis and experimental T cell transfer colitis. Our results thus show a critical role for de novo sphingolipid biosynthetic pathway in shaping adaptive immune responses with implications in autoimmune diseases.
Collapse
Affiliation(s)
| | - Velayoudame Parthibane
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Si-Hung Le
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Nagampalli Vijaykrishna
- Genomic Medicine Institute and Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Stephen D. Fox
- Mass Spectrometry Group, National Cancer Institute, Frederick, MD, USA
| | - Baktiar Karim
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Govind Kunduri
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Daniel Blankenberg
- Genomic Medicine Institute and Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Takeshi Bamba
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Usha Acharya
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Jairaj K. Acharya
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, USA
| |
Collapse
|
32
|
Wang H, He Y, Dang D, Zhao Y, Zhao J, Lu W. Gut Microbiota-Derived Tryptophan Metabolites Alleviate Allergic Asthma Inflammation in Ovalbumin-Induced Mice. Foods 2024; 13:1336. [PMID: 38731707 PMCID: PMC11082989 DOI: 10.3390/foods13091336] [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: 04/02/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Asthma is a prevalent respiratory disease. The present study is designed to determine whether gut microbiota-derived tryptophan metabolites alleviate allergic asthma inflammation in ovalbumin (OVA)-induced mice and explore the effect and potential mechanism therein. Asthma model mice were constructed by OVA treatment, and kynurenine (KYN), indole-3-lactic acid (ILA), in-dole-3-carbaldehyde (I3C), and indole acetic acid (IAA) were administered by intraperitoneal injection. The percent survival, weight and asthma symptom score of mice were recorded. The total immunoglobulin E and OVA-specific (s)IgE in the serum and the inflammatory cytokines in the bronchoalveolar lavage fluid (BALF) were detected by the corresponding ELISA kits. The composition of the gut microbiota and tryptophan-targeted metabolism in mouse feces were analyzed using 16S rRNA gene sequencing and targeted metabolomics, respectively. The four tryptophan metabolites improved the percent survival, weight and asthma symptoms of mice, and reduced the inflammatory cells in lung tissues, especially I3C. I3C and IAA significantly (p < 0.05) downregulated the levels of OVA-IgE and inflammatory cytokines. KYN was observed to help restore gut microbiota diversity. Additionally, I3C, KYN, and ILA increased the relative abundance of Anaeroplasma, Akkermansia, and Ruminococcus_1, respectively, which were connected with tryptophan metabolic pathways. IAA also enhanced capability of tryptophan metabolism by the gut microbiota, restoring tryptophan metabolism and increasing production of other tryptophan metabolites. These findings suggest that tryptophan metabolites may modulate asthma through the gut microbiota, offering potential benefits for clinical asthma management.
Collapse
Affiliation(s)
- Hongchao Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (H.W.); (Y.H.); (D.D.); (Y.Z.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yuan He
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (H.W.); (Y.H.); (D.D.); (Y.Z.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Danting Dang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (H.W.); (Y.H.); (D.D.); (Y.Z.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yurong Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (H.W.); (Y.H.); (D.D.); (Y.Z.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (H.W.); (Y.H.); (D.D.); (Y.Z.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (H.W.); (Y.H.); (D.D.); (Y.Z.); (J.Z.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
33
|
Zhou H, Qi Y, Xu Y, Qi X, Qi H. Reverse causation between multiple sclerosis and psoriasis: a genetic correlation and Mendelian randomization study. Sci Rep 2024; 14:8845. [PMID: 38632254 PMCID: PMC11024188 DOI: 10.1038/s41598-024-58182-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 03/26/2024] [Indexed: 04/19/2024] Open
Abstract
Observational studies have found a potential bidirectional positive association between multiple sclerosis and psoriasis, but these studies are susceptible to confounding factors. We examined the directionality of causation using Mendelian randomization and estimated the genetic correlation using the linkage disequilibrium score. We performed Mendelian randomization analysis using large-scale genome-wide association studies datasets from the International Multiple Sclerosis Genetics Consortium (IMSGC, 115,803 individuals of European ancestry) and FinnGen (252,323 individuals of European ancestry). We selected several Mendelian randomization methods including causal analysis using summary effect (CAUSE), inverse variance-weighted (IVW), and pleiotropy-robust methods. According to CAUSE and IVW the genetic liability to MS reduces the risk of psoriasis (CAUSE odds ratio [OR] 0.93, p = 0.045; IVW OR 0.93, p = 2.51 × 10-20), and vice versa (CAUSE OR 0.72, p = 0.001; IVW OR 0.71, p = 4.80 × 10-26). Pleiotropy-robust methods show the same results, with all p-values < 0.05. The linkage disequilibrium score showed no genetic correlation between psoriasis and MS (rg = - 0.071, p = 0.2852). In summary, there is genetic evidence that MS reduces the risk of psoriasis, and vice versa.
Collapse
Affiliation(s)
- Hao Zhou
- Peking University Shenzhen Hospital Clinical College, Anhui Medical University, Shenzhen, 518036, China
- The Fifth Clinical Medical College, Anhui Medical University, Hefei, 230000, China
- Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Yajie Qi
- Peking University Shenzhen Hospital Clinical College, Anhui Medical University, Shenzhen, 518036, China
- The Fifth Clinical Medical College, Anhui Medical University, Hefei, 230000, China
- Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Yingxin Xu
- Peking University Shenzhen Hospital Clinical College, Anhui Medical University, Shenzhen, 518036, China
- The Fifth Clinical Medical College, Anhui Medical University, Hefei, 230000, China
- Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Xiaoyi Qi
- Medical College, Shantou University, Shantou, 515000, China
| | - Hui Qi
- Peking University Shenzhen Hospital Clinical College, Anhui Medical University, Shenzhen, 518036, China.
- The Fifth Clinical Medical College, Anhui Medical University, Hefei, 230000, China.
