1
|
Prajapati SK, Lekkala L, Yadav D, Jain S, Yadav H. Microbiome and Postbiotics in Skin Health. Biomedicines 2025; 13:791. [PMID: 40299368 PMCID: PMC12025169 DOI: 10.3390/biomedicines13040791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/30/2025] Open
Abstract
The skin microbiome, a diverse and dynamic ecosystem of microorganisms, plays a pivotal role in maintaining skin health by interacting with skin cells, immune components, and structural barriers. It is essential for skin homeostasis, immune defense, and protection against pathogenic colonization. Dysbiosis in the microbiome has been implicated in numerous dermatological conditions, including acne, eczema, psoriasis, and rosacea. Acne, the most prevalent skin condition, affects up to 85% of individuals at some point in their lives, while eczema and psoriasis impose significant public health and economic burdens. The composition of the skin microbiome varies across skin types and anatomical sites, with sebaceous, moist, and dry areas fostering distinct microbial communities. Emerging therapeutic strategies such as microbiome-targeted treatments offer novel avenues for addressing skin diseases. Among these approaches, postbiotics have gained significant attention for their safety and efficacy. Unlike probiotics, postbiotics are non-viable microbial cells or their metabolites, which reduce safety concerns while providing functional benefits such as UV protection and wound healing. This review consolidates current insights into the role of the skin microbiome in health and disease, emphasizing postbiotics as a promising therapeutic strategy by exploring the clinical and commercial potential of microbiome-based treatments, particularly postbiotics, and their ability to redefine dermatological care and improve patient outcomes.
Collapse
Affiliation(s)
- Santosh Kumar Prajapati
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (S.K.P.); (L.L.); (D.Y.); (S.J.)
- Center of Excellence in Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Lalitha Lekkala
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (S.K.P.); (L.L.); (D.Y.); (S.J.)
- Center of Excellence in Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Dhananjay Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (S.K.P.); (L.L.); (D.Y.); (S.J.)
- Center of Excellence in Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Shalini Jain
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (S.K.P.); (L.L.); (D.Y.); (S.J.)
- Center of Excellence in Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (S.K.P.); (L.L.); (D.Y.); (S.J.)
- Center of Excellence in Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
2
|
Wu X, Hao J, Jiang K, Wu M, Zhao X, Zhang X. Neuroinflammation and pathways that contribute to tourette syndrome. Ital J Pediatr 2025; 51:63. [PMID: 40022157 PMCID: PMC11871796 DOI: 10.1186/s13052-025-01874-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/26/2025] [Indexed: 03/03/2025] Open
Abstract
Tourette syndrome (TS), a neurological and psychological disease, typically exhibit motor and phonic tics. The pathophysiology of TS remains controversial. Currently, the recognized pathogenesis of TS is the imbalance of neurotransmitters, involving abnormality of the cortex-striatum-thalamus-cortex circuit. Recently, clinical researches demonstrate that triggers such as infection and allergic reaction could lead to the onset or exacerbation of tic symptoms. Current studies have also suggested that neural-immune crosstalk caused by inflammation is also associated with TS, potentially leading to the occurrence of tics by inducing neurotransmitter abnormalities. Herein, we review inflammation-related factors contributing to the occurrence of TS as well as the mechanisms by which immune-inflammatory pathways mediate the onset of TS. This aims to clarify the pathogenesis of TS and provide a theoretical basis for the treatment of TS.
Collapse
Affiliation(s)
- Xinnan Wu
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juanjuan Hao
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Keyu Jiang
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Wu
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Zhao
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Zhang
- Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Saito M, McDonald KA, Grier AK, Meghwani H, Rangel-Moreno J, Becerril-Villanueva E, Gamboa-Dominguez A, Bruno J, Beck CA, Proctor RA, Kates SL, Schwarz EM, Muthukrishnan G. Immune Checkpoint Molecules as Biomarkers of Staphylococcus aureus Bone Infection and Clinical Outcome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.30.630837. [PMID: 39803468 PMCID: PMC11722373 DOI: 10.1101/2024.12.30.630837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Staphylococcus aureus prosthetic joint infections (PJIs) are broadly considered incurable, and clinical diagnostics that guide conservative vs. aggressive surgical treatments don't exist. Multi-omics studies in a humanized NSG-SGM3 BLT mouse model demonstrate human T cells: 1) are remarkably heterogenous in gene expression and numbers, and 2) exist as a mixed population of activated, progenitor-exhausted, and terminally-exhausted Th1/Th17 cells with increased expression of immune checkpoint proteins (LAG3, TIM-3). Importantly, these proteins are upregulated in the serum and the bone marrow of S. aureus PJI patients. A multiparametric nomogram combining high serum immune checkpoint protein levels with low proinflammatory cytokine levels (IFN-γ, IL-2, TNF-α, IL-17) revealed that TIM-3 was highly predictive of adverse disease outcomes (AUC=0.89). Hence, T cell impairment in the form of immune checkpoint expression and exhaustion could be a functional biomarker for S. aureus PJI disease outcome, and blockade of checkpoint proteins could potentially improve outcomes following surgery.
Collapse
Affiliation(s)
- Motoo Saito
- The Center for Musculoskeletal Research, Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
| | - Katya A. McDonald
- The Center for Musculoskeletal Research, Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Alex K. Grier
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Division of Gastroenterology and Hepatology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Himanshu Meghwani
- The Center for Musculoskeletal Research, Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Javier Rangel-Moreno
- Division of Allergy, Immunology, Rheumatology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Enrique Becerril-Villanueva
- Psychoimmunology laboratory, Instituto Nacional de Psiquiatría “Ramón de la Fuente Muñiz.” Mexico City, Mexico
| | - Armando Gamboa-Dominguez
- Deparment of Pathology, Instituto Nacional de Ciencias Médicas Y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jennifer Bruno
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Christopher A. Beck
- The Center for Musculoskeletal Research, Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
| | - Richard A. Proctor
- Departments of Medical Microbiology/Immunology and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Stephen L. Kates
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - Edward M. Schwarz
- The Center for Musculoskeletal Research, Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Gowrishankar Muthukrishnan
- The Center for Musculoskeletal Research, Department of Orthopedics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
4
|
Hussein H, Van Remoortel S, Boeckxstaens GE. Irritable bowel syndrome: When food is a pain in the gut. Immunol Rev 2024; 326:102-116. [PMID: 39037230 DOI: 10.1111/imr.13374] [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] [Indexed: 07/23/2024]
Abstract
Irritable bowel syndrome (IBS) is a chronic gastrointestinal condition associated with altered bowel habits and recurrent abdominal pain, often triggered by food intake. Current treatments focus on improving stool pattern, but effective treatments for pain in IBS are still lacking due to our limited understanding of pathophysiological mechanisms. Visceral hypersensitivity (VHS), or abnormal visceral pain perception, underlies abdominal pain development in IBS, and mast cell activation has been shown to play an important role in the development of VHS. Our work recently revealed that abdominal pain in response to food intake is induced by the sensitization of colonic pain-sensing neurons by histamine produced by activated mast cells following a local IgE response to food. In this review, we summarize the current knowledge on abdominal pain and VHS pathophysiology in IBS, we outline the work leading to the discovery of the role of histamine in abdominal pain, and we introduce antihistamines as a novel treatment option to manage chronic abdominal pain in patients with IBS.
Collapse
Affiliation(s)
- Hind Hussein
- Center for Intestinal Neuro-Immune Interactions, Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Samuel Van Remoortel
- Center for Intestinal Neuro-Immune Interactions, Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Guy E Boeckxstaens
- Center for Intestinal Neuro-Immune Interactions, Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism, and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| |
Collapse
|
5
|
Yun AE, Johnson LB. Recurrent streptococcal toxic shock syndrome due to distinct episodes of Streptococcus dysgalactiae and Streptococcus agalactiae septic arthritis. BMJ Case Rep 2024; 17:e260409. [PMID: 39142840 DOI: 10.1136/bcr-2024-260409] [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] [Indexed: 08/16/2024] Open
Abstract
Streptococcal toxic shock syndrome (STSS) is an uncommon disorder characterised by hypotension and multiorgan failure in the setting of streptococcal infection. Recurrent STSS is rare and has been due to recurrence of the same streptococcal species. Here, we present a case of a patient who developed recurrent STSS from a Streptococcus dysgalactiae right native joint septic arthritis and subsequently from a Streptococcus agalactiae left native joint septic arthritis.
Collapse
Affiliation(s)
| | - Leonard B Johnson
- Infectious Disease, Ascension St John Hospital, Detroit, Michigan, USA
| |
Collapse
|
6
|
Rampersadh K, Salie MT, Engel KC, Moodley C, Zühlke LJ, Engel ME. Presence of Group A streptococcus frequently assayed virulence genes in invasive disease: a systematic review and meta-analysis. Front Cell Infect Microbiol 2024; 14:1337861. [PMID: 39055978 PMCID: PMC11270091 DOI: 10.3389/fcimb.2024.1337861] [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: 11/13/2023] [Accepted: 01/18/2024] [Indexed: 07/28/2024] Open
Abstract
Introduction It is currently unclear what the role of Group A streptococcus (GAS) virulence factors (VFs) is in contributing to the invasive potential of GAS. This work investigated the evidence for the association of GAS VFs with invasive disease. Methods We employed a broad search strategy for studies reporting the presence of GAS VFs in invasive and non-invasive GAS disease. Data were independently extracted by two reviewers, quality assessed, and meta-analyzed using Stata®. Results A total of 32 studies reported on 45 putative virulence factors [invasive (n = 3,236); non-invasive (n = 5,218)], characterized by polymerase chain reaction (PCR) (n = 30) and whole-genome sequencing (WGS) (n = 2). The risk of bias was rated as low and moderate, in 23 and 9 studies, respectively. Meta-,analyses of high-quality studies (n = 23) revealed a significant association of speM [OR, 1.64 (95%CI, 1.06; 2.52)] with invasive infection. Meta-analysis of WGS studies demonstrated a significant association of hasA [OR, 1.91 (95%CI, 1.36; 2.67)] and speG [OR, 2.83 (95%CI, 1.63; 4.92)] with invasive GAS (iGAS). Meta-analysis of PCR studies indicated a significant association of speA [OR, 1.59 (95%CI, 1.10; 2.30)] and speK [OR, 2.95 (95%CI, 1.81; 4.80)] with invasive infection. A significant inverse association was observed between prtf1 [OR, 0.42 (95%CI, 0.20; 0.87)] and invasive infection. Conclusion This systematic review and genomic meta-analysis provides evidence of a statistically significant association with invasive infection for the hasA gene, while smeZ, ssa, pnga3, sda1, sic, and NaDase show statistically significantly inverse associations with invasive infection. SpeA, speK, and speG are associated with GAS virulence; however, it is unclear if they are markers of invasive infection. This work could possibly aid in developing preventative strategies.
Collapse
Affiliation(s)
- Kimona Rampersadh
- AFROStrep Research Group, Department of Medicine and Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - M. Taariq Salie
- AFROStrep Research Group, Department of Medicine and Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Kelin C. Engel
- AFROStrep Research Group, Department of Medicine and Cape Heart Institute, University of Cape Town, Cape Town, South Africa
| | - Clinton Moodley
- Department of Pathology, Division of Medical Microbiology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- The National Health Laboratory Service, Microbiology, Groote Schuur Hospital, Cape Town, South Africa
| | - Liesl J. Zühlke
- Division of Paediatric Cardiology, Department of Paediatrics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council, Parrow Valley, Cape Town, South Africa
| | - Mark E. Engel
- AFROStrep Research Group, Department of Medicine and Cape Heart Institute, University of Cape Town, Cape Town, South Africa
- South African Medical Research Council, Parrow Valley, Cape Town, South Africa
| |
Collapse
|
7
|
Lichota A, Gwozdzinski K, Kowalczyk E, Kowalczyk M, Sienkiewicz M. Contribution of staphylococcal virulence factors in the pathogenesis of thrombosis. Microbiol Res 2024; 283:127703. [PMID: 38537329 DOI: 10.1016/j.micres.2024.127703] [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: 08/23/2023] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 04/17/2024]
Abstract
Staphylococci are responsible for many infections in humans, starting with skin and soft tissue infections and finishing with invasive diseases such as endocarditis, sepsis and pneumonia, which lead to high mortality. Patients with sepsis often demonstrate activated clotting pathways, decreased levels of anticoagulants, decreased fibrinolysis, activated endothelial surfaces and activated platelets. This results in disseminated intravascular coagulation and formation of a microthrombus, which can lead to a multiorgan failure. This review describes various staphylococcal virulence factors that contribute to vascular thrombosis, including deep vein thrombosis in infected patients. The article presents mechanisms of action of different factors released by bacteria in various host defense lines, which in turn can lead to formation of blood clots in the vessels.
