1
|
Ullah A, Chen Y, Singla RK, Cao D, Shen B. Exploring cytokines dynamics: Uncovering therapeutic concepts for metabolic disorders in postmenopausal women- diabetes, metabolic bone diseases, and non-alcohol fatty liver disease. Ageing Res Rev 2024; 101:102505. [PMID: 39307315 DOI: 10.1016/j.arr.2024.102505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/18/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024]
Abstract
Menopause is an age-related change that persists for around one-third of a woman's life. Menopause increases the risk of metabolic illnesses such as diabetes, osteoporosis (OP), and nonalcoholic fatty liver disease (NAFLD). Immune mediators (pro-inflammatory cytokines), such as interleukin-1 (IL-1), IL-6, IL-17, transforming growth factor (TGF), and tumor necrosis factor (TNF), exacerbate the challenges of a woman undergoing menopause by causing inflammation and contributing to the development of these metabolic diseases in postmenopausal women. Furthermore, studies have shown that anti-inflammatory cytokines such as interleukin-1 receptor antagonists (IL-1Ra), IL-2, and IL-10 have a double-edged effect on diabetes and OP. Likewise, several interferon (IFN) members are double-edged swords in the OP. Therefore, addressing these immune mediators precisely may be an approach to improving the health of postmenopausal women. Hence, considering the significant changes in these cytokines, the present review focuses on the latest findings concerning the molecular mechanisms by which pro- and anti-inflammatory cytokines (interleukins) impact postmenopausal women with diabetes, OP, and NAFLD. Furthermore, we comprehensively discuss the therapeutic approaches that identify cytokines as therapeutic targets, such as hormonal therapy, physical activities, natural inhibitors (drugs), and others. Finally, this review aims to provide valuable insights into the role of cytokines in postmenopausal women's diabetes, OP, and NAFLD. Deeply investigating the mechanisms and therapeutic interventions involved will address the characteristics of immune mediators (cytokines) and improve the management of these illnesses, thereby enhancing the general quality of life and health of the corresponding populations of women.
Collapse
Affiliation(s)
- Amin Ullah
- Department of Abdominal Oncology, Cancer Center of West China Hospital and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yongxiu Chen
- Gynecology Department, Guangdong Women and Children Hospital, No. 521, Xingnan Road, Panyu District, Guangzhou 511442, China
| | - Rajeev K Singla
- Department of Abdominal Oncology, Cancer Center of West China Hospital and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Dan Cao
- Department of Abdominal Oncology, Cancer Center of West China Hospital and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Bairong Shen
- Department of Abdominal Oncology, Cancer Center of West China Hospital and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
2
|
Bleck D, Loacker-Schöch K, Classen T, Jose J, Schneider M, Pongratz G. Fibroblast-like synoviocytes preferentially induce terminal differentiation of IgD + memory B cells instead of naïve B cells. Immunology 2024; 173:520-535. [PMID: 39054787 DOI: 10.1111/imm.13840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 07/10/2024] [Indexed: 07/27/2024] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease driven by highly active autoantibody-producing B cells. Activation of B cells is maintained within ectopic germinal centres found in affected joints. Fibroblast-like synoviocytes (FLS) present in inflamed joints support B-cell survival, activation, and differentiation. CD27+ memory B cells and naive B cells show very different responses to activation, particularly by CD40 ligand (CD40L). We show that FLS-dependent activation of human B cells is dependent on interleukin-6 (IL-6) and CD40L. FLS have been shown to activate both naive and memory B cells. Whether the activating potential of FLS is different for naive and memory B cells has not been investigated. Our results suggest that FLS-induced activation of B cells is dependent on IL-6 and CD40L. While FLS are able to induce plasma cell differentiation, isotype switching, and antibody production in memory B cells, the ability of FLS to activate naive B cells is significantly lower.