- Peking University Shenzhen Hospital, Shenzhen, 518036, China.
| |
Collapse
|
34
|
Koudstaal T, van den Bosch T, Bergen I, Lila K, Bresser P, Bogaard HJ, Boomars K, Hendriks R, von der Thüsen J. Predominance of M2 macrophages in organized thrombi in chronic thromboembolic pulmonary hypertension patients. Eur J Immunol 2024:e2350670. [PMID: 38593342 DOI: 10.1002/eji.202350670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 04/11/2024]
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a debilitating disease characterized by thrombotic occlusion of pulmonary arteries and vasculopathy, leading to increased pulmonary vascular resistance and progressive right-sided heart failure. Thrombotic lesions in CTEPH contain CD68+ macrophages, and increasing evidence supports their role in disease pathogenesis. Macrophages are classically divided into pro-inflammatory M1 macrophages and anti-inflammatory M2 macrophages, which are involved in wound healing and tissue repair. Currently, the phenotype of macrophages and their localization within thrombotic lesions of CTEPH are largely unknown. In our study, we subclassified thrombotic lesions of CTEPH patients into developing fresh thrombi (FT) and organized thrombi (OT), based on the degree of fibrosis and remodeling. We used multiplex immunofluorescence histology to identify immune cell infiltrates in thrombotic lesions of CPTEH patients. Utilizing software-assisted cell detection and quantification, increased proportions of macrophages were observed in immune cell infiltrates of OT lesions, compared with FT. Strikingly, the proportions with a CD206+INOS- M2 phenotype were significantly higher in OT than in FT, which mainly contained unpolarized macrophages. Taken together, we observed a shift from unpolarized macrophages in FT toward an expanded population of M2 macrophages in OT, indicating a dynamic role of macrophages during CTEPH pathogenesis.
Collapse
Affiliation(s)
- Thomas Koudstaal
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Thierry van den Bosch
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Ingrid Bergen
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Karishma Lila
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Paul Bresser
- Department of Pulmonary Medicine, Onze Lieve Vrouwe Gasthuis, Amsterdam, the Netherlands
| | - Harm Jan Bogaard
- Department of Pulmonary Medicine, VU Medical Centre, Amsterdam, the Netherlands
| | - Karin Boomars
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Rudi Hendriks
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Jan von der Thüsen
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
35
|
Xu X, Niu M, Lamberty BG, Emanuel K, Trease AJ, Tabassum M, Lifson JD, Fox HS. Microglia and macrophages alterations in the CNS during acute SIV infection: a single-cell analysis in rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588047. [PMID: 38617282 PMCID: PMC11014596 DOI: 10.1101/2024.04.04.588047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Human Immunodeficiency Virus (HIV) is widely acknowledged for its profound impact on the immune system. Although HIV primarily affects peripheral CD4 T cells, its influence on the central nervous system (CNS) cannot be overlooked. Within the brain, microglia and CNS-associated macrophages (CAMs) serve as the primary targets for HIV, as well as for the simian immunodeficiency virus (SIV) in nonhuman primates. This infection can lead to neurological effects and the establishment of a viral reservoir. Given the gaps in our understanding of how these cells respond in vivo to acute CNS infection, we conducted single-cell RNA sequencing (scRNA-seq) on myeloid cells from the brains of three rhesus macaques 12-days after SIV infection, along with three uninfected controls. Our analysis revealed six distinct microglial clusters including homeostatic microglia, preactivated microglia, and activated microglia expressing high levels of inflammatory and disease-related molecules. In response to acute SIV infection, the population of homeostatic and preactivated microglia decreased, while the activated and disease-related microglia increased. All microglial clusters exhibited upregulation of MHC class I molecules and interferon-related genes, indicating their crucial roles in defending against SIV during the acute phase. All microglia clusters also upregulated genes linked to cellular senescence. Additionally, we identified two distinct CAM populations: CD14lowCD16hi and CD14hiCD16low CAMs. Interestingly, during acute SIV infection, the dominant CAM population changed to one with an inflammatory phenotype. Notably, specific upregulated genes within one microglia and one macrophage cluster were associated with neurodegenerative pathways, suggesting potential links to neurocognitive disorders. This research sheds light on the intricate interactions between viral infection, innate immune responses, and the CNS, providing valuable insights for future investigations.
Collapse
Affiliation(s)
- Xiaoke Xu
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Meng Niu
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Benjamin G. Lamberty
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Katy Emanuel
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Andrew J. Trease
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Mehnaz Tabassum
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, Maryland, USA
| | - Howard S. Fox
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
36
|
Bahadorian D, Faraj TA, Kheder RK, Najmaldin SK, Haghmorad D, Mollazadeh S, Esmaeili SA. A glance on the role of IL-35 in systemic lupus erythematosus (SLE). Cytokine 2024; 176:156501. [PMID: 38290255 DOI: 10.1016/j.cyto.2024.156501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/08/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024]
Abstract
It is well known that systemic lupus erythematosus (SLE) is an auto-inflammatory disease that is characterized by chronic and widespread inflammation. The exact pathogenesis of SLE is still a matter of debate. However, it has been suggested that the binding of autoantibodies to autoantigens forms immune complexes (ICs), activators of the immune response, in SLE patients. Ultimately, all of these responses lead to an imbalance between anti-inflammatory and pro-inflammatory cytokines, resulting in cumulative inflammation. IL-35, the newest member of the IL-12 family, is an immunosuppressive and anti-inflammatory cytokine secreted mainly by regulatory cells. Structurally, IL-35 is a heterodimeric cytokine, composed of Epstein-Barr virus-induced gene 3 (EBI3) and p35. IL-35 appears to hold therapeutic and diagnostic potential in cancer and autoimmune diseases. In this review, we summarized the most recent associations between IL and 35 and SLE. Unfortunately, the comparative review of IL-35 in SLE indicates many differences and contradictions, which make it difficult to generalize the use of IL-35 in the treatment of SLE. With the available information, it is not possible to talk about targeting this cytokine for the lupus treatment. So, further studies would be needed to establish the clear and exact levels of this cytokine and its related receptors in people with lupus to provide IL-35 as a preferential therapeutic or diagnostic candidate in SLE management.