Collapse
Affiliation(s)
- Anna Lichota
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Lodz, Poland.
| | | | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, Medical University of Lodz, Lodz, Poland
| | | | - Monika Sienkiewicz
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
8
|
Kaneko S, Noguchi Y, Hatano M, Shimbo A, Irabu H, Furuno K, Iwata N, Fujimura J, Akamine K, Kobayashi A, Endo T, Morio T, Shimizu M. Clinical Usefulness of T-Cell Receptor Vβ Repertoire Analysis for Differentiating Multisystem Inflammatory Syndrome in Japanese Children From Toxic Shock Syndrome and Kawasaki Disease. Pediatr Infect Dis J 2024; 43:e125-e127. [PMID: 38134372 DOI: 10.1097/inf.0000000000004227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
The specific expansion of T-cell receptor β chain variable region (TCR-Vβ21.3 + ) CD4 + and CD8 + T cells was observed in Japanese patients with multisystem inflammatory syndrome in children. In contrast, these findings were not observed in patients with toxic shock syndrome and Kawasaki disease. T-cell receptor β chain variable region repertoire analysis to detect specific expansion of Vβ21.3 + T cells might be useful for differentiating multisystem inflammatory syndrome in children from toxic shock syndrome and Kawasaki disease.
Collapse
Affiliation(s)
- Shuya Kaneko
- From the Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yusuke Noguchi
- From the Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Maho Hatano
- From the Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Asami Shimbo
- From the Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hitoshi Irabu
- From the Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kenji Furuno
- Department of General Pediatrics and Interdisciplinary Medicine, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Naomi Iwata
- Department of Infection and immunology, Aichi Children's Health and Medical Center, Aichi, Japan
| | - Junya Fujimura
- Department of Pediatrics, Kakogawa Central City Hospital, Kakogawa, Japan
| | - Keiji Akamine
- Department of Nephrology and Rheumatology, Tokyo Metropolitan Children's Medical Center, Fuchu, Tokyo, Japan
| | - Anna Kobayashi
- Department of Pediatrics, University of Yamanashi, Yamanashi, Japan
| | - Takuya Endo
- Department of Pediatrics, Saitama Medical University, Saitama, Japan
| | - Tomohiro Morio
- From the Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masaki Shimizu
- From the Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
9
|
Ohashi A, Murayama MA, Miyabe Y, Yudoh K, Miyabe C. Streptococcal infection and autoimmune diseases. Front Immunol 2024; 15:1361123. [PMID: 38464518 PMCID: PMC10920276 DOI: 10.3389/fimmu.2024.1361123] [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: 12/25/2023] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Excessive activation of immune cells by environmental factors, such as infection or individual genetic risk, causes various autoimmune diseases. Streptococcus species are gram-positive bacteria that colonize the nasopharynx, respiratory tract, gastrointestinal tract, genitourinary tract, and skin. Group A Streptococcus (GAS) species cause various symptoms, ranging from mild infections, such as tonsillitis and pharyngitis, to serious infections, such as necrotizing fasciitis and streptococcal toxic shock syndrome. The contribution of GAS infections to several autoimmune diseases, including acute rheumatic fever, vasculitis, and neuropsychiatric disorders, has been studied. In this review, we focus on the association between streptococcal infections and autoimmune diseases, and discuss current research on the mechanisms underlying the initiation and progression of autoimmune diseases.
Collapse
Affiliation(s)
- Ayaka Ohashi
- Department of Immunology and Parasitology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Masanori A. Murayama
- Department of Animal Models for Human Diseases, Institute of Biomedical Science, Kansai Medical University, Osaka, Japan
| | - Yoshishige Miyabe
- Department of Immunology and Parasitology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Kazuo Yudoh
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Chie Miyabe
- Department of Frontier Medicine, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
10
|
Zhao XC, Ju B, Xiu NN, Sun XY, Meng FJ. When inflammatory stressors dramatically change, disease phenotypes may transform between autoimmune hematopoietic failure and myeloid neoplasms. Front Immunol 2024; 15:1339971. [PMID: 38426096 PMCID: PMC10902444 DOI: 10.3389/fimmu.2024.1339971] [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: 11/17/2023] [Accepted: 01/24/2024] [Indexed: 03/02/2024] Open
Abstract
Aplastic anemia (AA) and hypoplastic myelodysplastic syndrome are paradigms of autoimmune hematopoietic failure (AHF). Myelodysplastic syndrome and acute myeloid leukemia are unequivocal myeloid neoplasms (MNs). Currently, AA is also known to be a clonal hematological disease. Genetic aberrations typically observed in MNs are detected in approximately one-third of AA patients. In AA patients harboring MN-related genetic aberrations, a poor response to immunosuppressive therapy (IST) and an increased risk of transformation to MNs occurring either naturally or after IST are predicted. Approximately 10%-15% of patients with severe AA transform the disease phenotype to MNs following IST, and in some patients, leukemic transformation emerges during or shortly after IST. Phenotypic transformations between AHF and MNs can occur reciprocally. A fraction of advanced MN patients experience an aplastic crisis during which leukemic blasts are repressed. The switch that shapes the disease phenotype is a change in the strength of extramedullary inflammation. Both AHF and MNs have an immune-active bone marrow (BM) environment (BME). In AHF patients, an inflamed BME can be evoked by infiltrated immune cells targeting neoplastic molecules, which contributes to the BM-specific autoimmune impairment. Autoimmune responses in AHF may represent an antileukemic mechanism, and inflammatory stressors strengthen antileukemic immunity, at least in a significant proportion of patients who have MN-related genetic aberrations. During active inflammatory episodes, normal and leukemic hematopoieses are suppressed, which leads to the occurrence of aplastic cytopenia and leukemic cell regression. The successful treatment of underlying infections mitigates inflammatory stress-related antileukemic activities and promotes the penetration of leukemic hematopoiesis. The effect of IST is similar to that of treating underlying infections. Investigating inflammatory stress-powered antileukemic immunity is highly important in theoretical studies and clinical practice, especially given the wide application of immune-activating agents and immune checkpoint inhibitors in the treatment of hematological neoplasms.
Collapse
Affiliation(s)
- Xi-Chen Zhao
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao, Shandong, China
| | - Bo Ju
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao, Shandong, China
| | - Nuan-Nuan Xiu
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao, Shandong, China
| | - Xiao-Yun Sun
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao, Shandong, China
| | - Fan-Jun Meng
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
11
|
Amarnani A, Silverman GJ. Understanding the roles of the microbiome in autoimmune rheumatic diseases. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2023; 4:177-187. [PMID: 38125641 PMCID: PMC10729600 DOI: 10.2478/rir-2023-0027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/06/2023] [Indexed: 12/23/2023]
Abstract
The gut microbiome represents a potential promising therapeutic target for autoimmune diseases. This review summarizes the current knowledge on the links between the gut microbiome and several autoimmune rheumatic diseases including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE) spondyloarthropathies (SpA), Sjogren's syndrome (SS), and systemic sclerosis (SSc). Evidence from studies of RA and SLE patients suggests that alterations in the gut microbiome composition and function contribute to disease development and progression through increased gut permeability, with microbes and microbial metabolites driving an excessive systemic activation of the immune system. Also, there is growing evidence that gut dysbiosis and subsequent immune cell activation may contribute to disease pathogenesis in SpA and SS. For SSc, there are fewer, but these are still informative, reports on alterations in the gut microbiome. In general, the complex interplay between the microbiome and the immune system is still not fully understood. Here we discuss the current knowledge of the link between the gut microbiome and autoimmune rheumatic diseases, highlighting potentially fertile areas for future research and make considerations on the potential benefits of strategies that restore gut microbiome homeostasis.
Collapse
Affiliation(s)
- Abhimanyu Amarnani
- Department of Medicine, NYU Grossman School of Medicine, New York, NYUSA
| | - Gregg J. Silverman
- Department of Medicine, NYU Grossman School of Medicine, New York, NYUSA
| |
Collapse
|
12
|
Moran MC, Brewer MG, Schlievert PM, Beck LA. S. aureus virulence factors decrease epithelial barrier function and increase susceptibility to viral infection. Microbiol Spectr 2023; 11:e0168423. [PMID: 37737609 PMCID: PMC10581065 DOI: 10.1128/spectrum.01684-23] [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: 04/24/2023] [Accepted: 07/03/2023] [Indexed: 09/23/2023] Open
Abstract
Individuals with atopic dermatitis (AD) are highly colonized by Staphylococcus aureus and are more susceptible to severe viral complications. We hypothesized that S. aureus secreted virulence factors may alter keratinocyte biology to enhance viral susceptibility through disruption of the skin barrier, impaired keratinocyte differentiation, and/or inflammation. To address this hypothesis, human keratinocytes were exposed to conditioned media from multiple S. aureus strains that vary in virulence factor production (USA300, HG003, and RN4220) or select purified virulence factors. We have identified the S. aureus enterotoxin-like superantigen SElQ, as a virulence factor of interest, since it is highly produced by USA300 and was detected on the skin of 53% of AD subjects (n = 72) in a study conducted by our group. Treatment with USA300 conditioned media or purified SElQ resulted in a significant increase in keratinocyte susceptibility to infection with vaccinia virus, and also significantly decreased barrier function. Importantly, we have previously demonstrated that keratinocyte differentiation influences susceptibility to viral infection, and our qPCR observations indicated that USA300 S. aureus and SElQ alter differentiation in keratinocytes. CRISPR/Cas9 was used to knock out CD40, a potential enterotoxin receptor on epithelial cells. We found that CD40 expression on keratinocytes was not completely necessary for SElQ-mediated responses, as measured by proinflammatory cytokine expression and barrier function. Together, these findings support that select S. aureus virulence factors, particularly SElQ, enhance the susceptibility of epidermal cells to viral infection, which may contribute to the increased cutaneous infections observed in individuals with AD. IMPORTANCE Staphylococcus aureus skin colonization and infection are frequently observed in individuals with atopic dermatitis. Many S. aureus strains belong to the clonal group USA300, and these strains produce superantigens including the staphylococcal enterotoxin-like Q (SElQ). Our studies highlight that SElQ may play a key role by altering keratinocyte differentiation and reducing barrier function; collectively, this may explain the AD-specific enhanced infection risk to cutaneous viruses. It is unclear what receptor mediates SElQ's effects on keratinocytes. We have shown that one putative surface receptor, CD40, was not critical for its effects on proinflammatory cytokine production or barrier function.
Collapse
Affiliation(s)
- Mary C. Moran
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology & Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Matthew G. Brewer
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | | | - Lisa A. Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
13
|
Duarte A, Montagna DR, Pastorini M, Alemán M. Apoptosis-mediated inhibition of human T-cell acute lymphoblastic leukemia upon treatment with Staphylococus Aureus enterotoxin-superantigen. Front Immunol 2023; 14:1176432. [PMID: 37377961 PMCID: PMC10291079 DOI: 10.3389/fimmu.2023.1176432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Patients with relapsed T cell acute lymphoblastic leukemia (T-ALL) have limited therapeutic options and poor prognosis. The finding of efficient strategies against this refractory neoplasm is a medical priority. Superantigens (SAgs) are viral and bacterial proteins that bind to major histocompatibility complex class II molecules as unprocessed proteins and subsequently interact with a high number of T cells expressing particular T cell receptor Vβ chains. Although on mature T cells, SAgs usually trigger massive cell proliferation producing deleterious effects on the organism, in contrast, on immature T cells, they may trigger their death by apoptosis. On this basis, it was hypothesized that SAgs could also induce apoptosis in neoplastic T cells that are usually immature cells that probably conserve their particular Vβ chains. In this work, we investigated the effect of the SAg Staphylococcus aureus enterotoxin E (SEE) (that specifically interacts with cells that express Vβ8 chain), on human Jurkat T- leukemia line, that expresses Vβ8 in its T receptor and it is a model of the highly aggressive recurrent T-ALL. Our results demonstrated that SEE could induce apoptosis in Jurkat cells in vitro. The induction of apoptosis was specific, correlated to the down regulation of surface Vβ8 TCR expression and was triggered, at least in part, through the Fas/FasL extrinsic pathway. The apoptotic effect induced by SEE on Jurkat cells was therapeutically relevant. In effect, upon transplantation of Jurkat cells in the highly immunodeficient NSG mice, SEE treatment reduced dramatically tumor growth, decreased the infiltration of neoplastic cells in the bloodstream, spleen and lymph nodes and, most importantly, increased significantly the survival of mice. Taken together, these results raise the possibility that this strategy can be, in the future, a useful option for the treatment of recurrent T-ALL.