Collapse
Affiliation(s)
- Dennis Bleck
- Clinic for Rheumatology, Medical Faculty of Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
- Hiller Research Center, Medical Faculty of Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Klara Loacker-Schöch
- Clinic for Rheumatology, Medical Faculty of Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
- Hiller Research Center, Medical Faculty of Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Tim Classen
- Clinic of Orthopedics/Orthopedic Rheumatology, St. Elisabeth-Hospital Meerbusch-Lank, Meerbusch, Germany
| | - Joachim Jose
- Institute of Pharmaceutical and Medicinal Chemistry, PharmaCampus, Westphalian Wilhelms-University, Muenster, Germany
| | - Matthias Schneider
- Clinic for Rheumatology, Medical Faculty of Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
- Hiller Research Center, Medical Faculty of Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Georg Pongratz
- Clinic for Rheumatology, Medical Faculty of Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
- Hiller Research Center, Medical Faculty of Heinrich Heine University, University Hospital Düsseldorf, Düsseldorf, Germany
- Department of Rheumatology, Barmherzige Brueder Hospital Regensburg, Regensburg, Germany
- Medical Faculty of the University of Regensburg, Regensburg, Germany
| |
Collapse
|
3
|
Fechtner S, Allen BE, Chriswell ME, Jubair WK, Robertson CE, Kofonow JN, Frank DN, Holers VM, Kuhn KA. 3,3-Dimethyl-1-Butanol and its Metabolite 3,3-Dimethylbutyrate Ameliorate Collagen-induced Arthritis Independent of Choline Trimethylamine Lyase Activity. Inflammation 2024:10.1007/s10753-024-02126-y. [PMID: 39153148 DOI: 10.1007/s10753-024-02126-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/18/2024] [Accepted: 08/08/2024] [Indexed: 08/19/2024]
Abstract
Conflicting data exist in rheumatoid arthritis and the collagen-induced arthritis (CIA) murine model of autoimmune arthritis regarding the role of bacterial carnitine and choline metabolism into the inflammatory product trimethylamine (TMA), which is oxidized in the liver to trimethylamine-N-oxide (TMAO). Using two published inhibitors of bacterial TMA lyase, 3,3-dimethyl-1-butanol (DMB) and fluoromethylcholine (FMC), we tested if TMA/TMAO were relevant to inflammation in the development of CIA. Surprisingly, DMB-treated mice demonstrated > 50% reduction in arthritis severity compared to FMC and vehicle-treated mice, but amelioration of disease was independent of TMA/TMAO production. Given the apparent contradiction that DMB did not inhibit TMA, we then investigated the mechanism of protection by DMB. After verifying that DMB acted independently of the intestinal microbiome, we traced the metabolism of DMB within the host and identified a novel host-derived metabolite of DMB, 3,3-dimethyl-1-butyric acid (DMBut). In vivo studies of mice treated with DMB or DMBut demonstrated efficacy of both molecules in significantly reducing disease and proinflammatory cytokines in CIA, while in vitro studies suggest these molecules may act by modulating secretion of proinflammatory cytokines from macrophages. Altogether, our study suggests that DMB and/or its metabolites are protective in CIA through direct immunomodulatory effects rather than inhibition of bacterial TMA lyases.
Collapse
Affiliation(s)
- Sabrina Fechtner
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brendan E Allen
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Meagan E Chriswell
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Widian K Jubair
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Charles E Robertson
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer N Kofonow
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Daniel N Frank
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - V Michael Holers
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kristine A Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
4
|
Netti GS, Soccio P, Catalano V, De Luca F, Khalid J, Camporeale V, Moriondo G, Papale M, Scioscia G, Corso G, Foschino MP, Lo Caputo S, Lacedonia D, Ranieri E. The Onset of Antinuclear Antibodies (ANAs) as a Potential Risk Factor for Mortality and Morbidity in COVID-19 Patients: A Single-Center Retrospective Study. Biomedicines 2024; 12:1306. [PMID: 38927513 PMCID: PMC11201662 DOI: 10.3390/biomedicines12061306] [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: 05/13/2024] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The immune system's amplified response to SARS-CoV-2 may lead to the production of autoantibodies, but their specific impact on disease severity and outcome remains unclear. This study aims to assess if hospitalized COVID-19 patients face a worse prognosis based on ANA presence, even without autoimmune diseases. We performed a retrospective, single-center, observational cohort study, enrolling 638 COVID-19 patients hospitalized from April 2020 to March 2021 at Hospital "Policlinico Riuniti" of Foggia (Italy). COVID-19 patients with a positive ANA test exhibited a significantly lower 30-day survival rate (64.4% vs. 83.0%) and a higher likelihood of severe respiratory complications during hospitalization than those with negative ANA screening (35.4% vs. 17.0%) (p < 0.001). The association between poor prognosis and ANA status was identified by calculating the HALP score (Hemoglobin-Albumin-Lymphocyte-Platelet), which was lower in COVID-19 patients with a positive ANA test compared to ANA-negative patients (108.1 ± 7.4 vs. 218.6 ± 11.2 AU; p < 0.011). In detail, COVID-19 patients with a low HALP showed a lower 30-day survival rate (99.1% vs. 83.6% vs. 55.2% for high, medium, and low HALP, respectively; p < 0.001) and a higher incidence of adverse respiratory events compared to those with high and medium HALP (13.1% vs. 35.2% vs. 64.6% for high, medium, and low HALP, respectively; p < 0.001). In summary, ANA positivity in COVID-19 patients appears to be linked to a more aggressive disease phenotype with a reduced survival rate. Furthermore, we propose that the HALP score could serve as a valuable parameter to assess prognosis for COVID-19 patients.
Collapse
Affiliation(s)
- Giuseppe Stefano Netti
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
- Unit of Clinical Pathology, Department of Laboratory Diagnostics, University Hospital “Policlinico Riuniti”, 71122 Foggia, Italy; (M.P.); (G.C.)
| | - Piera Soccio
- Unit of Respiratory Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.M.); (G.S.); (M.P.F.); (D.L.)
| | - Valeria Catalano
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
| | - Federica De Luca
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
| | - Javeria Khalid
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
| | - Valentina Camporeale
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
| | - Giorgia Moriondo
- Unit of Respiratory Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.M.); (G.S.); (M.P.F.); (D.L.)
| | - Massimo Papale
- Unit of Clinical Pathology, Department of Laboratory Diagnostics, University Hospital “Policlinico Riuniti”, 71122 Foggia, Italy; (M.P.); (G.C.)
| | - Giulia Scioscia
- Unit of Respiratory Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.M.); (G.S.); (M.P.F.); (D.L.)
| | - Gaetano Corso
- Unit of Clinical Pathology, Department of Laboratory Diagnostics, University Hospital “Policlinico Riuniti”, 71122 Foggia, Italy; (M.P.); (G.C.)