Collapse
Affiliation(s)
- Davood Bahadorian
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tola Abdulsattar Faraj
- Department of Basic Sciences, College of Medicine, Hawler Medical University, Erbil, Iraq; Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Ramiar Kamal Kheder
- Medical Laboratory Science Department, College of Science, University of Raparin, Rania, Sulaymaniyah, Iraq; Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Soran K Najmaldin
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Dariush Haghmorad
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran; Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
37
|
Nilsen M, Nygaard UC, Brodin P, Carlsen KCL, Fredheim C, Haugen G, Hedlin G, Jonassen CM, Jonsmoen ULA, Lakshmikanth T, Nordlund B, Olin A, Rehbinder EM, Skjerven HO, Snipen L, Staff AC, Söderhäll C, Vettukattil R, Rudi K. Gut bacteria at 6 months of age are associated with immune cell status in 1-year-old children. Scand J Immunol 2024; 99:e13346. [PMID: 39007947 DOI: 10.1111/sji.13346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 07/16/2024]
Abstract
Age-related gut bacterial changes during infancy have been widely studied, but it remains still unknown how these changes are associated with immune cell composition. This study's aim was to explore if the temporal development of gut bacteria during infancy prospectively affects immune cell composition. Faecal bacteria and short-chain fatty acids were analysed from 67 PreventADALL study participants at four timepoints (birth to 12 months) using reduced metagenome sequencing and gas chromatography. Immune cell frequencies were assessed using mass cytometry in whole blood samples at 12 months. The infants clustered into four groups based on immune cell composition: clusters 1 and 2 showed a high relative abundance of naïve cells, cluster 3 exhibited increased abundance of classical- and non-classical monocytes and clusters 3 and 4 had elevated neutrophil levels. At all age groups, we did observe significant associations between the gut microbiota and immune cell clusters; however, these were generally from low abundant species. Only at 6 months of age we observed significant associations between abundant (>8%) species and immune cell clusters. Bifidobacterium adolescentis and Porphyromonadaceae are associated with cluster 1, while Bacteroides fragilis and Bifidobacterium longum are associated with clusters 3 and 4 respectively. These species have been linked to T-cell polarization and maturation. No significant correlations were found between short-chain fatty acids and immune cell composition. Our findings suggest that abundant gut bacteria at 6 months may influence immune cell frequencies at 12 months, highlighting the potential role of gut microbiota in shaping later immune cell composition.
Collapse
Affiliation(s)
- Morten Nilsen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Unni Cecilie Nygaard
- Section for Immunology, Department of Method Development and Analytics, Norwegian Institute of Public Health, Oslo, Norway
| | - Petter Brodin
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Pediatric Rheumatology, Karolinska University Hospital, Solna, Sweden
| | - Karin Cecilie Lødrup Carlsen
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Cecilie Fredheim
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Guttorm Haugen
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
| | - Gunilla Hedlin
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Christine Monceyron Jonassen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
- Genetic Unit, Centre for Laboratory Medicine, Østfold Hospital Trust, Kalnes, Norway
| | | | | | - Björn Nordlund
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Axel Olin
- Human Evolutionary Genetics, Institut Pasteur, Paris, France
| | - Eva Maria Rehbinder
- Department of Dermatology and Vaenorology, Oslo University Hospital, Oslo, Norway
| | - Håvard O Skjerven
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lars Snipen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Anne Cathrine Staff
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
| | - Cilla Söderhäll
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
- Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Riyas Vettukattil
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Knut Rudi
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
38
|
Montero-Herradón S, García-Ceca J, Villarejo-Torres M, Zapata AG. Peripheral T-cell responses of EphB2- and EphB3-deficient mice in a model of collagen-induced arthritis. Cell Mol Life Sci 2024; 81:159. [PMID: 38558087 PMCID: PMC10984909 DOI: 10.1007/s00018-024-05197-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 12/20/2023] [Accepted: 03/02/2024] [Indexed: 04/04/2024]
Abstract
Both EphB2- and EphB3-deficient mice exhibit profound histological alterations in the thymic epithelial network but few changes in T-cell differentiation, suggesting that this organization would be sufficient to produce functional T lymphocytes. Also, other antigen-presenting cells involved in immunological education could substitute the thymic epithelium. Accordingly, we found an increased frequency of plasmacytoid dendritic cells but not of conventional dendritic cells, medullary fibroblasts or intrathymic B lymphocytes. In addition, there are no lymphoid infiltrates in the organs of mutant mice nor do they contain circulating autoantibodies. Furthermore, attempts to induce arthritic lesions after chicken type II collagen administration fail totally in EphB2-deficient mice whereas all WT and half of the immunized EphB3-/- mice develop a typical collagen-induced arthritis. Our results point out that Th17 cells, IL4-producing Th2 cells and regulatory T cells are key for the induction of disease, but mutant mice appear to have deficits in T cell activation or cell migration properties. EphB2-/- T cells show reduced in vitro proliferative responses to anti-CD3/anti-CD28 antibodies, produce low levels of anti-type II collagen antibodies, and exhibit low proportions of T follicular helper cells. On the contrary, EphB3-/- lymph node cells respond accurately to the different immune stimuli although in lower levels than WT cells but show a significantly reduced migration in in vitro transwell assays, suggesting that no sufficient type II collagen-dependent activated lymphoid cells reached the joints, resulting in reduced arthritic lesions.