Collapse
Affiliation(s)
- Alejandra Duarte
- Institute of Experimental Medicine, National Council of Scientific and Technical Research, National Medicine Academy (IMEX-CONICET-ANM), Buenos Aires, Argentina
- Fundación Héctor Alejandro (H.A.) Barceló, Instituto Universitario de Ciencias de la Salud, Buenos Aires, Argentina
| | - Daniela R. Montagna
- Institute of Experimental Medicine, National Council of Scientific and Technical Research, National Medicine Academy (IMEX-CONICET-ANM), Buenos Aires, Argentina
| | - Mercedes Pastorini
- Institute of Experimental Medicine, National Council of Scientific and Technical Research, National Medicine Academy (IMEX-CONICET-ANM), Buenos Aires, Argentina
| | - Mercedes Alemán
- Institute of Experimental Medicine, National Council of Scientific and Technical Research, National Medicine Academy (IMEX-CONICET-ANM), Buenos Aires, Argentina
| |
Collapse
|
14
|
Francis D, Bhairaddy A, Joy A, Hari GV, Francis A. Secretory proteins in the orchestration of microbial virulence: The curious case of Staphylococcus aureus. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 133:271-350. [PMID: 36707204 DOI: 10.1016/bs.apcsb.2022.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Microbial virulence showcases an excellent model for adaptive changes that enable an organism to survive and proliferate in a hostile environment and exploit host resources to its own benefit. In Staphylococcus aureus, an opportunistic pathogen of the human host, known for the diversity of the disease conditions it inflicts and the rapid evolution of antibiotic resistance, virulence is a consequence of having a highly plastic genome that is amenable to quick reprogramming and the ability to express a diverse arsenal of virulence factors. Virulence factors that are secreted to the host milieu effectively manipulate the host conditions to favor bacterial survival and growth. They assist in colonization, nutrient acquisition, immune evasion, and systemic spread. The structural and functional characteristics of the secreted virulence proteins have been shaped to assist S. aureus in thriving and disseminating effectively within the host environment and exploiting the host resources to its best benefit. With the aim of highlighting the importance of secreted virulence proteins in bacterial virulence, the present chapter provides a comprehensive account of the role of the major secreted proteins of S. aureus in orchestrating its virulence in the human host.
Collapse
Affiliation(s)
- Dileep Francis
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India.
| | - Anusha Bhairaddy
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India
| | - Atheene Joy
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India
| | | | - Ashik Francis
- Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| |
Collapse
|
15
|
Jang JH, Kim S, Kim SG, Lee J, Lee DG, Jang J, Jeong YS, Song DH, Min JK, Park JG, Lee MS, Han BS, Son JS, Lee J, Lee NK. A Sensitive Immunodetection Assay Using Antibodies Specific to Staphylococcal Enterotoxin B Produced by Baculovirus Expression. BIOSENSORS 2022; 12:bios12100787. [PMID: 36290925 PMCID: PMC9599101 DOI: 10.3390/bios12100787] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/30/2022]
Abstract
Staphylococcal enterotoxin B (SEB) is a potent bacterial toxin that causes inflammatory stimulation and toxic shock, thus it is necessary to detect SEB in food and environmental samples. Here, we developed a sensitive immunodetection system using monoclonal antibodies (mAbs). Our study is the first to employ a baculovirus expression vector system (BEVS) to produce recombinant wild-type SEB. BEVS facilitated high-quantity and pure SEB production from suspension-cultured insect cells, and the SEB produced was characterized by mass spectrometry analysis. The SEB was stable at 4 °C for at least 2 years, maintaining its purity, and was further utilized for mouse immunization to generate mAbs. An optimal pair of mAbs non-competitive to SEB was selected for sandwich enzyme-linked immunosorbent assay-based immunodetection. The limit of detection of the immunodetection method was 0.38 ng/mL. Moreover, it displayed higher sensitivity in detecting SEB than commercially available immunodetection kits and retained detectability in various matrices and S. aureus culture supernatants. Thus, the results indicate that BEVS is useful for producing pure recombinant SEB with its natural immunogenic property in high yield, and that the developed immunodetection assay is reliable and sensitive for routine identification of SEB in various samples, including foods.
Collapse
Affiliation(s)
- Ju-Hong Jang
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Department of Biomolecular Science, Korea Research Institute of Bioscience and Biotechnology, School of Bioscience, Korea University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
| | - Sungsik Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Seul-Gi Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Department of Biomolecular Science, Korea Research Institute of Bioscience and Biotechnology, School of Bioscience, Korea University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
| | - Jaemin Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Dong-Gwang Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Jieun Jang
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Department of Biomolecular Science, Korea Research Institute of Bioscience and Biotechnology, School of Bioscience, Korea University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
| | - Young-Su Jeong
- Agency for Defense Development, 488 Bugyuseoung-daero, Daejeon 34060, Korea
| | - Dong-Hyun Song
- Agency for Defense Development, 488 Bugyuseoung-daero, Daejeon 34060, Korea
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Department of Biomolecular Science, Korea Research Institute of Bioscience and Biotechnology, School of Bioscience, Korea University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
| | - Jong-Gil Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Moo-Seung Lee
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Baek-Soo Han
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Jee-Soo Son
- iNtRON Biotechnology, 137 Sagimakgol-ro, Jungwon-gu, Seongnam-si 13202, Korea
| | - Jangwook Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Department of Biomolecular Science, Korea Research Institute of Bioscience and Biotechnology, School of Bioscience, Korea University of Science and Technology, 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Korea
- Correspondence: (J.L.); (N.-K.L.); Tel.: +82-42-860-4123 (J.L.); +82-42-860-4117 (N.-K.L.)
| | - Nam-Kyung Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea
- Correspondence: (J.L.); (N.-K.L.); Tel.: +82-42-860-4123 (J.L.); +82-42-860-4117 (N.-K.L.)
| |
Collapse
|
16
|
Manipulating Microbiota to Treat Atopic Dermatitis: Functions and Therapies. Pathogens 2022; 11:pathogens11060642. [PMID: 35745496 PMCID: PMC9228373 DOI: 10.3390/pathogens11060642] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 12/13/2022] Open
Abstract
Atopic dermatitis (AD) is a globally prevalent skin inflammation with a particular impact on children. Current therapies for AD are challenged by the limited armamentarium and the high heterogeneity of the disease. A novel promising therapeutic target for AD is the microbiota. Numerous studies have highlighted the involvement of the skin and gut microbiota in the pathogenesis of AD. The resident microbiota at these two epithelial tissues can modulate skin barrier functions and host immune responses, thus regulating AD progression. For example, the pathogenic roles of Staphylococcus aureus in the skin are well-established, making this bacterium an attractive target for AD treatment. Targeting the gut microbiota is another therapeutic strategy for AD. Multiple oral supplements with prebiotics, probiotics, postbiotics, and synbiotics have demonstrated promising efficacy in both AD prevention and treatment. In this review, we summarize the association of microbiota dysbiosis in both the skin and gut with AD, and the current knowledge of the functions of commensal microbiota in AD pathogenesis. Furthermore, we discuss the existing therapies in manipulating both the skin and gut commensal microbiota to prevent or treat AD. We also propose potential novel therapies based on the cutting-edge progress in this area.
Collapse
|
17
|
Noorbakhsh S, Ashouri S, Moradkhani M. Role of Superantigens In Various Childhood Inflamatory Diseases. Infect Disord Drug Targets 2022; 22:IDDT-EPUB-124062. [PMID: 35638540 DOI: 10.2174/1871526522666220530141031] [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: 10/20/2022] [Revised: 01/14/2022] [Accepted: 01/28/2022] [Indexed: 06/15/2023]
Abstract
----:: Super antigens (Sags) are some part of virus or bacteria proteins which stimulate T cells and antigen-presenting cells leading to systemic immune repose and inflammation. ---SAgs might have possible role in in various inflammatory childhood diseases (eg Kawasaki disease, atopic dermatitis, and chronic rhinosinusitis). ----Worldwide studies had done to determine the role of staphylococcal SAgs (TSST-1 ) in various inflammatory diseases. The SAgs (TSST-1) not only induce sepsis and septic shock (even in negative blood culture for S.aureus) ,but also might have significant role in various childhood inflammatory diseases ( eg KD, OMS, Polyp, dermatitis, psoriasis ) . In proven SAgs induced inflammatory diseases, inhibition the cell-destructive process by SAgs suppressants might be helpful. ----In toxic shock or sepsis like presentation even in cases with negative blood cultures immediate use pf anti staphylococcal drugs is required. ---Occasionaly, clinical presentation of some human viruses (eg coronavirus and adenovirus) mimic KD. ----- In addition ,coinfection with adenovirus, coronavirus, and para-influenza virus type 3 were observed with KD. -- Bacterial Sags induced increasement of acute-phase reactants and number of white blood cell, and neutrophil counts In developed KD. ----Multisystem inflammatory syndrome in children (MISC) and KS observed during the recent COVID-19 pandemia. This study summarized the relation between viral and bacterial SAgs and childhood inflammatory diseases.
Collapse
Affiliation(s)
- Samileh Noorbakhsh
- Pediatric Infectious Diseases department, Iran University of Medical Sciences, Tehran, Iran
| | - Sarvenaz Ashouri
- ENT and Head and Neck Research center and department, Iran university of medical sciences, Tehran, Iran
| | - Masoumeh Moradkhani
- Pediatric Resident ,Rasoul Hospital ,Iran university of medical sciences, Tehran, Iran
| |
Collapse
|
18
|
Massicotte-Azarniouch D, Herrera CA, Jennette JC, Falk RJ, Free ME. Mechanisms of vascular damage in ANCA vasculitis. Semin Immunopathol 2022; 44:325-345. [PMID: 35254509 PMCID: PMC9064952 DOI: 10.1007/s00281-022-00920-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
Abstract
The discovery of anti-neutrophil cytoplasmic antibodies (ANCA) and their antigenic targets, myeloperoxidase (MPO) and proteinase 3 (PR3), has led to further understanding as to the pathophysiologic processes that underlie vascular and tissue damage in ANCA vasculitis. ANCA trigger neutrophil activation leading to vascular damage in ANCA vasculitis. However, decades of study have determined that neutrophil activation alone is not sufficient to cause disease. Inflammatory stimuli are drivers of ANCA autoantigen expression and ANCA production. Certain infections or bacterial peptides may be crucial players in the initial steps of ANCA immunopathogenesis. Genetic and epigenetic alterations of gene encoding for MPO and PR3 provide additional disturbances to the immune homeostasis which provide a substrate for pathogenic ANCA formation from an adaptive immune system predisposed to autoreactivity. Promoted by inflammatory cytokines, ANCA binding leads to neutrophil activation, a process characterized by conformational changes, production and release of cytotoxic substances, and alternative complement pathway activation, thus creating an intense inflammatory milieu. This cascade of events perpetuates a vicious cycle of further inflammatory cell recruitment and activation, culminating in tissue necrosis. Our understanding of the pathogenic process in ANCA vasculitis paves the way for the development of therapies targeting crucial steps in this process. The greater appreciation of the role for complement, monocytes, and the adaptive immune system has already led to novel complement blockers and is poised to lead to further innovations which will allow for tailored antigen- or cell-specific immunotherapy targeting the autoimmune process without exposure to undue risks or toxicities.