- Clinical Biochemistry, Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Maria Pia Foschino
- Unit of Respiratory Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.M.); (G.S.); (M.P.F.); (D.L.)
| | - Sergio Lo Caputo
- Unit of Infectious Diseases, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
| | - Donato Lacedonia
- Unit of Respiratory Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.M.); (G.S.); (M.P.F.); (D.L.)
| | - Elena Ranieri
- Unit of Clinical Pathology, Advanced Research Center on Kidney Aging (A.R.K.A.), Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (V.C.); (F.D.L.); (J.K.); (V.C.); (E.R.)
- Unit of Clinical Pathology, Department of Laboratory Diagnostics, University Hospital “Policlinico Riuniti”, 71122 Foggia, Italy; (M.P.); (G.C.)
| |
Collapse
|
5
|
Doctor GT, Dudreuilh C, Perera R, Dorling A. Granulomatous Tubulointerstitial Nephritis in a Kidney Allograft: Treatment with Interleukin-6 Receptor Antagonist Stabilises Kidney Function. J Clin Med 2024; 13:3427. [PMID: 38929956 PMCID: PMC11205090 DOI: 10.3390/jcm13123427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Granulomatous tubulointerstitial nephritis (GTIN) attributed to early onset sarcoidosis is an ultrarare finding in an allograft kidney biopsy. We present the case of a young man with allograft dysfunction who had GTIN upon biopsy. We performed a thorough case review based on recovered records from early childhood and reassessed genetic testing results. We revised his underlying diagnosis from cryopyrin-associated periodic syndrome to early-onset sarcoidosis with wild-type NOD2 and established a rationale to use the interleukin-6 (IL-6) receptor blocker tocilizumab (TCZ). This suppressed his inflammatory disease and stabilised kidney function. We performed a literature review related to the emerging role of IL-6 pathway blockade in kidney transplantation. We identified 18 reports with 417 unique patients treated with TCZ for indications including HLA-desensitisation, transplant immunosuppression induction, treatment of chronic antibody-mediated rejection, and treatment of subclinical rejection. Both TCZ and the direct IL-6 inhibitor clazakizumab are being studied in ongoing randomised control trials.
Collapse
Affiliation(s)
- Gabriel T. Doctor
- Department of Transplantation, Renal and Urology, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 9RT, UK; (C.D.); (R.P.); (A.D.)
| | | | | | | |
Collapse
|
6
|
Cieślik M, Strobel SD, Bryniarski P, Twardowska H, Chmielowski A, Rudek M, Felkle D, Zięba K, Kaleta K, Jarczyński M, Nowak B, Bryniarski K, Nazimek K. Hypotensive drugs mitigate the high-sodium diet-induced pro-inflammatory activation of mouse macrophages in vivo. Biomed Pharmacother 2024; 175:116648. [PMID: 38677242 DOI: 10.1016/j.biopha.2024.116648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 04/29/2024] Open
Abstract
Nowadays, there is an increasing emphasis on the need to alleviate the chronic inflammatory response to effectively treat hypertension. However, there are still gaps in our understanding on how to achieve this. Therefore, research on interaction of antihypertensive drugs with the immune system is extremely interesting, since their therapeutic effect could partly result from amelioration of hypertension-related inflammation, in which macrophages seem to play a pivotal role. Thus, current comprehensive studies have investigated the impact of repeatedly administered hypotensive drugs (captopril, olmesartan, propranolol, carvedilol, amlodipine, verapamil) on macrophage functions in the innate and adaptive immunity, as well as if drug-induced effects are affected by a high-sodium diet (HSD), one of the key environmental risk factors of hypertension. Although the assayed medications increased the generation of reactive oxygen and nitrogen intermediates by macrophages from standard fed donors, they reversed HSD-induced enhancing effects on macrophage oxidative burst and secretion of pro-inflammatory cytokines. On the other hand, some drugs increased macrophage phagocytic activity and the expression of surface markers involved in antigen presentation, which translated into enhanced macrophage ability to activate B cells for antibody production. Moreover, the assayed medications augmented macrophage function and the effector phase of contact hypersensitivity reaction, but suppressed the sensitization phase of cell-mediated hypersensitivity under HSD conditions. Our current findings contribute to the recognition of mechanisms, by which excessive sodium intake affects macrophage immune activity in hypertensive individuals, and provide evidence that the assayed medications mitigate most of the HSD-induced adverse effects, suggesting their additional protective therapeutic activity.