Collapse
Affiliation(s)
- Sara Montero-Herradón
- Department of Cell Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain
- Health Research Institute, Hospital 12 de Octubre (imas12), 28041, Madrid, Spain
| | - Javier García-Ceca
- Department of Cell Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain
- Health Research Institute, Hospital 12 de Octubre (imas12), 28041, Madrid, Spain
| | - Marta Villarejo-Torres
- Department of Cell Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain
| | - Agustín G Zapata
- Department of Cell Biology, Faculty of Biological Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
- Health Research Institute, Hospital 12 de Octubre (imas12), 28041, Madrid, Spain.
| |
Collapse
|
39
|
Yue M, Tao S, Gaietto K, Chen W. Omics approaches in asthma research: Challenges and opportunities. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:1-9. [PMID: 39170962 PMCID: PMC11332849 DOI: 10.1016/j.pccm.2024.02.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Indexed: 08/23/2024]
Abstract
Asthma, a chronic respiratory disease with a global prevalence of approximately 300 million individuals, presents a significant societal and economic burden. This multifaceted syndrome exhibits diverse clinical phenotypes and pathogenic endotypes influenced by various factors. The advent of omics technologies has revolutionized asthma research by delving into the molecular foundation of the disease to unravel its underlying mechanisms. Omics technologies are employed to systematically screen for potential biomarkers, encompassing genes, transcripts, methylation sites, proteins, and even the microbiome components. This review provides an insightful overview of omics applications in asthma research, with a special emphasis on genetics, transcriptomics, epigenomics, and the microbiome. We explore the cutting-edge methods, discoveries, challenges, and potential future directions in the realm of asthma omics research. By integrating multi-omics and non-omics data through advanced statistical techniques, we aspire to advance precision medicine in asthma, guiding diagnosis, risk assessment, and personalized treatment strategies for this heterogeneous condition.
Collapse
Affiliation(s)
- Molin Yue
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Shiyue Tao
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Kristina Gaietto
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Wei Chen
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15224, USA
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA
| |
Collapse
|
40
|
Mancuso RA, Ross KM, Accortt E, Coussons-Read M, Okun ML, Irwin J, Carroll J, Hobel CJ, Schetter CD. Prenatal mood and anxiety disorders and associated cytokine changes. J Affect Disord 2024; 347:635-644. [PMID: 38070749 PMCID: PMC11375962 DOI: 10.1016/j.jad.2023.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 11/11/2023] [Accepted: 12/02/2023] [Indexed: 12/25/2023]
Abstract
BACKGROUND We examined whether women with prenatal mood and anxiety disorders would exhibit differential pro- and anti-inflammatory marker trajectories during the prenatal and postpartum periods compared to women without these disorders. METHODS Approximately 179 pregnant women participated in a longitudinal study conducted in two urban areas. Blood samples for inflammatory markers were collected at six study visits. The Structured Clinical Interview for the DSM-IV (SCID) was administered to participants scoring above cutoffs on anxiety and depression. Pregnant women with SCID Axis I diagnoses of mood and/or anxiety disorders were compared to other participants on inflammatory markers. Multilevel modeling tested associations between SCID diagnoses and within-person interleukin (IL)6 and IL10 trajectories. RESULTS Prenatal SCID diagnoses were associated with linear, quadratic and cubic change in IL6 from prenatal to postpartum timepoints. Women with a prenatal SCID diagnosis had steeper decreases and increases in IL6 during prenatal and postpartum periods. SCID diagnoses were associated with lower IL10 in mid-pregnancy to postpartum (b = -0.078, SE = 0.019; p = .015). LIMITATIONS Future studies would benefit from a larger sample size and a larger number of participants with SCID diagnoses. Future research should also examine whether different prenatal Axis 1 diagnoses are associated with different patterns of immune response in pregnancy. CONCLUSIONS Pregnant women with prenatal mood and anxiety disorders had greater fluctuations in IL6 across prenatal and postpartum periods and lower IL10 through pregnancy and postpartum. They may have different proinflammatory states that remain after birth without a reciprocal anti-inflammatory response.
Collapse
Affiliation(s)
- Roberta A Mancuso
- Department of Psychology and Neuroscience, Regis University, Denver, CO, USA.
| | - Kharah M Ross
- Centre for Social Sciences, Athabasca University, Athabasca, AB, Canada
| | - Eynav Accortt
- Reproductive Psychology Program, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mary Coussons-Read
- Department of Psychology, University of Colorado - Colorado Springs, Colorado Springs, CO, USA
| | - Michele L Okun
- Sleep and Biobehavioral Health Research Laboratory, University of Colorado - Colorado Springs, Colorado Springs, CO, USA
| | - Jessica Irwin
- Department of Psychology, University of La Verne, La Verne, CA, USA
| | - Judith Carroll
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Calvin J Hobel
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | |
Collapse
|
41
|
Kapembwa MS, Batman PA, Fleming SC, Griffin GE. HIV enteropathy and 'Slim disease': Historical and current perspectives. Int J Infect Dis 2024; 139:86-91. [PMID: 38052315 DOI: 10.1016/j.ijid.2023.11.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 12/07/2023] Open
Abstract
OBJECTIVES Chronic diarrhoea and severe wasting associated with HIV infection were first described in East African patients as slim disease (SD) in 1985. The main histological features are flattening of the villi (villous atrophy) and crypt hyperplasia (elongated crypts), i.e., HIV enteropathy (HIVE). Selective loss of mucosal clusters of differentiation 4 (CD4)+ T helper (Th)17+ lymphocytes is the immunological hallmark of HIVE. This review explores (i) the historical background of HIVE and SD, (ii) the relationship between gut mucosal CD4+ Th17+ and intestinal-resident intra-epithelial gamma delta (IRIE) T lymphocytes in pathogenesis of HIVE, (iii) the role of cytokines in regulation of intestinal epithelial proliferation, and (iv) the role of antiretroviral therapy in HIVE. METHODS Recent studies have highlighted the role of IRIE T lymphocytes, mostly CD8+, in regulating gut epithelial regeneration. CD4+Th17+ and IRIE T cells are necessary to maintain intestinal barrier integrity and mucosal antimicrobial immune defence. However, the immunological cross-talk between such lymphocyte sub-sets culminating in HIVE is uncertain. We undertook a narrative literature review under the headings 'HIVE', 'SD', and 'Highly active antiretroviral therapy (HAART). Relevant studies were located using the electronic search engines Google Scholar and PubMed from 1984 to 2022. RESULTS Depletion of Th17+ cells in the lamina propria, attributed to low-level viraemia, is accompanied by concomitant increase in the density of gut mucosal IRIE T lymphocytes in AIDS. The latter express a broad range of cytokines (interferon-gamma, tumor necrosis factor-alpha, interleukin-17) and chemokines e.g., keratinocyte growth factor, post exposure to HIV-infected cells. Keratinocyte growth factor induces epithelial proliferation mainly in the crypts, leading to functional immaturity of enterocytes, reduced gut absorptive surface area and malabsorption in animal experiments. Of note, the absence of IRIE T cells is associated with a reduction in epithelial cell turnover. Patients with HIVE receiving early HAART show enhanced expression of mucosal repair genes and improvement of gut symptoms. CONCLUSION Multiple lines of enquiry suggest HIVE is directly related to HIV infection and is a consequence of perturbations in mucosal CD4+Th17+ and IRIE T lymphocytes. The pathological result is enterocyte immaturity and dysfunction. SD whose main features are malabsorption, diarrhoea and weight loss, is a severe clinical expression of HIVE. A better understanding of immuno-pathogenesis of HIVE opens a window of opportunity for the potential use of immunotherapy in HIV disease and other T cell-mediated enteropathies.