Collapse
Affiliation(s)
- David Massicotte-Azarniouch
- Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Carolina A Herrera
- Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - J Charles Jennette
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ronald J Falk
- Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Meghan E Free
- Division of Nephrology and Hypertension, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
19
|
Gan SKE, Derrick JP, Fraternali F. Editorial: Understanding and Engineering Antibody-Superantigen Interactions. Front Immunol 2022; 13:857339. [PMID: 35222446 PMCID: PMC8865624 DOI: 10.3389/fimmu.2022.857339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 01/24/2022] [Indexed: 12/04/2022] Open
Affiliation(s)
- Samuel Ken-En Gan
- Antibody & Product Development (APD) Lab, Agency for Science, Technology, and Research (ASTAR), Singapore, Singapore.,APD SKEG Pte Ltd, Singapore, Singapore.,James Cook University, Singapore, Singapore
| | - Jeremy P Derrick
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
| | - Franca Fraternali
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom.,The Francis Crick Institute, London, United Kingdom.,The Thomas Young Centre for Theory and Simulation of Materials, London, United Kingdom
| |
Collapse
|
20
|
Tabiś A, Gonet M, Schubert J, Miazek A, Nowak M, Tomaszek A, Bania J. Analysis of enterotoxigenic effect of Staphylococcus aureus and Staphylococcus epidermidis enterotoxins C and L on mice. Microbiol Res 2022; 258:126979. [DOI: 10.1016/j.micres.2022.126979] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/03/2022] [Accepted: 02/03/2022] [Indexed: 01/31/2023]
|
21
|
Baz AA, Bakhiet EK, Abdul-Raouf U, Abdelkhalek A. Prevalence of enterotoxin genes (SEA to SEE) and antibacterial resistant pattern of Staphylococcus aureus isolated from clinical specimens in Assiut city of Egypt. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-021-00199-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Infections in communities and hospitals are mostly caused by Staphylococcus aureus strains. This study aimed to determine the prevalence of five genes (SEA, SEB, SEC, SED and SEE) encoding staphylococcal enterotoxins in S. aureus isolates from various clinical specimens, as well as to assess the relationship of these isolates with antibiotic susceptibility. Traditional PCR was used to detect enterotoxin genes, and the ability of isolates expressing these genes was determined using Q.RT-PCR.
Results
Overall; 61.3% (n = 46) of the samples were positive for S. aureus out of 75 clinical specimens, including urine, abscess, wounds, and nasal swabs. The prevalence of antibiotic resistance showed S. aureus isolates were resistant to Nalidixic acid, Ampicillin and Amoxicillin (100%), Cefuroxime (94%), Ceftriaxone (89%), Ciprofloxacin (87%), Erythromycin and Ceftaxime (85%), Cephalexin and Clarithromycin (83%), Cefaclor (81%), Gentamicin (74%), Ofloxacin (72%), Chloramphenicol(59%), Amoxicillin/Clavulanic acid (54%), while all isolates sensitive to Imipinem (100%). By employing specific PCR, about 39.1% of isolates were harbored enterotoxin genes, enterotoxin A was the most predominant toxin in 32.6% of isolates, enterotoxin B with 4.3% of isolates and enterotoxin A and B were detected jointly in 2.1% of isolates, while enterotoxin C, D and E weren’t detected in any isolate.
Conclusion
This study revealed a high prevalence of S. aureus among clinical specimens. The isolates were also multidrug resistant to several tested antibiotics. Enterotoxin A was the most prevalent gene among isolates. The presence of antibiotic resistance and enterotoxin genes may facilitate the spread of S. aureus strains and pose a potential threat to public health.
Collapse
|
22
|
Borowska MT, Drees C, Yarawsky AE, Viswanathan M, Ryan SM, Bunker JJ, Herr AB, Bendelac A, Adams EJ. The molecular characterization of antibody binding to a superantigen-like protein from a commensal microbe. Proc Natl Acad Sci U S A 2021; 118:e2023898118. [PMID: 34548394 PMCID: PMC8488583 DOI: 10.1073/pnas.2023898118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2021] [Indexed: 11/18/2022] Open
Abstract
Microorganisms have coevolved diverse mechanisms to impair host defenses. A major one, superantigens, can result in devastating effects on the immune system. While all known superantigens induce vast immune cell proliferation and come from opportunistic pathogens, recently, proteins with similar broad specificity to antibody variable (V) domain families were identified in a commensal microbiota. These proteins, identified in the human commensal Ruminococcus gnavus, are called immunoglobulin-binding protein (Ibp) A and B and have been shown to activate B cells in vitro expressing either human VH3 or murine VH5/6/7. Here, we provide molecular and functional studies revealing the basis of this Ibp/immunoglobulin (Ig) interaction. The crystal structure and biochemical assays of a truncated IbpA construct in complex with mouse VH5 antigen-binding fragment (Fab) shows a binding of Ig heavy chain framework residues to the Ibp Domain D and the C-terminal heavy chain binding domain (HCBD). We used targeted mutagenesis of contact residues and affinity measurements and performed studies of the Fab-IbpA complex to determine the stoichiometry between Ibp and VH domains, suggesting Ibp may serve to cluster full-length IgA antibodies in vivo. Furthermore, in vitro stimulation experiments indicate that binding of the Ibp HCBD alone is sufficient to activate responsive murine B cell receptors. The presence of these proteins in a commensal microbe suggest that binding a broad repertoire of immunoglobulins, particularly in the gut/microbiome environment, may provide an important function in the maintenance of host/microbiome homeostasis contrasting with the pathogenic role of structurally homologous superantigens expressed by pathogens.
Collapse
Affiliation(s)
- Marta T Borowska
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637
| | - Christoph Drees
- Department of Pathology, University of Chicago, Chicago, IL 60637
| | - Alexander E Yarawsky
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | | | - Sean M Ryan
- Committee on Immunology, University of Chicago, Chicago, IL 60637
| | - Jeffrey J Bunker
- Department of Pathology, University of Chicago, Chicago, IL 60637
- Committee on Immunology, University of Chicago, Chicago, IL 60637
| | - Andrew B Herr
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | - Albert Bendelac
- Department of Pathology, University of Chicago, Chicago, IL 60637;
- Committee on Immunology, University of Chicago, Chicago, IL 60637
| | - Erin J Adams
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637;
- Committee on Immunology, University of Chicago, Chicago, IL 60637
| |
Collapse
|
23
|
Deacy AM, Gan SKE, Derrick JP. Superantigen Recognition and Interactions: Functions, Mechanisms and Applications. Front Immunol 2021; 12:731845. [PMID: 34616400 PMCID: PMC8488440 DOI: 10.3389/fimmu.2021.731845] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/30/2021] [Indexed: 12/27/2022] Open
Abstract
Superantigens are unconventional antigens which recognise immune receptors outside their usual recognition sites e.g. complementary determining regions (CDRs), to elicit a response within the target cell. T-cell superantigens crosslink T-cell receptors and MHC Class II molecules on antigen-presenting cells, leading to lymphocyte recruitment, induction of cytokine storms and T-cell anergy or apoptosis among many other effects. B-cell superantigens, on the other hand, bind immunoglobulins on B-cells, affecting opsonisation, IgG-mediated phagocytosis, and driving apoptosis. Here, through a review of the structural basis for recognition of immune receptors by superantigens, we show that their binding interfaces share specific physicochemical characteristics when compared with other protein-protein interaction complexes. Given that antibody-binding superantigens have been exploited extensively in industrial antibody purification, these observations could facilitate further protein engineering to optimize the use of superantigens in this and other areas of biotechnology.
Collapse
Affiliation(s)
- Anthony M. Deacy
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
| | - Samuel Ken-En Gan
- Antibody & Product Development Lab, Experimental Drug Development Centre – Bioinformatics Institute (EDDC-BII), Agency for Science Technology and Research (ASTAR), Singapore, Singapore
- James Cook University, Singapore, Singapore
| | - Jeremy P. Derrick
- School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
24
|
Liu D, Van Belleghem JD, de Vries CR, Burgener E, Chen Q, Manasherob R, Aronson JR, Amanatullah DF, Tamma PD, Suh GA. The Safety and Toxicity of Phage Therapy: A Review of Animal and Clinical Studies. Viruses 2021; 13:1268. [PMID: 34209836 PMCID: PMC8310247 DOI: 10.3390/v13071268] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/23/2021] [Accepted: 06/23/2021] [Indexed: 12/13/2022] Open
Abstract
Increasing rates of infection by antibiotic resistant bacteria have led to a resurgence of interest in bacteriophage (phage) therapy. Several phage therapy studies in animals and humans have been completed over the last two decades. We conducted a systematic review of safety and toxicity data associated with phage therapy in both animals and humans reported in English language publications from 2008-2021. Overall, 69 publications met our eligibility criteria including 20 animal studies, 35 clinical case reports or case series, and 14 clinical trials. After summarizing safety and toxicity data from these publications, we discuss potential approaches to optimize safety and toxicity monitoring with the therapeutic use of phage moving forward. In our systematic review of the literature, we found some adverse events associated with phage therapy, but serious events were extremely rare. Comprehensive and standardized reporting of potential toxicities associated with phage therapy has generally been lacking in the published literature. Structured safety and tolerability endpoints are necessary when phages are administered as anti-infective therapeutics.
Collapse
Affiliation(s)
- Dan Liu
- Department of Burn, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China;
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (J.D.V.B.); (C.R.d.V.); (Q.C.); (J.R.A.)
| | - Jonas D. Van Belleghem
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (J.D.V.B.); (C.R.d.V.); (Q.C.); (J.R.A.)
| | - Christiaan R. de Vries
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (J.D.V.B.); (C.R.d.V.); (Q.C.); (J.R.A.)
| | - Elizabeth Burgener
- Center for Excellence in Pulmonary Biology, Department of Pediatrics, Stanford University, Stanford, CA 94305, USA;
| | - Qingquan Chen
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (J.D.V.B.); (C.R.d.V.); (Q.C.); (J.R.A.)
| | - Robert Manasherob
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.M.); (D.F.A.)
| | - Jenny R. Aronson
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (J.D.V.B.); (C.R.d.V.); (Q.C.); (J.R.A.)
| | - Derek F. Amanatullah
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.M.); (D.F.A.)
| | - Pranita D. Tamma
- Division of Pediatric Infectious Diseases, Department of Pediatrics, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA;
| | - Gina A. Suh
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
25
|
Gao SX, Sun C, Zhu YT, Zhao JB, Sun J, Zhou P, Jiang HY, Fan YA, Wei L, Zhang T, Guan JC. Exposure of pregnant rats to staphylococcal enterotoxin B increases offspring splenic Treg number and function via decreasing FoxP3 methylation. Immunobiology 2021; 226:152060. [PMID: 33529803 DOI: 10.1016/j.imbio.2021.152060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/12/2021] [Accepted: 01/20/2021] [Indexed: 12/30/2022]
Abstract
Staphylococcus aureus is an infectious pathogen that is relatively common, but that can cause severe disease in pregnant women and their fetus. We previously demonstrated that exposing pregnant rats to staphylococcal enterotoxin B (SEB) altered splenic CD4/CD8 T cell frequencies in their offspring. Whether prenatal SEB exposure impacts Tregs in these offspring, however, remains to be determined. As such, in this study, we intravenously injected pregnant rats with 15 μg of SEB on gestational day 16. Splenic tissue was then harvested from 1-, 3-, and 5-day-old neonatal rats and analyzed via flow cytometry to assess Treg numbers. In addition, FoxP3 expression levels were assessed via qPCR and western blotting, while FoxP3 methylation status was evaluated via methyl-DNA immunoprecipitation qPCR. Immunosuppression assays were additionally used to gauge Treg suppressive functionality. We found that exposing pregnant rats to SEB resulted in a significant increase in Treg numbers, FoxP3 expression, and Treg suppressive capacity in the spleens of both neonatal and adult offspring. In addition, total T cell, CD4+T cell, and non-Treg CD4+ T cell numbers were elevated in the spleens of offspring following prenatal SEB exposure. We additionally determined that SEB exposure resulted in a significant reduction in FoxP3 DNA methylation. Together, our results indicate that prenatal SEB exposure can markedly enhance offspring splenic Treg numbers and functionality at least in part by decreasing FoxP3 methylation.