Collapse
Affiliation(s)
- Martyna Cieślik
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Spencer D Strobel
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Paweł Bryniarski
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Hanna Twardowska
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Adam Chmielowski
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Michał Rudek
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Dominik Felkle
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Katarzyna Zięba
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Konrad Kaleta
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Mateusz Jarczyński
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Bernadeta Nowak
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Krzysztof Bryniarski
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland
| | - Katarzyna Nazimek
- Department of Immunology, Jagiellonian University Medical College, 18 Czysta St., Krakow 31-121, Poland.
| |
Collapse
|
7
|
Chen W, Toda E, Takeuchi K, Sawa Y, Wakamatsu K, Kuwahara N, Ishikawa A, Igarashi Y, Terasaki M, Kunugi S, Terasaki Y, Yamada K, Terashima Y, Shimizu A. Disulfiram treatment suppresses antibody-producing reactions by inhibiting macrophage activation and B cell pyrimidine metabolism. Commun Biol 2024; 7:488. [PMID: 38649462 PMCID: PMC11035657 DOI: 10.1038/s42003-024-06183-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Antibody responses, involving B cells, CD4 + T cells, and macrophages, are implicated in autoimmune diseases and organ transplant rejection. We have previously shown that inhibiting FROUNT with disulfiram (DSF) suppresses macrophage activation and migration, effectively treating inflammatory diseases. In this study, we investigated the effectiveness of DSF in antibody-producing reactions. Using a heart transplantation mouse model with antibody-mediated rejection, we administered anti-CD8 antibody to exclude cellular rejection. DSF directly inhibited B cell responses in vitro and significantly reduced plasma donor-specific antibodies and graft antibody deposition in vivo, resulting in prolonged survival of the heart graft. DSF also mediated various effects, including decreased macrophage infiltration and increased Foxp3+ regulatory T-cells in the grafts. Additionally, DSF inhibited pyrimidine metabolism-related gene expression induced by B-cell stimulation. These findings demonstrate that DSF modulates antibody production in the immune response complexity by regulating B-cell and macrophage responses.
Collapse
Affiliation(s)
- Weili Chen
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Etsuko Toda
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan.
- Laboratory for Morphological and Biomolecular Imaging, Nippon Medical School, Tokyo, Japan.
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan.
| | - Kazuhiro Takeuchi
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
- Division of Organ Replacement and Xenotransplantation Surgery, Center for Advanced Biomedical Science and Swine Research, Kagoshima University, Kagoshima, Japan
| | - Yurika Sawa
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Kyoko Wakamatsu
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Naomi Kuwahara
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Arimi Ishikawa
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Yuri Igarashi
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Mika Terasaki
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Shinobu Kunugi
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | | | - Kazuhiko Yamada
- Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Yuya Terashima
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Akira Shimizu
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan.
| |
Collapse
|
8
|
Isaac AH, Recalde Phillips SY, Ruben E, Estes M, Rajavel V, Baig T, Paleti C, Landsgaard K, Lee RH, Guda T, Criscitiello MF, Gregory C, Alge DL. Impact of PEG sensitization on the efficacy of PEG hydrogel-mediated tissue engineering. Nat Commun 2024; 15:3283. [PMID: 38637507 PMCID: PMC11026400 DOI: 10.1038/s41467-024-46327-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 02/22/2024] [Indexed: 04/20/2024] Open
Abstract
While poly(ethylene glycol) (PEG) hydrogels are generally regarded as biologically inert blank slates, concerns over PEG immunogenicity are growing, and the implications for tissue engineering are unknown. Here, we investigate these implications by immunizing mice against PEG to stimulate anti-PEG antibody production and evaluating bone defect regeneration after treatment with bone morphogenetic protein-2-loaded PEG hydrogels. Quantitative analysis reveals that PEG sensitization increases bone formation compared to naive controls, whereas histological analysis shows that PEG sensitization induces an abnormally porous bone morphology at the defect site, particularly in males. Furthermore, immune cell recruitment is higher in PEG-sensitized mice administered the PEG-based treatment than their naive counterparts. Interestingly, naive controls that were administered a PEG-based treatment also develop anti-PEG antibodies. Sex differences in bone formation and immune cell recruitment are also apparent. Overall, these findings indicate that anti-PEG immune responses can impact tissue engineering efficacy and highlight the need for further investigation.