Collapse
Affiliation(s)
- Moses Silungwe Kapembwa
- London Northwest Teaching Hospitals NHS Trust & Imperial College of Medicine, Northwick Park & St Mark's Hospitals, Harrow, UK.
| | - Philip Anthony Batman
- Department of Histopathology, Bradford Hospitals NHS Trust, Bradford Royal Infirmary, Bradford, UK
| | | | - George Edward Griffin
- Department of Cellular and Molecular Sciences. St George's Hospital Medical School. Cranmer Terrace, London, UK
| |
Collapse
|
42
|
Ahuja S, Zaheer S. Multifaceted TGF-β signaling, a master regulator: From bench-to-bedside, intricacies, and complexities. Cell Biol Int 2024; 48:87-127. [PMID: 37859532 DOI: 10.1002/cbin.12097] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/08/2023] [Accepted: 10/02/2023] [Indexed: 10/21/2023]
Abstract
Physiological embryogenesis and adult tissue homeostasis are regulated by transforming growth factor-β (TGF-β), an evolutionarily conserved family of secreted polypeptide factors, acting in an autocrine and paracrine manner. The role of TGF-β in inflammation, fibrosis, and cancer is complex and sometimes even contradictory, exhibiting either inhibitory or promoting effects depending on the stage of the disease. Under pathological conditions, especially fibrosis and cancer, overexpressed TGF-β causes extracellular matrix deposition, epithelial-mesenchymal transition, cancer-associated fibroblast formation, and/or angiogenesis. In this review article, we have tried to dive deep into the mechanism of action of TGF-β in inflammation, fibrosis, and carcinogenesis. As TGF-β and its downstream signaling mechanism are implicated in fibrosis and carcinogenesis blocking this signaling mechanism appears to be a promising avenue. However, targeting TGF-β carries substantial risk as this pathway is implicated in multiple homeostatic processes and is also known to have tumor-suppressor functions. There is a need for careful dosing of TGF-β drugs for therapeutic use and patient selection.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
43
|
Wu KY, Kulbay M, Daigle P, Nguyen BH, Tran SD. Nonspecific Orbital Inflammation (NSOI): Unraveling the Molecular Pathogenesis, Diagnostic Modalities, and Therapeutic Interventions. Int J Mol Sci 2024; 25:1553. [PMID: 38338832 PMCID: PMC10855920 DOI: 10.3390/ijms25031553] [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: 12/31/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Nonspecific orbital inflammation (NSOI), colloquially known as orbital pseudotumor, sometimes presents a diagnostic and therapeutic challenge in ophthalmology. This review aims to dissect NSOI through a molecular lens, offering a comprehensive overview of its pathogenesis, clinical presentation, diagnostic methods, and management strategies. The article delves into the underpinnings of NSOI, examining immunological and environmental factors alongside intricate molecular mechanisms involving signaling pathways, cytokines, and mediators. Special emphasis is placed on emerging molecular discoveries and approaches, highlighting the significance of understanding molecular mechanisms in NSOI for the development of novel diagnostic and therapeutic tools. Various diagnostic modalities are scrutinized for their utility and limitations. Therapeutic interventions encompass medical treatments with corticosteroids and immunomodulatory agents, all discussed in light of current molecular understanding. More importantly, this review offers a novel molecular perspective on NSOI, dissecting its pathogenesis and management with an emphasis on the latest molecular discoveries. It introduces an integrated approach combining advanced molecular diagnostics with current clinical assessments and explores emerging targeted therapies. By synthesizing these facets, the review aims to inform clinicians and researchers alike, paving the way for molecularly informed, precision-based strategies for managing NSOI.