Collapse
Affiliation(s)
- Shu-Xian Gao
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, PR China
| | - Chao Sun
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, PR China
| | - Yu-Ting Zhu
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, PR China
| | - Jia-Bao Zhao
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, PR China
| | - Jing Sun
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, PR China
| | - Ping Zhou
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, PR China
| | - Hao-Yuan Jiang
- An Under Graduate Student Majored Clinical Medicine, Bengbu Medical College, Anhui 233030, PR China
| | - Ying-Ao Fan
- An Under Graduate Student Majored Clinical Medicine, Bengbu Medical College, Anhui 233030, PR China
| | - Li Wei
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, PR China
| | - Tao Zhang
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, PR China
| | - Jun-Chang Guan
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui 233030, PR China; Department of Microbiology, Bengbu Medical College, Bengbu, Anhui 233030, PR China.
| |
Collapse
|
26
|
Venkatasubramaniam A, Liao G, Cho E, Adhikari RP, Kort T, Holtsberg FW, Elsass KE, Kobs DJ, Rudge TL, Kauffman KD, Lora NE, Barber DL, Aman MJ, Karauzum H. Safety and Immunogenicity of a 4-Component Toxoid-Based Staphylococcus aureus Vaccine in Rhesus Macaques. Front Immunol 2021; 12:621754. [PMID: 33717122 PMCID: PMC7947289 DOI: 10.3389/fimmu.2021.621754] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/19/2021] [Indexed: 12/17/2022] Open
Abstract
Staphylococcus aureus is a leading cause of significant morbidity and mortality and an enormous economic burden to public health worldwide. Infections caused by methicillin-resistant S. aureus (MRSA) pose a major threat as MRSA strains are becoming increasingly prevalent and multi-drug resistant. To this date, vaccines targeting surface-bound antigens demonstrated promising results in preclinical testing but have failed in clinical trials. S. aureus pathogenesis is in large part driven by immune destructive and immune modulating toxins and thus represent promising vaccine targets. Hence, the objective of this study was to evaluate the safety and immunogenicity of a staphylococcal 4-component vaccine targeting secreted bi-component pore-forming toxins (BCPFTs) and superantigens (SAgs) in non-human primates (NHPs). The 4-component vaccine proved to be safe, even when repeated vaccinations were given at a dose that is 5 to 10- fold higher than the proposed human dose. Vaccinated rhesus macaques did not exhibit clinical signs, weight loss, or changes in hematology or serum chemistry parameters related to the administration of the vaccine. No acute, vaccine-related elevation of serum cytokine levels was observed after vaccine administration, confirming the toxoid components lacked superantigenicity. Immunized animals demonstrated high level of toxin-specific total and neutralizing antibodies toward target antigens of the 4-component vaccine as well as cross-neutralizing activity toward staphylococcal BCPFTs and SAgs that are not direct targets of the vaccine. Cross-neutralization was also observed toward the heterologous streptococcal pyogenic exotoxin B. Ex vivo stimulation of PBMCs with individual vaccine components demonstrated an overall increase in several T cell cytokines measured in supernatants. Immunophenotyping of CD4 T cells ex vivo showed an increase in Ag-specific polyfunctional CD4 T cells in response to antigen stimulation. Taken together, we demonstrate that the 4-component vaccine is well-tolerated and immunogenic in NHPs generating both humoral and cellular immune responses. Targeting secreted toxin antigens could be the next-generation vaccine approach for staphylococcal vaccines if also proven to provide efficacy in humans.
Collapse
Affiliation(s)
| | - Grant Liao
- Integrated BioTherapeutics, Rockville, MD, United States
| | - Eunice Cho
- Integrated BioTherapeutics, Rockville, MD, United States
| | | | - Tom Kort
- Integrated BioTherapeutics, Rockville, MD, United States
| | | | | | - Dean J. Kobs
- Batelle - West Jefferson, West Jefferson, OH, United States
| | | | - Keith D. Kauffman
- Laboratory of Parasitic Diseases, T Lymphocyte Biology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Nickiana E. Lora
- Laboratory of Parasitic Diseases, T Lymphocyte Biology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Daniel L. Barber
- Laboratory of Parasitic Diseases, T Lymphocyte Biology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - M. Javad Aman
- Integrated BioTherapeutics, Rockville, MD, United States
| | | |
Collapse
|
27
|
Riahi Rad Z, Riahi Rad Z, Goudarzi H, Goudarzi M, Mahmoudi M, Yasbolaghi Sharahi J, Hashemi A. MicroRNAs in the interaction between host-bacterial pathogens: A new perspective. J Cell Physiol 2021; 236:6249-6270. [PMID: 33599300 DOI: 10.1002/jcp.30333] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/17/2022]
Abstract
Gene expression regulation plays a critical role in host-pathogen interactions, and RNAs function is essential in this process. miRNAs are small noncoding, endogenous RNA fragments that affect stability and/or translation of mRNAs, act as major posttranscriptional regulators of gene expression. miRNA is involved in regulating many biological or pathological processes through targeting specific mRNAs, including development, differentiation, apoptosis, cell cycle, cytoskeleton organization, and autophagy. Deregulated microRNA expression is associated with many types of diseases, including cancers, immune disturbances, and infection. miRNAs are a vital section of the host immune response to bacterial-made infection. Bacterial pathogens suppress host miRNA expression for their benefit, promoting survival, replication, and persistence. The role played through miRNAs in interaction with host-bacterial pathogen has been extensively studied in the past 10 years, and knowledge about these staggering molecules' function can clarify the complicated and ambiguous interactions of the host-bacterial pathogen. Here, we review how pathogens prevent the host miRNA expression. We briefly discuss emerging themes in this field, including their role as biomarkers in identifying bacterial infections, as part of the gut microbiota, on host miRNA expression.
Collapse
Affiliation(s)
- Zohreh Riahi Rad
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Riahi Rad
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahmoudi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Javad Yasbolaghi Sharahi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Hashemi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Systems Genetics Approaches in Mouse Models of Group A Streptococcal Necrotizing Soft-Tissue Infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 33079368 DOI: 10.1007/978-3-030-57616-5_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Mouse models are invaluable resources for studying the pathogenesis and preclinical evaluation of therapeutics and vaccines against many human pathogens. Infections caused by group A streptococcus (GAS, Streptococcus pyogenes) are heterogeneous ranging from mild pharyngitis to severe invasive necrotizing fasciitis, a subgroup of necrotizing soft-tissue infections (NSTIs). While several strains of mice including BALB/c, C3H/HeN, CBA/J, and C57BL/10 offered significant insights, the human specificity and the interindividual variations on susceptibility or resistance to GAS infections limit their ability to mirror responses as seen in humans. In this chapter, we discuss the advanced recombinant inbred (ARI) BXD mouse model that mimics the genetic diversity as seen in humans and underpins the feasibility to map multiple genes (genetic loci) modulating GAS NSTI. GAS produces a myriad of virulence factors, including superantigens (SAg). Superantigens are potent immune toxins that activate T cells by cross-linking T cell receptors with human leukocyte antigen class-II (HLA-II) molecules expressed on antigen-presenting cells. This leads to a pro-inflammatory cytokine storm and the subsequent multiple organ damage and shock. Inbred mice are innately refractive to SAg-mediated responses. In this chapter, we discuss the versatility of the HLA-II transgenic mouse model that allowed the biological validation of known genetic associations to GAS NSTI. The combined utility of ARI-BXD and HLA-II mice as complementary approaches that offer clinically translatable insights into pathomechanisms driven by complex traits and host genetic context and novel means to evaluate the in vivo efficiency of therapies to improve outcomes of GAS NSTI are also discussed.
Collapse
|
29
|
Felce JH, Parolini L, Sezgin E, Céspedes PF, Korobchevskaya K, Jones M, Peng Y, Dong T, Fritzsche M, Aarts D, Frater J, Dustin ML. Single-Molecule, Super-Resolution, and Functional Analysis of G Protein-Coupled Receptor Behavior Within the T Cell Immunological Synapse. Front Cell Dev Biol 2021; 8:608484. [PMID: 33537301 PMCID: PMC7848080 DOI: 10.3389/fcell.2020.608484] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/21/2020] [Indexed: 12/31/2022] Open
Abstract
A central process in immunity is the activation of T cells through interaction of T cell receptors (TCRs) with agonistic peptide-major histocompatibility complexes (pMHC) on the surface of antigen presenting cells (APCs). TCR-pMHC binding triggers the formation of an extensive contact between the two cells termed the immunological synapse, which acts as a platform for integration of multiple signals determining cellular outcomes, including those from multiple co-stimulatory/inhibitory receptors. Contributors to this include a number of chemokine receptors, notably CXC-chemokine receptor 4 (CXCR4), and other members of the G protein-coupled receptor (GPCR) family. Although best characterized as mediators of ligand-dependent chemotaxis, some chemokine receptors are also recruited to the synapse and contribute to signaling in the absence of ligation. How these and other GPCRs integrate within the dynamic structure of the synapse is unknown, as is how their normally migratory Gαi-coupled signaling is terminated upon recruitment. Here, we report the spatiotemporal organization of several GPCRs, focusing on CXCR4, and the G protein Gαi2 within the synapse of primary human CD4+ T cells on supported lipid bilayers, using standard- and super-resolution fluorescence microscopy. We find that CXCR4 undergoes orchestrated phases of reorganization, culminating in recruitment to the TCR-enriched center. This appears to be dependent on CXCR4 ubiquitination, and does not involve stable interactions with TCR microclusters, as viewed at the nanoscale. Disruption of this process by mutation impairs CXCR4 contributions to cellular activation. Gαi2 undergoes active exclusion from the synapse, partitioning from centrally-accumulated CXCR4. Using a CRISPR-Cas9 knockout screen, we identify several diverse GPCRs with contributions to T cell activation, most significantly the sphingosine-1-phosphate receptor S1PR1, and the oxysterol receptor GPR183. These, and other GPCRs, undergo organization similar to CXCR4; including initial exclusion, centripetal transport, and lack of receptor-TCR interactions. These constitute the first observations of GPCR dynamics within the synapse, and give insights into how these receptors may contribute to T cell activation. The observation of broad GPCR contributions to T cell activation also opens the possibility that modulating GPCR expression in response to cell status or environment may directly regulate responsiveness to pMHC.
Collapse
Affiliation(s)
- James H Felce
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Lucia Parolini
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Erdinc Sezgin
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Pablo F Céspedes
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | | | - Mathew Jones
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Yanchun Peng
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Tao Dong
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom.,Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Marco Fritzsche
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom.,Rosalind Franklin Institute, Didcot, United Kingdom
| | - Dirk Aarts
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, Oxford, United Kingdom
| | - John Frater
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.,National Institute of Health Research Biomedical Research Centre, Oxford, United Kingdom
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
30
|
Aguilera-Lizarraga J, Florens MV, Viola MF, Jain P, Decraecker L, Appeltans I, Cuende-Estevez M, Fabre N, Van Beek K, Perna E, Balemans D, Stakenborg N, Theofanous S, Bosmans G, Mondelaers SU, Matteoli G, Ibiza Martínez S, Lopez-Lopez C, Jaramillo-Polanco J, Talavera K, Alpizar YA, Feyerabend TB, Rodewald HR, Farre R, Redegeld FA, Si J, Raes J, Breynaert C, Schrijvers R, Bosteels C, Lambrecht BN, Boyd SD, Hoh RA, Cabooter D, Nelis M, Augustijns P, Hendrix S, Strid J, Bisschops R, Reed DE, Vanner SJ, Denadai-Souza A, Wouters MM, Boeckxstaens GE. Local immune response to food antigens drives meal-induced abdominal pain. Nature 2021; 590:151-156. [PMID: 33442055 DOI: 10.1038/s41586-020-03118-2] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 11/27/2020] [Indexed: 12/19/2022]
Abstract
Up to 20% of people worldwide develop gastrointestinal symptoms following a meal1, leading to decreased quality of life, substantial morbidity and high medical costs. Although the interest of both the scientific and lay communities in this issue has increased markedly in recent years, with the worldwide introduction of gluten-free and other diets, the underlying mechanisms of food-induced abdominal complaints remain largely unknown. Here we show that a bacterial infection and bacterial toxins can trigger an immune response that leads to the production of dietary-antigen-specific IgE antibodies in mice, which are limited to the intestine. Following subsequent oral ingestion of the respective dietary antigen, an IgE- and mast-cell-dependent mechanism induced increased visceral pain. This aberrant pain signalling resulted from histamine receptor H1-mediated sensitization of visceral afferents. Moreover, injection of food antigens (gluten, wheat, soy and milk) into the rectosigmoid mucosa of patients with irritable bowel syndrome induced local oedema and mast cell activation. Our results identify and characterize a peripheral mechanism that underlies food-induced abdominal pain, thereby creating new possibilities for the treatment of irritable bowel syndrome and related abdominal pain disorders.