Collapse
Affiliation(s)
- Alisa H Isaac
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, USA
| | | | - Elizabeth Ruben
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Matthew Estes
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Varsha Rajavel
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Talia Baig
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Carol Paleti
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University, College Station, TX, USA
| | - Kirsten Landsgaard
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Ryang Hwa Lee
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University, College Station, TX, USA
| | - Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX, USA
- Department of Cell Systems and Anatomy, The University of Texas Health San Antonio, San Antonio, TX, USA
| | - Michael F Criscitiello
- Comparative Immunogenetics Laboratory, Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, USA
| | - Carl Gregory
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University, College Station, TX, USA
| | - Daniel L Alge
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Materials Science and Engineering, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
9
|
Bracken SJ, Suthers AN, DiCioccio RA, Su H, Anand S, Poe JC, Jia W, Visentin J, Basher F, Jordan CZ, McManigle WC, Li Z, Hakim FT, Pavletic SZ, Bhuiya NS, Ho VT, Horwitz ME, Chao NJ, Sarantopoulos S. Heightened TLR7 signaling primes BCR-activated B cells in chronic graft-versus-host disease for effector functions. Blood Adv 2024; 8:667-680. [PMID: 38113462 PMCID: PMC10839617 DOI: 10.1182/bloodadvances.2023010362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 11/02/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
ABSTRACT Chronic graft-versus-host disease (cGVHD) is a debilitating, autoimmune-like syndrome that can occur after allogeneic hematopoietic stem cell transplantation. Constitutively activated B cells contribute to ongoing alloreactivity and autoreactivity in patients with cGVHD. Excessive tissue damage that occurs after transplantation exposes B cells to nucleic acids in the extracellular environment. Recognition of endogenous nucleic acids within B cells can promote pathogenic B-cell activation. Therefore, we hypothesized that cGVHD B cells aberrantly signal through RNA and DNA sensors such as Toll-like receptor 7 (TLR7) and TLR9. We found that B cells from patients and mice with cGVHD had higher expression of TLR7 than non-cGVHD B cells. Using ex vivo assays, we found that B cells from patients with cGVHD also demonstrated increased interleukin-6 production after TLR7 stimulation with R848. Low-dose B-cell receptor (BCR) stimulation augmented B-cell responses to TLR7 activation. TLR7 hyperresponsiveness in cGVHD B cells correlated with increased expression and activation of the downstream transcription factor interferon regulatory factor 5. Because RNA-containing immune complexes can activate B cells through TLR7, we used a protein microarray to identify RNA-containing antigen targets of potential pathological relevance in cGVHD. We found that many of the unique targets of active cGVHD immunoglobulin G (IgG) were nucleic acid-binding proteins. This unbiased assay identified the autoantigen and known cGVHD target Ro-52, and we found that RNA was required for IgG binding to Ro-52. Herein, we find that BCR-activated B cells have aberrant TLR7 signaling responses that promote potential effector responses in cGVHD.
Collapse
Affiliation(s)
- Sonali J. Bracken
- Division of Rheumatology and Immunology, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Amy N. Suthers
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Rachel A. DiCioccio
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Hsuan Su
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Sarah Anand
- Division of Hematology and Medical Oncology, Department of Medicine, University of Michigan, Ann Arbor, MI
| | - Jonathan C. Poe
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Wei Jia
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Jonathan Visentin
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
- Department of Immunology and Immunogenetics, Bordeaux University Hospital, Bordeaux, France
- UMR CNRS 5164 ImmunoConcEpT, Bordeaux University, Bordeaux, France
| | - Fahmin Basher
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Collin Z. Jordan
- Division of Nephrology, Department of Medicine, Duke University Medical Center, Durham NC
| | - William C. McManigle
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Duke University Medical Center, Durham NC
| | - Zhiguo Li
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham NC
- Duke Cancer Institute, Duke University Medical Center, Durham NC
| | - Frances T. Hakim
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Bethesda, MD
| | - Steven Z. Pavletic
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Bethesda, MD
| | - Nazmim S. Bhuiya
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Bethesda, MD
| | - Vincent T. Ho
- Division of Hematologic Malignancies and Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Mitchell E. Horwitz
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
- Duke Cancer Institute, Duke University Medical Center, Durham NC
| | - Nelson J. Chao
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
- Duke Cancer Institute, Duke University Medical Center, Durham NC
- Department of Integrated Immunobiology, Duke University School of Medicine, Durham, NC
| | - Stefanie Sarantopoulos
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC
- Duke Cancer Institute, Duke University Medical Center, Durham NC
- Department of Integrated Immunobiology, Duke University School of Medicine, Durham, NC
| |
Collapse
|
10
|
Liu N, Zhou R, Tam AR, Chu WM, Yang D, Huang H, Peng Q, Du Z, Chen B, To KKW, Chen Z. Immune correlates with disease severity in older patients after SARS-CoV-2 BA.2 infection in Hong Kong. Clin Immunol 2024; 259:109900. [PMID: 38218212 DOI: 10.1016/j.clim.2024.109900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Affiliation(s)
- Na Liu
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China
| | - Runhong Zhou
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China.
| | - Anthony Raymond Tam
- Department of Medicine, Queen Mary Hospital, Hong Kong Special Administrative Region, People's Republic of China
| | - Wing-Ming Chu
- Department of Medicine, Queen Mary Hospital, Hong Kong Special Administrative Region, People's Republic of China
| | - Dawei Yang
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Haode Huang
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Qiaoli Peng
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Zhenglong Du
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Bohao Chen
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Kelvin Kai-Wang To
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China; State Key Laboratory for Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China
| | - Zhiwei Chen
- AIDS Institute, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China.; Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, People's Republic of China; State Key Laboratory for Emerging Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, People's Republic of China.