Collapse
Affiliation(s)
- Kevin Y. Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Merve Kulbay
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 0A4, Canada
| | - Patrick Daigle
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Bich H. Nguyen
- CHU Sainte Justine Hospital, Montreal, QC H3T 1C5, Canada
| | - Simon D. Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
44
|
Tokifuji Y, Hayabuchi H, Sasaki T, Hara-Chikuma M, Hirota K, Takahashi H, Amagai M, Yoshimura A, Chikuma S. Targeting abatacept-resistant T-helper-17 cells by aldehyde dehydrogenase inhibition. iScience 2024; 27:108646. [PMID: 38226171 PMCID: PMC10788227 DOI: 10.1016/j.isci.2023.108646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/06/2023] [Accepted: 12/04/2023] [Indexed: 01/17/2024] Open
Abstract
IL-17-producing helper T (Th17) cells are long-lived and serve as central effector cells in chronic autoimmune diseases. The underlying mechanisms of Th17 persistence remain unclear. We demonstrated that abatacept, a CD28 antagonist, effectively prevented the development of skin disease in a Th17-dependent experimental autoimmune dermatitis model. Abatacept selectively inhibited the emergence of IL-7R-negative effector-phenotype T cells while allowing the survival and proliferation of IL-7R+ memory-phenotype cells. The surviving IL-7R+ Th17 cells expressed genes associated with alcohol/aldehyde detoxification and showed potential to transdifferentiate into IL-7R-negative effector cells. Inhibiting aldehyde dehydrogenase reduced IL-7R+ Th17 cells in vivo, independently of CD28, and exhibited additive effects when combined with abatacept. Our findings suggest that CD28 blockade prevents inflammation without eliminating persistent memory cells. These remaining memory cells can be targeted by other drugs, such as aldehyde dehydrogenase inhibitors, to limit their survival, thereby facilitating the treatment of chronic autoimmune diseases.
Collapse
Affiliation(s)
- Yukiko Tokifuji
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, East Lecture Hall 4F, Shinjuku, Tokyo 160-8582, Japan
| | - Hodaka Hayabuchi
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, East Lecture Hall 4F, Shinjuku, Tokyo 160-8582, Japan
| | - Takashi Sasaki
- Center for Supercentenarian Medical Research, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Mariko Hara-Chikuma
- Department of Pharmacology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Keiji Hirota
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Hayato Takahashi
- Department of Dermatology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, East Lecture Hall 4F, Shinjuku, Tokyo 160-8582, Japan
| | - Shunsuke Chikuma
- Department of Microbiology and Immunology, Keio University School of Medicine, 35 Shinanomachi, East Lecture Hall 4F, Shinjuku, Tokyo 160-8582, Japan
| |
Collapse
|
45
|
Khan IA, Singh N, Gunjan D, Dash NR, Nayak B, Gupta S, Saraya A. Elevated levels of peripheral Th17 cells and Th17-related cytokines in patients with periampullary adenocarcinoma. Hum Immunol 2024; 85:110748. [PMID: 38177009 DOI: 10.1016/j.humimm.2023.110748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/12/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024]
Abstract
AIM Periampullary adenocarcinoma (PAC) is a malignant tumor originating at the ampulla of Vater, distal common bile duct, head of the pancreas, ampulla and duodenum. The levels of circulating Th17 cells and Th17-related cytokines in patients with PAC remain unreported. Therefore, the aim of this study was to determine the levels of circulating Th17 cells and Th17-related cytokines in patients with PAC. MATERIALS AND METHODS Flow cytometry was used to measure Th17 cell proportions in PBMCs from 60 PAC patients and 30 healthy controls. Enzyme-linked immunosorbent assay (ELISA) was used to quantify IL-17A and IL-23 levels in serum samples, while quantitative reverse transcription polymerase chain reaction (qRT-PCR) assessed IL-17A mRNA expression and Th17-related transcription factors (RORγt and STAT3) in tissue samples. RESULTS The findings showed a substantial increase in Th17 cell percentages, elevated concentrations of IL-17A and IL-23, and higher mRNA expression levels of IL-17A, RORγt, and STAT3 in patients with PAC when compared to healthy controls (HCs). CONCLUSION Th17 cells play an important role in the pathogenesis of PAC and may represent potential therapeutic targets.
Collapse
Affiliation(s)
- Imteyaz Ahmad Khan
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Nidhi Singh
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Deepak Gunjan
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Nihar Ranjan Dash
- Department of Gastrointestinal Surgery, All India Institute of Medical Sciences, New Delhi, India
| | - Baibaswata Nayak
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Surabhi Gupta
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, India
| | - Anoop Saraya
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
46
|
Borlongan MC, Saha D, Wang H. Tumor Microenvironment: A Niche for Cancer Stem Cell Immunotherapy. Stem Cell Rev Rep 2024; 20:3-24. [PMID: 37861969 DOI: 10.1007/s12015-023-10639-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2023] [Indexed: 10/21/2023]
Abstract
Tumorigenic Cancer Stem Cells (CSCs), often called tumor-initiating cells (TICs), represent a unique subset of cells within the tumor milieu. They stand apart from the bulk of tumor cells due to their exceptional self-renewal, metastatic, and differentiation capabilities. Despite significant progress in classifying CSCs, these cells remain notably resilient to conventional radiotherapy and chemotherapy, contributing to cancer recurrence. In this review, our objective is to explore novel avenues of research that delve into the distinctive characteristics of CSCs within their surrounding tumor microenvironment (TME). We will start with an overview of the defining features of CSCs and then delve into their intricate interactions with cells from the lymphoid lineage, namely T cells, B cells, and natural killer (NK) cells. Furthermore, we will discuss their dynamic interplay with myeloid lineage cells, including macrophages, neutrophils, and myeloid-derived suppressor cells (MDSCs). Moreover, we will illuminate the crosstalk between CSCs and cells of mesenchymal origin, specifically fibroblasts, adipocytes, and endothelial cells. Subsequently, we will underscore the pivotal role of CSCs within the context of the tumor-associated extracellular matrix (ECM). Finally, we will highlight pre-clinical and clinical studies that target CSCs within the intricate landscape of the TME, including CAR-T therapy, oncolytic viruses, and CSC-vaccines, with the ultimate goal of uncovering novel avenues for CSC-based cancer immunotherapy.