Collapse
Affiliation(s)
- Javier Aguilera-Lizarraga
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Morgane V Florens
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Maria Francesca Viola
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Piyush Jain
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Lisse Decraecker
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Iris Appeltans
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Maria Cuende-Estevez
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Naomi Fabre
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Kim Van Beek
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Eluisa Perna
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Dafne Balemans
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Nathalie Stakenborg
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Stavroula Theofanous
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Goele Bosmans
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Stéphanie U Mondelaers
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Gianluca Matteoli
- Laboratory for Mucosal Immunology, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Sales Ibiza Martínez
- Laboratory for Mucosal Immunology, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium.,Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Cintya Lopez-Lopez
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | | | - Karel Talavera
- Laboratory for Ion Channel Research, VIB Center for Brain and Disease Research, KU Leuven Department of Cellular and Molecular Medicine, Leuven, Belgium
| | - Yeranddy A Alpizar
- Neuroscience Research group, BIOMED, Hasselt University, Hasselt, Belgium
| | | | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Ricard Farre
- Mucosal Permeability Lab, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Frank A Redegeld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Jiyeon Si
- KU Leuven Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, Leuven, Belgium.,VIB KU Leuven Center for Microbiology, Leuven, Belgium
| | - Jeroen Raes
- KU Leuven Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, Leuven, Belgium.,VIB KU Leuven Center for Microbiology, Leuven, Belgium
| | - Christine Breynaert
- Allergy and Clinical Immunology Research Group, KU Leuven Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Rik Schrijvers
- Allergy and Clinical Immunology Research Group, KU Leuven Department of Microbiology, Immunology and Transplantation, Leuven, Belgium
| | - Cédric Bosteels
- Laboratory of Immunoregulation, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Scott D Boyd
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA, USA
| | - Ramona A Hoh
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Deirdre Cabooter
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Maxim Nelis
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Patrick Augustijns
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Sven Hendrix
- Department of Morphology, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.,Medical School Hamburg, Hamburg, Germany
| | - Jessica Strid
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Raf Bisschops
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - David E Reed
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Stephen J Vanner
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Alexandre Denadai-Souza
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Mira M Wouters
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium
| | - Guy E Boeckxstaens
- Laboratory for Intestinal Neuroimmune Interactions, Translational Research Center for Gastrointestinal Disorders, KU Leuven Department of Chronic Diseases, Metabolism and Ageing, Leuven, Belgium.
| |
Collapse
|
31
|
Bachert C, Marple B, Schlosser RJ, Hopkins C, Schleimer RP, Lambrecht BN, Bröker BM, Laidlaw T, Song WJ. Adult chronic rhinosinusitis. Nat Rev Dis Primers 2020; 6:86. [PMID: 33122665 DOI: 10.1038/s41572-020-00218-1] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/09/2020] [Indexed: 02/06/2023]
Abstract
Chronic rhinosinusitis (CRS) occurs in >10% of the adult population in Europe and the USA and can be differentiated into CRS without nasal polyps and CRS with nasal polyps (CRSwNP). Both phenotypes are characterized by a high disease burden and an overlapping spectrum of symptoms, with facial pain and loss of smell being the most differentiating. Great progress has been made in the understanding of CRS pathophysiology: from the epithelium and epithelial-mesenchymal transition to innate and adaptive immunity pathways and, finally, on the role of eosinophils and Staphylococcus aureus in the persistence of disease. Although clinical manifestations and diagnostic tools (including nasal endoscopy and imaging) have undergone major changes over the past few years, management (including pharmacotherapy, surgery and biologics) has experienced enormous progress based on the growing knowledge of key mediators in severe CRSwNP. The introduction of endotyping has led to a differentiation of 'tailored' surgical approaches, focusing on the mucosal concept in those with severe CRSwNP and on the identification of patients eligible for extended surgery and possibly biologics in the future.
Collapse
Affiliation(s)
- Claus Bachert
- Sun Yat-sen University, International Airway Research Center, First Affiliated Hospital, Guangzhou, China.
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium.
- Division of ENT diseases, CLINTEC, Karolinska Institute, University of Stockholm, Stockholm, Sweden.
| | - Bradley Marple
- University of Texas, Southwestern Medical Center, Department of Otolaryngology - Head and Neck Surgery, Dallas, TX, USA
| | - Rodney J Schlosser
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC, USA
| | | | - Robert P Schleimer
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bart N Lambrecht
- Laboratory of Immunoregulation, VIB-UGhent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Pulmonary Medicine, ErasmusMC, Rotterdam, Netherlands
| | - Barbara M Bröker
- Department of Immunology, Institute of Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Tanya Laidlaw
- Department of Medicine, Harvard Medical School, the Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Woo-Jung Song
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
32
|
Siggins MK, Lynskey NN, Lamb LE, Johnson LA, Huse KK, Pearson M, Banerji S, Turner CE, Woollard K, Jackson DG, Sriskandan S. Extracellular bacterial lymphatic metastasis drives Streptococcus pyogenes systemic infection. Nat Commun 2020; 11:4697. [PMID: 32943639 PMCID: PMC7498588 DOI: 10.1038/s41467-020-18454-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 08/24/2020] [Indexed: 12/15/2022] Open
Abstract
Unassisted metastasis through the lymphatic system is a mechanism of dissemination thus far ascribed only to cancer cells. Here, we report that Streptococcus pyogenes also hijack lymphatic vessels to escape a local infection site, transiting through sequential lymph nodes and efferent lymphatic vessels to enter the bloodstream. Contrasting with previously reported mechanisms of intracellular pathogen carriage by phagocytes, we show S. pyogenes remain extracellular during transit, first in afferent and then efferent lymphatics that carry the bacteria through successive draining lymph nodes. We identify streptococcal virulence mechanisms important for bacterial lymphatic dissemination and show that metastatic streptococci within infected lymph nodes resist and subvert clearance by phagocytes, enabling replication that can seed intense bloodstream infection. The findings establish the lymphatic system as both a survival niche and conduit to the bloodstream for S. pyogenes, explaining the phenomenon of occult bacteraemia. This work provides new perspectives in streptococcal pathogenesis with implications for immunity. Pathogenic agents can spread from an initial to a secondary site via the lymphatics. Here, using a mouse model of infection, the authors show that S. pyogenes readily transit through sequential lymph nodes within efferent lymphatics to reach the bloodstream and drive systemic infection, while remaining extracellular.
Collapse
Affiliation(s)
- Matthew K Siggins
- Department of Infectious Disease, Imperial College London, London, W12 0NN, UK. .,MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, SW7 2DD, UK. .,NLHI, Imperial College London, London, W2 1PG, UK.
| | - Nicola N Lynskey
- Department of Infectious Disease, Imperial College London, London, W12 0NN, UK.,MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, SW7 2DD, UK.,The Roslin Institute, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Lucy E Lamb
- Department of Infectious Disease, Imperial College London, London, W12 0NN, UK
| | - Louise A Johnson
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Kristin K Huse
- Department of Infectious Disease, Imperial College London, London, W12 0NN, UK.,MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, SW7 2DD, UK
| | - Max Pearson
- Department of Infectious Disease, Imperial College London, London, W12 0NN, UK.,MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, SW7 2DD, UK
| | - Suneale Banerji
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Claire E Turner
- Department of Infectious Disease, Imperial College London, London, W12 0NN, UK.,The Florey Institute, University of Sheffield, Sheffield, S10 2TN, UK
| | - Kevin Woollard
- Centre for Inflammatory Disease, Department of Immunology & Inflammation, Imperial College London, London, W12 0NN, UK
| | - David G Jackson
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Shiranee Sriskandan
- Department of Infectious Disease, Imperial College London, London, W12 0NN, UK. .,MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, SW7 2DD, UK.
| |
Collapse
|
33
|
Chajęcka-Wierzchowska W, Gajewska J, Wiśniewski P, Zadernowska A. Enterotoxigenic Potential of Coagulase-Negative Staphylococci from Ready-to-Eat Food. Pathogens 2020; 9:pathogens9090734. [PMID: 32899905 PMCID: PMC7559265 DOI: 10.3390/pathogens9090734] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/01/2020] [Accepted: 09/04/2020] [Indexed: 11/16/2022] Open
Abstract
Although coagulase-positive staphylococci are considered to be the main factor responsible for food poisoning, an increasing role for the coagulase-negative staphylococci in the production of enterotoxins has been observed in recent years. This study was conducted to assess the occurrence of genes responsible for the production of staphylococcal enterotoxins (SE), enterotoxin-like toxins (SEI) and toxic shock syndrome toxin-1 (TSST-1) in coagulase-negative staphylococci (CoNS) isolated from ready-to-eat food from bars and restaurants. One hundred and eighteen CoNS strains were tested using polymerase chain reaction (PCR) to five superantigenic toxin genes, including five different types of classical enterotoxins (sea, seb, sec, sed and see) and the toxic shock syndrome toxin-1 (tsst-1) as well as to supertoxin-like genes. PCR-positive isolates were then tested using immunoenzymatic methods (SET-RPLA, Vidas SET 2) for toxin expression. Out of 118 CoNS strains, the presence of staphylococcal enterotoxins was confirmed in 72% of them. The most frequently found enterotoxin-like genotype was ser, selu. Two of the tested strains had up to ten different enterotoxin genes in the genome at the same time. Although no production of enterotoxins was detected in the CoNS, which means that their possible role in the epidemiology of food-borne diseases is minimal, the data demonstrated that the toxigenic capacity of the CoNS should not be ignored, and that this group of microorganisms should be continuously monitored in food.
Collapse
|
34
|
SpeS: A Novel Superantigen and Its Potential as a Vaccine Adjuvant against Strangles. Int J Mol Sci 2020; 21:ijms21124467. [PMID: 32586031 PMCID: PMC7352279 DOI: 10.3390/ijms21124467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 11/16/2022] Open
Abstract
Bacterial superantigens (sAgs) are powerful activators of the immune response that trigger unspecific T cell responses accompanied by the release of proinflammatory cytokines. Streptococcus equi (S. equi) and Streptococcus zooepidemicus (S. zooepidemicus) produce sAgs that play an important role in their ability to cause disease. Strangles, caused by S. equi, is one of the most common infectious diseases of horses worldwide. Here, we report the identification of a new sAg of S. zooepidemicus, SpeS, and show that mutation of the putative T cell receptor (TCR)-binding motif (YAY to IAY) abrogated TCR-binding, whilst maintaining interaction with major histocompatibility complex (MHC) class II molecules. The fusion of SpeS and SpeSY39I to six S. equi surface proteins using two different peptide linkers was conducted to determine if MHC class II-binding properties were maintained. Proliferation assays, qPCR and flow cytometry analysis showed that SpeSY39I and its fusion proteins induced less mitogenic activity and interferon gamma expression when compared to SpeS, whilst retaining Antigen-Presenting Cell (APC)-binding properties. Our data suggest that SpeSY39I-surface protein fusions could be used to direct vaccine antigens towards antigen-presenting cells in vivo with the potential to enhance antigen presentation and improve immune responses.