| |
Collapse
|
11
|
Wijaya LK, Stumbles PA, Finch PM, Drummond PD. Inflammation induces α 1-adrenoceptor expression in peripheral blood mononuclear cells of patients with complex regional pain syndrome. Brain Behav Immun 2024; 115:201-208. [PMID: 37848097 DOI: 10.1016/j.bbi.2023.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/09/2023] [Accepted: 10/14/2023] [Indexed: 10/19/2023] Open
Abstract
Persistent regional and systemic inflammation may promote pain and hyperalgesia in complex regional pain syndrome (CRPS). In this study, we investigated whether stimulation of α1-adrenoceptors (α1-AR) on peripheral blood mononuclear cells (PBMC) might contribute to this inflammatory state. PBMC were isolated from venous blood collected from 21 CRPS patients and 21 sex and age-matched controls. Lipopolysaccharide (LPS), a bacterial toxin, was administered to cultured PBMC for 24 h to trigger inflammation. Exposure to LPS resulted in heightened gene expression of α1-AR subtype B (α1B-AR) in PBMC of CRPS patients relative to controls. Interleukin (IL)-1β and IL-6 levels did not change when the α1-AR agonist phenylephrine was administered to naïve PBMC. However, α1-AR stimulation following LPS treatment increased IL-6 mRNA and protein levels in PBMC of patients and controls. To investigate the possible consequence of heightened IL-6 levels on immunoglobulin G antibody production, PBMC were stimulated with CD40 ligand and IL-21 to generate plasmablasts (B cells that secrete antibodies). This response was similar in patients and controls. Adding IL-6 to the cell culture medium increased plasmablast differentiation in controls and antibody production both in patients and controls. These findings suggest that the inflammatory cascade associated with elevated levels of IL-6 may generate α1B-AR expression in CRPS PBMC. A reciprocal interaction between heightened α1-AR expression in PBMC and IL-6 secretion may contribute to systemic inflammation and antibody production in CRPS.
Collapse
Affiliation(s)
- Linda K Wijaya
- School of Psychology, College of Health and Education, Murdoch University, 90 South St, Perth, Western Australia 6150, Australia; Telethon Kids Institute, Perth Children Hospital, 15 Hospital Avenue, Perth, Western Australia 6009, Australia.
| | - Philip A Stumbles
- School of Psychology, College of Health and Education, Murdoch University, 90 South St, Perth, Western Australia 6150, Australia; Telethon Kids Institute, Perth Children Hospital, 15 Hospital Avenue, Perth, Western Australia 6009, Australia.
| | - Philip M Finch
- School of Psychology, College of Health and Education, Murdoch University, 90 South St, Perth, Western Australia 6150, Australia.
| | - Peter D Drummond
- School of Psychology, College of Health and Education, Murdoch University, 90 South St, Perth, Western Australia 6150, Australia.
| |
Collapse
|
12
|
Sawada K, Chung H, Softic S, Moreno-Fernandez ME, Divanovic S. The bidirectional immune crosstalk in metabolic dysfunction-associated steatotic liver disease. Cell Metab 2023; 35:1852-1871. [PMID: 37939656 PMCID: PMC10680147 DOI: 10.1016/j.cmet.2023.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is an unabated risk factor for end-stage liver diseases with no available therapies. Dysregulated immune responses are critical culprits of MASLD pathogenesis. Independent contributions from either the innate or adaptive arms of the immune system or their unidirectional interplay are commonly studied in MASLD. However, the bidirectional communication between innate and adaptive immune systems and its impact on MASLD remain insufficiently understood. Given that both innate and adaptive immune cells are indispensable for the development and progression of inflammation in MASLD, elucidating pathogenic contributions stemming from the bidirectional interplay between these two arms holds potential for development of novel therapeutics for MASLD. Here, we review the immune cell types and bidirectional pathways that influence the pathogenesis of MASLD and highlight potential pharmacologic approaches to combat MASLD based on current knowledge of this bidirectional crosstalk.
Collapse
Affiliation(s)
- Keisuke Sawada
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Hak Chung
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Samir Softic
- Department of Pediatrics and Gastroenterology, University of Kentucky, Lexington, KY 40536, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Maria E Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
13
|
Chu E, Mychasiuk R, Green TRF, Zamani A, Dill LK, Sharma R, Raftery AL, Tsantikos E, Hibbs ML, Semple BD. Regulation of microglial responses after pediatric traumatic brain injury: exploring the role of SHIP-1. Front Neurosci 2023; 17:1276495. [PMID: 37901420 PMCID: PMC10603304 DOI: 10.3389/fnins.2023.1276495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 09/18/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Severe traumatic brain injury (TBI) is the world's leading cause of permanent neurological disability in children. TBI-induced neurological deficits may be driven by neuroinflammation post-injury. Abnormal activity of SH2 domain-containing inositol 5' phosphatase-1 (SHIP-1) has been associated with dysregulated immunological responses, but the role of SHIP-1 in the brain remains unclear. The current study investigated the immunoregulatory role of SHIP-1 in a mouse model of moderate-severe pediatric TBI. Methods SHIP-1+/- and SHIP-1-/- mice underwent experimental TBI or sham surgery at post-natal day 21. Brain gene expression was examined across a time course, and immunofluorescence staining was evaluated to determine cellular immune responses, alongside peripheral serum cytokine levels by immunoassays. Brain tissue volume loss was measured using volumetric analysis, and behavior changes both acutely and chronically post-injury. Results Acutely, inflammatory gene expression was elevated in the injured cortex alongside increased IBA-1 expression and altered microglial morphology; but to a similar extent in SHIP-1-/- mice and littermate SHIP-1+/- control mice. Similarly, the infiltration and activation of CD68-positive macrophages, and reactivity of GFAP-positive astrocytes, was increased after TBI but comparable between genotypes. TBI increased anxiety-like behavior acutely, whereas SHIP-1 deficiency alone reduced general locomotor activity. Chronically, at 12-weeks post-TBI, SHIP-1-/- mice exhibited reduced body weight and increased circulating cytokines. Pro-inflammatory gene expression in the injured hippocampus was also elevated in SHIP-1-/- mice; however, GFAP immunoreactivity at the injury site in TBI mice was lower. TBI induced a comparable loss of cortical and hippocampal tissue in both genotypes, while SHIP-1-/- mice showed reduced general activity and impaired working memory, independent of TBI. Conclusion Together, evidence does not support SHIP-1 as an essential regulator of brain microglial morphology, brain immune responses, or the extent of tissue damage after moderate-severe pediatric TBI in mice. However, our data suggest that reduced SHIP-1 activity induces a greater inflammatory response in the hippocampus chronically post-TBI, warranting further investigation.