Collapse
Affiliation(s)
- Mia C Borlongan
- College of Medicine, California Northstate University, 9700 West Taron Drive, Elk Grove, CA, 95757, USA
| | - Dipongkor Saha
- Department of Pharmaceutical and Biomedical Sciences College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, CA, 95757, USA.
| | - Hongbin Wang
- College of Medicine, California Northstate University, 9700 West Taron Drive, Elk Grove, CA, 95757, USA.
- Department of Pharmaceutical and Biomedical Sciences College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, CA, 95757, USA.
- Master Program of Pharmaceutical Sciences College of Graduate Studies, Department of Pharmaceutical and Biomedical Sciences College of Pharmacy, Department of Basic Science College of Medicine, California Northstate University, 9700 West Taron Drive, Elk Grove, CA, 95757, USA.
| |
Collapse
|
47
|
Manohar K, Mesfin FM, Liu J, Shelley WC, Brokaw JP, Markel TA. Effect of Oral Chondroitin Sulfate Supplementation on Acute Brain Injury in a Murine Necrotizing Enterocolitis Model. J Am Coll Surg 2024; 238:82-98. [PMID: 37870229 DOI: 10.1097/xcs.0000000000000896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a devastating condition where inflammatory changes and necrosis in the gut results in activation of brain microglia and subsequent neurodevelopmental impairment. Chondroitin sulfate (CS) is a glycosaminoglycan in human breast milk that is absent in conventional formulas. We hypothesized that oral formula supplementation with CS during a murine model of experimental NEC would not only attenuate intestinal injury, but also brain injury. STUDY DESIGN NEC was induced in mouse pups on postnatal days (PNDs) 5 to 8. Three conditions were studied: (1) breastfed controls, (2) NEC, and (3) NEC+enteral CS (formula+200 mg/kg/d of CS). Pups were euthanized on PND 9 or reunited with dams by the evening of PND 8. Intestinal segments were H&E stained, and immunohistochemistry was performed on brain tissue for Iba-1 to assess for microglial morphology and cortical changes. Neurodevelopmental assays were performed on mice reunited with foster dams on PND 9. Single-cell RNA-sequencing analysis was performed on human intestinal epithelial cells exposed to (1) nothing, (2) hydrogen peroxide (H 2 O 2 ) alone, or (3) H 2 O 2 + CS to look at the differential gene expression between groups. Groups were compared with ANOVA or Kruskal-Wallis tests as appropriate with p < 0.05 considered significant. RESULTS Compared with NEC, mice treated with oral CS showed improved clinical outcomes, decreased intestinal injury, and attenuated microglial activation and deleterious cortical change. Mice with CS performed better on early neurodevelopmental assays when compared with NEC alone. Single-cell analysis of HIEC-6 cells demonstrated that CS treatment down regulated several inflammatory pathways including nuclear factor κB-suggesting an explanation for the improved Th17 intestinal cytokine profile. CONCLUSIONS Oral CS supplementation improved both physiological, clinical, and developmental outcomes. These data suggest that CS is a safe compound for formula supplementation for the prevention of NEC.
Collapse
Affiliation(s)
- Krishna Manohar
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| | - Fikir M Mesfin
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| | - Jianyun Liu
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| | - W Christopher Shelley
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| | - John P Brokaw
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| | - Troy A Markel
- From the Department of Surgery, Indiana University School of Medicine, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
- Riley Hospital for Children at Indiana University Health, Indianapolis, IN (Manohar, Mesfin, Liu, Shelley, Brokaw, Markel)
| |
Collapse
|
48
|
Fanizza J, D’Amico F, Lusetti F, Fasulo E, Allocca M, Furfaro F, Zilli A, Parigi TL, Radice S, Peyrin-Biroulet L, Danese S, Fiorino G. The Role of IL-23 Inhibitors in Crohn's Disease. J Clin Med 2023; 13:224. [PMID: 38202231 PMCID: PMC10779938 DOI: 10.3390/jcm13010224] [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: 11/24/2023] [Revised: 12/27/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Promoting a Th17 pathogenic response, the interleukin (IL)-23 pathway is crucial in the pathophysiology of inflammatory bowel disease (IBD). With a favorable safety profile, ustekinumab, a monoclonal antibody targeting the shared p40 component of IL-12/23, is currently approved for the treatment of IBD in patients with disease refractory to corticosteroids and biologic drugs. Risankizumab, mirikizumab, and guselkumab are specific IL-23p19 antagonists tested for the treatment of Crohn's disease (CD). However, only risankizumab currently has been approved for its treatment. Trials with guselkumab and mirikizumab are currently ongoing, with promising preliminary efficacy and safety results. In this review, we provide a summary of the current knowledge about selective IL-23 inhibitors, focusing on their positioning in the therapeutic algorithm of patients with moderate to severe CD.
Collapse
Affiliation(s)
- Jacopo Fanizza
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (J.F.); (F.D.); (F.L.); (E.F.); (M.A.); (F.F.); (A.Z.); (T.L.P.); (S.R.); (S.D.)
| | - Ferdinando D’Amico
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (J.F.); (F.D.); (F.L.); (E.F.); (M.A.); (F.F.); (A.Z.); (T.L.P.); (S.R.); (S.D.)
- Department of Biomedical Sciences, Humanitas University, 20089 Milan, Italy
| | - Francesca Lusetti
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (J.F.); (F.D.); (F.L.); (E.F.); (M.A.); (F.F.); (A.Z.); (T.L.P.); (S.R.); (S.D.)