Collapse
|
35
|
Altun M, Mericli Yapıcı B. Detection of Group A Beta Hemolytic Streptococci Species, emm, and Exotoxin Genes Isolated from Patients with Tonsillopharyngitis. Curr Microbiol 2020; 77:2064-2070. [PMID: 32367278 PMCID: PMC7222056 DOI: 10.1007/s00284-020-01994-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 04/10/2020] [Indexed: 10/25/2022]
Abstract
Group A Beta Hemolytic Streptococci (GAS) is the most critical human pathogen that leads to tonsillopharyngitis. The aims of this study were to identify GAS isolates and to determine emm typing, the coverage rate of available vaccines, and the distribution of superantigen gene profiles. 15 GAS isolates were isolated from throat cultures of 200 patients with tonsillopharyngitis, who were admitted to Canakkale Health Application and Research Hospital between October 2017 and May 2018. Identification of the isolates was performed by conventional methods and 16S rRNA sequence analysis. emm typing and exotoxin profiling of the isolates were performed by polymerase chain reaction. 7.5% GAS was detected in 200 patients. All the GAS isolates were identified as S. pyogenes. emm typing can be carried out in 13 S. pyogenes isolates. emm89 (33.3%), emm44 (20%), emm6 (13.3%), emm84 (6.7%), emm1 (6.7%), and emm18.1 (6.7%) were found to be six emm types. The coverage rate of S. pyogenes strains for 26-valent vaccine was 61.5% and for the 30-valent vaccine 84.6%. The most common exotoxin was speB (86.7%), followed by speC (60%), speF (33.3%), ssa (26.7%), speA (20%), speM (20%), speJ (13.3%), speL (6.7%), and speI (6.7%). As a result of determining the emm types of S. pyogenes species in Canakkale, it was concluded that the potential of 30-valent vaccine should be considered in Turkey and development of vaccines containing exotoxin types may be beneficial.
Collapse
Affiliation(s)
- Mehzat Altun
- Vocational School of Health Services, Canakkale Onsekiz Mart University, Canakkale, Turkey
| | - Binnur Mericli Yapıcı
- Department of Biology, Faculty of Sciences and Arts, Canakkale Onsekiz Mart University, 17020, Canakkale, Turkey.
| |
Collapse
|
36
|
Zhou P, Chen J, Li HH, Sun J, Gao SX, Zheng QW, Wei L, Jiang CY, Guan JC. Exposure of pregnant rats to staphylococcal enterotoxin B attenuates the response of increased Tregs to re-exposure to SEB in the thymus of adult offspring. Microb Pathog 2020; 145:104225. [PMID: 32353581 DOI: 10.1016/j.micpath.2020.104225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/06/2020] [Accepted: 04/20/2020] [Indexed: 11/17/2022]
Abstract
Regulatory T cells (Tregs) play an essential role during homeostasis and tolerance of the immune system. Based on our previous study that exposure of pregnant rats to staphylococcal enterotoxin B (SEB) can alter the percentage of CD4/CD8 subsets in the thymus of the offspring, in this study, we focus on the influence of exposure of pregnant rats to SEB on number, function and response of Tregs in the thymus of the offspring. Pregnant rats at gestational day of 16 were intravenously injected with 15 μg SEB and the thymuses of the neonatal and adult offspring were harvested for this study. We found that exposure of pregnant rats to SEB could significantly increase the absolute number of Tregs and the FoxP3 expression level in the thymus of not only neonatal but also adult offspring. Re-exposure of adult offspring to SEB remarkably reduced the suppressive capacity of Tregs to CD4+ T cells and the expression levels of TGF-β and IL-10 in the thymus, but had no effect on production of IL-4 and IFN-γ. Furthermore, it also notedly decreased the absolute number of Tregs and the FoxP3 expression level. These data suggest that prenatal exposure of pregnant rats to SEB attenuates the response of increased Tregs to re-exposure to SEB in the thymus of adult offspring.
Collapse
Affiliation(s)
- Ping Zhou
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Department of Microbiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Jie Chen
- Department of Cardiology, Jiande Branch, Second Affiliated Hospital, Zhejiang University School of Medicine, Jiande, 311600, PR China
| | - Hui-Hui Li
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Jing Sun
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Shu-Xian Gao
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Department of Microbiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Qing-Wei Zheng
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Li Wei
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China
| | - Cheng-Yi Jiang
- Department of Otolaryngology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233033, PR China
| | - Jun-Chang Guan
- Anhui Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, Anhui, 233030, PR China; Department of Microbiology, Bengbu Medical College, Bengbu, Anhui, 233030, PR China.
| |
Collapse
|
37
|
Allergy-A New Role for T Cell Superantigens of Staphylococcus aureus? Toxins (Basel) 2020; 12:toxins12030176. [PMID: 32178378 PMCID: PMC7150838 DOI: 10.3390/toxins12030176] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 12/16/2022] Open
Abstract
Staphylococcus aureus superantigens (SAgs) are among the most potent T cell mitogens known. They stimulate large fractions of T cells by cross-linking their T cell receptor with major histocompatibility complex class-II molecules on antigen presenting cells, resulting in T cell proliferation and massive cytokine release. To date, 26 different SAgs have been described in the species S. aureus; they comprise the toxic shock syndrome toxin (TSST-1), as well as 25 staphylococcal enterotoxins (SEs) or enterotoxin-like proteins (SEls). SAgs can cause staphylococcal food poisoning and toxic shock syndrome and contribute to the clinical symptoms of staphylococcal infection. In addition, there is growing evidence that SAgs are involved in allergic diseases. This review provides an overview on recent epidemiological data on the involvement of S. aureus SAgs and anti-SAg-IgE in allergy, demonstrating that being sensitized to SEs—in contrast to inhalant allergens—is associated with a severe disease course in patients with chronic airway inflammation. The mechanisms by which SAgs trigger or amplify allergic immune responses, however, are not yet fully understood. Here, we discuss known and hypothetical pathways by which SAgs can drive an atopic disease.
Collapse
|
38
|
Glik A, Douvdevani A. T Lymphocytes: The “Cellular” Arm of Acquired Immunity in the Peritoneum. Perit Dial Int 2020. [DOI: 10.1177/089686080602600407] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
T cells are an important part of the acquired immune response and target specific antigen with their T cell receptor. The peritoneum is a special milieu within which T cells react. We describe briefly the anatomy important for T cell function. T cell biology including antigen presentation, T cell activation, and the different T cell subpopulations are reviewed. We also define innate and acquired immunity and describe the role of polymorphonuclear cells and peritoneal mesothelial cells in the regulation of leukocyte population recruitment during peritonitis. We focus particularly on peritoneal lymphocytes and compare them to the regular lymphocyte populations in the circulation. We illustrate the role of PMCs in antigen presentation and discuss the changes of CD4+ helper T cell subtypes (Th1 and Th2) during peritoneal dialysis. The role of CD8+ cytotoxic T lymphocytes and their possible destructive role for the peritoneal membrane modified by advanced glycation end products are discussed. Polymorphonuclear cells play an important role in the regulation of inflammation and immunity. We describe their possible role in supporting T cells and particularly for generating memory CD8+ T cells by secretion of interleukin-15, a potent T cell growth factor. Light is shed on γδ T cells, a special T cell population that is able to recognize antigens without the restriction of antigen presentation. We end our review with a description of regulatory T cells. This cell population is extremely important in preventing autoimmunity and in the regulation of acquired immunity.
Collapse
Affiliation(s)
- Amir Glik
- Department of Nephrology, Soroka Medical Center, and
Clinical Biochemistry Department, Faculty of Health Sciences, Ben Gurion University
of the Negev, Beer Sheva, Israel
| | - Amos Douvdevani
- Department of Nephrology, Soroka Medical Center, and
Clinical Biochemistry Department, Faculty of Health Sciences, Ben Gurion University
of the Negev, Beer Sheva, Israel
| |
Collapse
|
39
|
Hnasko R, Lin AV, McGarvey JA. Rapid Detection of Staphylococcal Enterotoxin-B by Lateral Flow Assay. Monoclon Antib Immunodiagn Immunother 2020; 38:209-212. [PMID: 31603743 PMCID: PMC6791480 DOI: 10.1089/mab.2019.0028] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A cohort of monoclonal antibodies (mAbs) were generated against Staphylococcal enterotoxin-B (SEB) and selected by double sandwich enzyme-linked immunosorbent assay (ELISA) for solution capture of the toxin. Clonal hybridoma cell lines were established and a pair of anti-SEB mAbs selected for the development of a sandwich ELISA. Immobilized 3D6 mAb (IgG1, kappa) when paired with 4C9 mAb (IgG1, kappa) conjugated to horseradish peroxidase generates a typical dose–response curve with an EC50 of 24.8 ng/mL for purified SEB using chemiluminescent detection. These mAbs bind SEB by Western blot and ELISA binding to classical enterotoxin serotypes show that the 3D6 mAb binds both SEB and the SEC1 serotypes, whereas 4C9 binds only SEB. These mAbs effectively port onto lateral flow test strips with a visual detection sensitivity for SEB of 5 ng/mL in <10 minutes using a 4C9 conjugated to a 40 nm gold reporter.
Collapse
Affiliation(s)
- Robert Hnasko
- USDA-ARS Produce Safety and Microbiology Research Unit (PSM), Albany, California
| | - Alice V Lin
- USDA-ARS Produce Safety and Microbiology Research Unit (PSM), Albany, California
| | - Jeffery A McGarvey
- USDA-ARS Foodborne Toxin Detection and Prevention Research Unit (FTDP), Albany, California
| |
Collapse
|
40
|
Wrotek S, Sobocińska J, Kozłowski HM, Pawlikowska M, Jędrzejewski T, Dzialuk A. New Insights into the Role of Glutathione in the Mechanism of Fever. Int J Mol Sci 2020; 21:ijms21041393. [PMID: 32092904 PMCID: PMC7073131 DOI: 10.3390/ijms21041393] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/14/2020] [Accepted: 02/17/2020] [Indexed: 12/17/2022] Open
Abstract
Glutathione is one of the most important and potent antioxidants. The development of pharmacological compounds that can either increase or decrease glutathione concentrations has allowed investigation into the role of glutathione in various biological processes, including immune responses. Recent findings have shown that glutathione not only affects certain factors involved in immunological processes but also modifies complex immune reactions such as fever. Until recently, it was not known why some patients do not develop fever during infection. Data suggest that fever induction is associated with oxidative stress; therefore, antioxidants such as glutathione can reduce pyrexia. Surprisingly, new studies have shown that low glutathione levels can also inhibit fever. In this review, we focus on recent advances in this area, with an emphasis on the role of glutathione in immune responses accompanied by fever. We describe evidence showing that disturbed glutathione homeostasis may be responsible for the lack of fever during infections. We also discuss the biological significance of the antipyretic effects produced by pharmacological glutathione modulators.
Collapse
Affiliation(s)
- Sylwia Wrotek
- Department of Immunology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 1 Lwowska Str., 87-100 Torun, Poland; (J.S.); (H.M.K.); (M.P.); (T.J.)
- Correspondence: (S.W.); (A.D.)
| | - Justyna Sobocińska
- Department of Immunology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 1 Lwowska Str., 87-100 Torun, Poland; (J.S.); (H.M.K.); (M.P.); (T.J.)
| | - Henryk M. Kozłowski
- Department of Immunology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 1 Lwowska Str., 87-100 Torun, Poland; (J.S.); (H.M.K.); (M.P.); (T.J.)
| | - Małgorzata Pawlikowska
- Department of Immunology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 1 Lwowska Str., 87-100 Torun, Poland; (J.S.); (H.M.K.); (M.P.); (T.J.)
| | - Tomasz Jędrzejewski
- Department of Immunology, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 1 Lwowska Str., 87-100 Torun, Poland; (J.S.); (H.M.K.); (M.P.); (T.J.)
| | - Artur Dzialuk
- Department of Genetics, Faculty of Biological Sciences, Kazimierz Wielki University, 10 Powstańców Wielkopolskich Ave., 85-090 Bydgoszcz, Poland
- Correspondence: (S.W.); (A.D.)
| |
Collapse
|
41
|
Noli Truant S, De Marzi MC, Sarratea MB, Antonoglou MB, Meo AP, Iannantuono López LV, Fernández Lynch MJ, Todone M, Malchiodi EL, Fernández MM. egc Superantigens Impair Monocytes/Macrophages Inducing Cell Death and Inefficient Activation. Front Immunol 2020; 10:3008. [PMID: 32010128 PMCID: PMC6974467 DOI: 10.3389/fimmu.2019.03008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022] Open
Abstract
Bacterial superantigens (SAgs) are enterotoxins that bind to MHC-II and TCR molecules, activating as much as 20% of the T cell population and promoting a cytokine storm which enhances susceptibility to endotoxic shock, causing immunosuppression, and hindering the immune response against bacterial infection. Since monocytes/macrophages are one of the first cells SAgs find in infected host and considering the effect these cells have on directing the immune response, here, we investigated the effect of four non-classical SAgs of the staphylococcal egc operon, namely, SEG, SEI, SEO, and SEM on monocytic-macrophagic cells, in the absence of T cells. We also analyzed the molecular targets on APCs which could mediate SAg effects. We found that egc SAgs depleted the pool of innate immune effector cells and induced an inefficient activation of monocytic-macrophagic cells, driving the immune response to an impaired proinflammatory profile, which could be mediated directly or indirectly by interactions with MHC class II. In addition, performing surface plasmon resonance assays, we demonstrated that non-classical SAgs bind the gp130 molecule, which is also present in the monocytic cell surface, among other cells.