Collapse
Affiliation(s)
- Erskine Chu
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
- Deparment of Neurology, Alfred Health, Prahran, VIC, Australia
| | - Tabitha R. F. Green
- Department of Integrative Physiology, The University of Colorado Boulder, Boulder, CO, United States
| | - Akram Zamani
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Larissa K. Dill
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
- Alfred Health, Prahran, VIC, Australia
| | - Rishabh Sharma
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - April L. Raftery
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Evelyn Tsantikos
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Margaret L. Hibbs
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Bridgette D. Semple
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
- Deparment of Neurology, Alfred Health, Prahran, VIC, Australia
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
14
|
Zhang L, More KR, Ojha A, Jackson CB, Quinlan BD, Li H, He W, Farzan M, Pardi N, Choe H. Effect of mRNA-LNP components of two globally-marketed COVID-19 vaccines on efficacy and stability. NPJ Vaccines 2023; 8:156. [PMID: 37821446 PMCID: PMC10567765 DOI: 10.1038/s41541-023-00751-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/25/2023] [Indexed: 10/13/2023] Open
Abstract
During the COVID-19 pandemic, Pfizer-BioNTech and Moderna successfully developed nucleoside-modified mRNA lipid nanoparticle (LNP) vaccines. SARS-CoV-2 spike protein expressed by those vaccines are identical in amino acid sequence, but several key components are distinct. Here, we compared the effect of ionizable lipids, untranslated regions (UTRs), and nucleotide composition of the two vaccines, focusing on mRNA delivery, antibody generation, and long-term stability. We found that the ionizable lipid, SM-102, in Moderna's vaccine performs better than ALC-0315 in Pfizer-BioNTech's vaccine for intramuscular delivery of mRNA and antibody production in mice and long-term stability at 4 °C. Moreover, Pfizer-BioNTech's 5' UTR and Moderna's 3' UTR outperform their counterparts in their contribution to transgene expression in mice. We further found that varying N1-methylpseudouridine content at the wobble position of mRNA has little effect on vaccine efficacy. These findings may contribute to the further improvement of nucleoside-modified mRNA-LNP vaccines and therapeutics.
Collapse
Affiliation(s)
- Lizhou Zhang
- Division of Infectious Disease, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA.
| | - Kunal R More
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Amrita Ojha
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Cody B Jackson
- Division of Infectious Disease, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Brian D Quinlan
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Hao Li
- Division of Infectious Disease, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
- Skaggs Graduate School, The Scripps Research Institute, La Jolla, CA, USA
| | - Wenhui He
- Division of Infectious Disease, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
- Center For Integrated Solutions for Infectious Diseases, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael Farzan
- Division of Infectious Disease, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
- Skaggs Graduate School, The Scripps Research Institute, La Jolla, CA, USA
- Center For Integrated Solutions for Infectious Diseases, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hyeryun Choe
- Division of Infectious Disease, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
- Department of Immunology and Microbiology, UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA.
| |
Collapse
|
15
|
Chunder R, Schropp V, Marzin M, Amor S, Kuerten S. A Dual Role of Osteopontin in Modifying B Cell Responses. Biomedicines 2023; 11:1969. [PMID: 37509608 PMCID: PMC10377065 DOI: 10.3390/biomedicines11071969] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
The occurrence of B cell aggregates within the central nervous system (CNS) has prompted the investigation of the potential sources of pathogenic B cell and T cell responses in a subgroup of secondary progressive multiple sclerosis (MS) patients. Nevertheless, the expression profile of molecules associated with these aggregates and their role in aggregate development and persistence is poorly described. Here, we focused on the expression pattern of osteopontin (OPN), which is a well-described cytokine, in MS brain tissue. Autopsied brain sections from MS cases with and without B cell pathology were screened for the presence of CD20+ B cell aggregates and co-expression of OPN. To demonstrate the effect of OPN on B cells, flow cytometry, ELISA and in vitro aggregation assays were conducted using the peripheral blood of healthy volunteers. Although OPN was expressed in MS brain tissue independent of B cell pathology, it was also highly expressed within B cell aggregates. In vitro studies demonstrated that OPN downregulated the co-stimulatory molecules CD80 and CD86 on B cells. OPN-treated B cells produced significantly lower amounts of IL-6. However, OPN-treated B cells also exhibited a higher tendency to form homotypic cell aggregates in vitro. Taken together, our data indicate a conflicting role of OPN in modulating B cell responses.