- Department of Gastroenterology, IRCCS Policlinico San Matteo, University of Pavia, 27100 Pavia, Italy
| | - Ernesto Fasulo
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (J.F.); (F.D.); (F.L.); (E.F.); (M.A.); (F.F.); (A.Z.); (T.L.P.); (S.R.); (S.D.)
| | - Mariangela Allocca
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (J.F.); (F.D.); (F.L.); (E.F.); (M.A.); (F.F.); (A.Z.); (T.L.P.); (S.R.); (S.D.)
| | - Federica Furfaro
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (J.F.); (F.D.); (F.L.); (E.F.); (M.A.); (F.F.); (A.Z.); (T.L.P.); (S.R.); (S.D.)
| | - Alessandra Zilli
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (J.F.); (F.D.); (F.L.); (E.F.); (M.A.); (F.F.); (A.Z.); (T.L.P.); (S.R.); (S.D.)
| | - Tommaso Lorenzo Parigi
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (J.F.); (F.D.); (F.L.); (E.F.); (M.A.); (F.F.); (A.Z.); (T.L.P.); (S.R.); (S.D.)
| | - Simona Radice
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (J.F.); (F.D.); (F.L.); (E.F.); (M.A.); (F.F.); (A.Z.); (T.L.P.); (S.R.); (S.D.)
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France;
- Inserm, NGERE, University of Lorraine, F-54000 Nancy, France
- INFINY Institute, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France
- FHU-CURE, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France
- Groupe Hospitalier Privé Ambroise Paré-Hartmann, Paris IBD Center, F-92200 Neuilly-sur-Seine, France
- Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Silvio Danese
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (J.F.); (F.D.); (F.L.); (E.F.); (M.A.); (F.F.); (A.Z.); (T.L.P.); (S.R.); (S.D.)
| | - Gionata Fiorino
- Department of Gastroenterology and Endoscopy, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, 20132 Milan, Italy; (J.F.); (F.D.); (F.L.); (E.F.); (M.A.); (F.F.); (A.Z.); (T.L.P.); (S.R.); (S.D.)
- IBD Unit, Department of Gastroenterology and Digestive Endoscopy, San Camillo-Forlanini Hospital, 00152 Rome, Italy
| |
Collapse
|
49
|
Lu L, Huang Y, Song M, Sun N, Xia L, Yu M, Zhao M, Qiu R, Chen JA, Zhao Y, Wang H, Guo H, Li Y, Zhu D, Wang Y, Xie Q. Discovery of Biaryl Amide Derivatives as Potent, Selective, and Orally Bioavailable RORγt Agonists for Cancer Immunotherapy. J Med Chem 2023; 66:16091-16108. [PMID: 37982494 DOI: 10.1021/acs.jmedchem.3c01492] [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: 11/21/2023]
Abstract
The master transcription factor receptor retinoic acid receptor-related orphan receptor γt (RORγt) regulates the differentiation of T-helper 17 (Th17) cells and the production of interleukin-17 (IL-17). Activation of RORγt+ T cells in the tumor microenvironment promotes immune infiltration to more effectively inhibit tumor growth. Therefore, RORγt agonists provide a reachable approach to cancer immunotherapy. Herein, a series of biaryl amide derivatives as novel RORγt agonists were designed, synthesized, and evaluated. Starting from the reported RORγt inverse agonist GSK805 (1), "functionality switching" and structure-based drug optimization led to the discovery of a promising RORγt agonist lead compound 14, which displayed potent and selective RORγt agonist activity and significantly improved metabolic stability. With excellent in vivo pharmacokinetic profiles, compound 14 demonstrated robust efficacy in preclinical tumor models of mouse B16F10 melanoma and LLC lung adenocarcinoma. Taken together, current studies indicate that 14 deserves further investigation as a potential lead RORγt agonist for cancer immunotherapy.
Collapse
Affiliation(s)
- Lixue Lu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yafei Huang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Meiqi Song
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Nannan Sun
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Li Xia
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mingcheng Yu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Meiling Zhao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Ruomeng Qiu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Ji-An Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yunpeng Zhao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Haojie Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Huimin Guo
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yan Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Di Zhu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yonghui Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Qiong Xie
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
50
|
Emami Nejad A, Mostafavi Zadeh SM, Nickho H, Sadoogh Abbasian A, Forouzan A, Ahmadlou M, Nedaeinia R, Shaverdi S, Manian M. The role of microRNAs involved in the disorder of blood-brain barrier in the pathogenesis of multiple sclerosis. Front Immunol 2023; 14:1281567. [PMID: 38193092 PMCID: PMC10773759 DOI: 10.3389/fimmu.2023.1281567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/30/2023] [Indexed: 01/10/2024] Open
Abstract
miRNAs are involved in various vital processes, including cell growth, development, apoptosis, cellular differentiation, and pathological cellular activities. Circulating miRNAs can be detected in various body fluids including serum, plasma, saliva, and urine. It is worth mentioning that miRNAs remain stable in the circulation in biological fluids and are released from membrane-bound vesicles called exosomes, which protect them from RNase activity. It has been shown that miRNAs regulate blood-brain barrier integrity by targeting both tight junction and adherens junction molecules and can also influence the expression of inflammatory cytokines. Some recent studies have examined the impact of certain commonly used drugs in Multiple Sclerosis on miRNA levels. In this review, we will focus on the recent findings on the role of miRNAs in multiple sclerosis, including their role in the cause of MS and molecular mechanisms of the disease, utilizing miRNAs as diagnostic and clinical biomarkers, using miRNAs as a therapeutic modality or target for Multiple Sclerosis and drug responses in patients, elucidating their importance as prognosticators of disease progression, and highlighting their potential as a future treatment for MS.
Collapse
Affiliation(s)
| | - Seyed Mostafa Mostafavi Zadeh
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid Nickho
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Sadoogh Abbasian
- Department of Internal Medicine, School of Medicine, Amiralmomenin Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Azim Forouzan
- Department of Internal Medicine, School of Medicine, Amiralmomenin Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Mojtaba Ahmadlou
- Department of Biostatistics, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saham Shaverdi
- Department of Biology, Payame Noor University (PNU), Tehran, Iran
| | - Mostafa Manian
- Department of Medical Laboratory Science, Faculty of Medicine, Islamic Azad University, Kermanshah, Iran
- Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|