Collapse
Affiliation(s)
- Sofia Noli Truant
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mauricio C De Marzi
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina.,Instituto de Ecología y Desarrollo Sustentable (INEDES), UNLU-CONICET, Universidad Nacional de Luján, Luján, Argentina
| | - María B Sarratea
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María B Antonoglou
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana P Meo
- Hospital Dr. J. M. Ramos Mejía, Buenos Aires, Argentina
| | - Laura V Iannantuono López
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María J Fernández Lynch
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marcos Todone
- Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Ciencias Básicas, Universidad Nacional de Luján, Luján, Argentina.,Instituto de Ecología y Desarrollo Sustentable (INEDES), UNLU-CONICET, Universidad Nacional de Luján, Luján, Argentina
| | - Emilio L Malchiodi
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marisa M Fernández
- Cátedra de Inmunología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Estudios de la Inmunidad Humoral "Prof. Ricardo A. Margni" (IDEHU), UBA-CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
42
|
Dey I, Bishayi B. Impact of simultaneous neutralization of IL-17A and treatment with recombinant IL-2 on Th17-Treg cell population in S.aureus induced septic arthritis. Microb Pathog 2019; 139:103903. [PMID: 31790794 DOI: 10.1016/j.micpath.2019.103903] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 01/05/2023]
Abstract
The contribution of Th17 and Treg in the pathogenesis of septic arthritis is well known. The imbalance of Th17/Treg ratio, especially the skewed CD4+ T cell differentiation towards pathogenic Th17 lineage is a major reason that mediates bone damage through one of its prime cytokine member IL-17A. The neutralization of released IL-17A, as well as exogenous administration of IL-2 at a lower dose, was seen to be potent in dampening the inflammatory response in many cases. Interestingly the effect of IL-17A neutralization to limit IL-17 mediated inflammation and induction of Tregs by the administration of IL-2 has not been studied in experimental arthritis. So in this study, we have treated arthritic mice with IL-17A Ab and recombinant mouse IL-2 either alone or in combination at 3, 9 and 15 days post-infection. We have found a marked decrease in Th17 cell population and their related pro-inflammatory cytokine levels at 15DPI in arthritic mice after IL-17 neutralization. An increased Treg cell population was also observed in mice after application of rIL-2 with a significantly heightened TGF-β level in serum and synovial joints compared to the untreated one. However, in the case of combination therapy of IL-17A Ab and rIL-2 we have observed a beneficial effect in ameliorating the disease outcome as the arthritic index was decreased maximally at 15DPI with a significant reduction of arthritis compared to individual treatment. Overall the inflammatory microenvironment was counterbalanced most effectively in combination treatment by lowering the Th17/Treg ratio and their related cytokines that resulted in reducing the immunopathogenesis of the destructive arthritis.
Collapse
Affiliation(s)
- Ipsita Dey
- Department of Physiology, Immunology laboratory, University of Calcutta, University Colleges of Science and Technology, Calcutta, West Bengal, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology laboratory, University of Calcutta, University Colleges of Science and Technology, Calcutta, West Bengal, India.
| |
Collapse
|
43
|
Staphylococcus aureus Toxins: From Their Pathogenic Roles to Anti-virulence Therapy Using Natural Products. BIOTECHNOL BIOPROC E 2019. [DOI: 10.1007/s12257-019-0059-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
44
|
Babbar A, Barrantes I, Pieper DH, Itzek A. Superantigen SpeA attenuates the biofilm forming capacity of Streptococcus pyogenes. J Microbiol 2019; 57:626-636. [DOI: 10.1007/s12275-019-8648-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/15/2019] [Accepted: 02/01/2019] [Indexed: 12/22/2022]
|
45
|
Multivariate profiling of African green monkey and rhesus macaque T lymphocytes. Sci Rep 2019; 9:4834. [PMID: 30886198 PMCID: PMC6423277 DOI: 10.1038/s41598-019-41209-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 02/27/2019] [Indexed: 12/22/2022] Open
Abstract
The complexity of immune responses limits the usefulness of univariate methods in answering complex immunology questions. To demonstrate the utility of a multivariate approach, we employ such approach to compare T cells of African green monkeys (AGMs) and rhesus macaques (RMs). Among the most prominent distinguishing features we found were lower CD3 and higher CD28 surface expression in AGMs compared to RMs. After in vitro stimulation, a larger proportion of AGM T cells secreted cytokines, especially those producing more than one cytokine (i.e. multifunctional cells). To find out whether multifunctional responses associate with protection in other species, we compared T cells of cynomolgus macaques (CMs) infected with wild-type Simian Immunodeficiency Virus (SIV) to those of CMs infected (vaccinated) with a replication-defective virus. Wild-type SIV infection in macaques leads to simian Acquired Immunodeficiency Syndrome (AIDS), which does not happen in animals previously vaccinated with a replication-defective virus. Interestingly, after in vitro stimulation, multifunctional cells were more abundant among T cells of vaccinated CMs. Our results propose T-cell multifunctionality as a potentially useful marker of immunity, although additional verification is needed. Finally, we hope our multivariate model and its associated validation methods will inform future studies in the field of immunology.
Collapse
|
46
|
Malke H. Genetics and Pathogenicity Factors of Group C and G Streptococci. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0002-2017. [PMID: 30873932 PMCID: PMC11590425 DOI: 10.1128/microbiolspec.gpp3-0002-2017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Indexed: 12/17/2022] Open
Abstract
Of the eight phylogenetic groups comprising the genus Streptococcus, Lancefield group C and G streptococci (GCS and GGS, resp.) occupy four of them, including the Pyogenic, Anginosus, and Mitis groups, and one Unnamed group so far. These organisms thrive as opportunistic commensals in both humans and animals but may also be associated with clinically serious infections, often resembling those due to their closest genetic relatives, the group A streptoccci (GAS). Advances in molecular genetics, taxonomic approaches and phylogenomic studies have led to the establishment of at least 12 species, several of which being subdivided into subspecies. This review summarizes these advances, citing 264 early and recent references. It focuses on the molecular structure and genetic regulation of clinically important proteins associated with the cell wall, cytoplasmic membrane and extracellular environment. The article also addresses the question of how, based on the current knowledge, basic research and translational medicine might proceed to further advance our understanding of these multifaceted organisms. Particular emphasis in this respect is placed on streptokinase as the protein determining the host specificity of infection and the Rsh-mediated stringent response with its potential for supporting bacterial survival under nutritional stress conditions.
Collapse
Affiliation(s)
- Horst Malke
- Friedrich Schiller University Jena, Faculty of Biology and Pharmacy, D-07743 Jena, Germany, and University of Oklahoma Health Sciences Center, Department of Microbiology and Immunology, Oklahoma City, OK 73190
| |
Collapse
|
47
|
Huang SM, Huang SH, Weng KP, Chien KJ, Lin CC, Huang YF. Update on association between Kawasaki disease and infection. J Chin Med Assoc 2019; 82:172-174. [PMID: 30913113 DOI: 10.1097/jcma.0000000000000025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The relationship between infection and Kawasaki disease (KD) remains unclear. Infection has long been considered a key predisposing factor for KD. Bacterial and viral agents may be related to the onset of KD because of superantigen and cytokine production. Various bacterial and viral infections have been reported to be associated with KD, but the actual mechanism remains unknown. The higher association between KD and enterovirus has been well documented by using Taiwan National Health Insurance Research Database. However, no evidence has been obtained that various bacterial and viral infections induce KD. Comprehensive research, including infectious agents, should be conducted to elucidate the pathogenesis of KD.
Collapse
Affiliation(s)
- Shih-Ming Huang
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
- Department of Pediatrics, Kaohsiung Municipal United Hospital, Kaohsiung, Taiwan, ROC
| | - Shih-Hui Huang
- Department of Nursing, Fooyin University, Kaohsiung, Taiwan, ROC
| | - Ken-Pen Weng
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
- Department of Physical Therapy, Shu-Zen College of Medicine and Management, Kaohsiung, Taiwan, ROC
| | - Kuang-Jen Chien
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Chu-Chuan Lin
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Yung-Feng Huang
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| |
Collapse
|
48
|
Golob-Urbanc A, Rajčević U, Strmšek Ž, Jerala R. Design of split superantigen fusion proteins for cancer immunotherapy. J Biol Chem 2019; 294:6294-6305. [PMID: 30782846 DOI: 10.1074/jbc.ra118.006742] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/26/2019] [Indexed: 11/06/2022] Open
Abstract
Several antibody-targeting cancer immunotherapies have been developed based on T cell activation at the target cells. One of the most potent activators of T cells are bacterial superantigens, which bind to major histocompatibility complex class II on antigen-presenting cells and activate T cells through T cell receptor. Strong T cell activation is also one of the main weaknesses of this strategy as it may lead to systemic T cell activation. To overcome the limitation of conventional antibody-superantigen fusion proteins, we have split a superantigen into two fragments, individually inactive, until both fragments came into close proximity and reassembled into a biologically active form capable of activating T cell response. A screening method based on fusion between SEA and coiled-coil heterodimers was developed that enabled detection of functional split SEA designs. The split SEA design that demonstrated efficacy in fusion with coiled-coil dimer forming polypeptides was fused to a single chain antibody specific for tumor antigen CD20. This design selectively activated T cells by split SEA-scFv fusion binding to target cells.
Collapse
Affiliation(s)
- Anja Golob-Urbanc
- From the Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia.,Graduate School of Biomedicine, University of Ljubljana, 1000 Ljubljana, Slovenia, and
| | - Uroš Rajčević
- Department of Research and Development, Blood Transfusion Centre of Slovenia, Šlajmerjeva 6, 1000 Ljubljana, Slovenia
| | - Žiga Strmšek
- From the Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia.,Graduate School of Biomedicine, University of Ljubljana, 1000 Ljubljana, Slovenia, and
| | - Roman Jerala
- From the Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia,
| |
Collapse
|
49
|
Sant AJ. Overview of T-Cell Recognition. Clin Immunol 2019. [DOI: 10.1016/b978-0-7020-6896-6.00006-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
50
|
Nakamura A, Ikeda K, Hamaoka K. Aetiological Significance of Infectious Stimuli in Kawasaki Disease. Front Pediatr 2019; 7:244. [PMID: 31316950 PMCID: PMC6611380 DOI: 10.3389/fped.2019.00244] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 05/29/2019] [Indexed: 01/23/2023] Open
Abstract
Kawasaki disease (KD) is a pediatric vasculitis syndrome that is often involves coronary artery lesions (e. g., coronary artery aneurysms). Although its causal factors and entire pathogenesis remain elusive, the available evidence indicates that the pathogenesis of KD is closely associated with dysregulation of immune responses to various viruses or microbes. In this short review, we address several essential aspects of the etiology of KD with respect to the immune response to infectious stimuli: 1) the role of viral infections, 2) the role of bacterial infections and the superantigen hypothesis, 3) involvement of innate immune response including pathogens/microbe-associated molecular patterns and complement pathways, and 4) the influence of genetic background on the response to infectious stimuli. Based on the clinical and experimental evidence, we discuss the possibility that a wide range of microbes and viruses could cause KD through common and distinct immune processes.
Collapse
Affiliation(s)
- Akihiro Nakamura
- Central Research Laboratory, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuyuki Ikeda
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenji Hamaoka
- Pediatric Cardiology and Kawasaki Disease Center, Uji-Tokushukai Medical Center, Kyoto, Japan.,Faculty of Life and Medical Sciences, Doshisha University, Kyoto, Japan
| |
Collapse
|