Collapse
Affiliation(s)
- Rittika Chunder
- Faculty of Medicine, Institute of Neuroanatomy, University of Bonn, 53115 Bonn, Germany
- University Hospital Bonn, 53127 Bonn, Germany
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Verena Schropp
- Faculty of Medicine, Institute of Neuroanatomy, University of Bonn, 53115 Bonn, Germany
- University Hospital Bonn, 53127 Bonn, Germany
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Manuel Marzin
- Department of Pathology, Amsterdam University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Sandra Amor
- Department of Pathology, Amsterdam University Medical Center, 1007 MB Amsterdam, The Netherlands
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Stefanie Kuerten
- Faculty of Medicine, Institute of Neuroanatomy, University of Bonn, 53115 Bonn, Germany
- University Hospital Bonn, 53127 Bonn, Germany
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
16
|
Pan C, Wang L, Zhang M, Li J, Liu J, Liu J. In Situ Polymerization-Mediated Antigen Presentation. J Am Chem Soc 2023. [PMID: 37262440 DOI: 10.1021/jacs.3c02682] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Activating antigen-presenting cells is essential to generate adaptive immunity, while the efficacy of conventional activation strategies remains unsatisfactory due to suboptimal antigen-specific priming. Here, in situ polymerization-mediated antigen presentation (IPAP) is described, in which antigen-loaded nanovaccines are spontaneously formed and efficiently anchored onto the surface of dendritic cells in vivo through co-deposition with dopamine. The resulting chemically bound nanovaccines can promote antigen presentation by elevating macropinocytosis-based cell uptake and reducing lysosome-related antigen degradation. IPAP is able to prolong the duration of antigen reservation in the injection site and enhance subsequent accumulation in the draining lymph nodes, thereby eliciting robust antigen-specific cellular and humoral immune responses. IPAP is also applicable for different antigens and capable of circumventing the disadvantages of complicated preparation and purification. By implementation with ovalbumin, IPAP induces a significant protective immunity against ovalbumin-overexpressing tumor cell challenge in a prophylactic murine model. The use of the SARS-CoV-2 Spike protein S1 subunit also remarkably increases the production of S1-specific immunoglobulin G in mice. IPAP offers a unique strategy for stimulating antigen-presenting cells to boost antigen-specific adaptive responses and proposes a facile yet versatile method for immunization against various diseases.
Collapse
Affiliation(s)
- Chao Pan
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lu Wang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Mengmeng Zhang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Juanjuan Li
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Junqiu Liu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
17
|
Wang Y, Jin X, Li M, Gao J, Zhao X, Ma J, Shi C, He B, Hu L, Shi J, Liu G, Qu G, Zheng Y, Jiang G. PM 2.5 Increases Systemic Inflammatory Cells and Associated Disease Risks by Inducing NRF2-Dependent Myeloid-Biased Hematopoiesis in Adult Male Mice. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:7924-7937. [PMID: 37184982 DOI: 10.1021/acs.est.2c09024] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Although PM2.5 (fine particles with aerodynamic diameter <2.5 μm) exposure shows the potential to impact normal hematopoiesis, the detailed alterations in systemic hematopoiesis and the underlying mechanisms remain unclear. For hematopoiesis under steady-state or stress conditions, nuclear factor erythroid 2-related factor 2 (NRF2) is essential for regulating hematopoietic processes to maintain blood homeostasis. Herein, we characterized changes in the populations of hematopoietic stem progenitor cells and committed hematopoietic progenitors in the lungs and bone marrow (BM) of wild-type and Nrf2-/- C57BL/6J male mice. PM2.5-induced NRF2-dependent biased hematopoiesis toward myeloid lineage in the lungs and BM generates excessive numbers of various inflammatory immune cells, including neutrophils, monocytes, and platelets. The increased population of these immune cells in the lungs, BM, and peripheral blood has been associated with observed pulmonary fibrosis and high disease risks in an NRF2-dependent manner. Therefore, although NRF2 is a protective factor against stressors, upon PM2.5 exposure, NRF2 is involved in stress myelopoiesis and enhanced PM2.5 toxicity in pulmonary injury, even leading to systemic inflammation.
Collapse
Affiliation(s)
- Yuanyuan Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoting Jin
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Min Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, China
| | - Jie Gao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environmental, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310000, China
| | - Xingchen Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Juan Ma
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunzhen Shi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Ecology and Environment, Beijing Technology and Business University, Beijing 100048, China
| | - Bin He
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environmental, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310000, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ligang Hu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environmental, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310000, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Jianbo Shi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environmental, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310000, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Guoliang Liu
- Department of Pulmonary and Critical Care Medicine, National Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China
- Institute of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Chinese Academy of Medical Sciences, Beijing 100029, China
| | - Guangbo Qu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environmental, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310000, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| | - Yuxin Zheng
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- School of Environmental, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310000, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Environment and Health, Jianghan University, Wuhan 430056, China
| |
Collapse
|
18
|
Abnousian A, Vasquez J, Sasaninia K, Kelley M, Venketaraman V. Glutathione Modulates Efficacious Changes in the Immune Response against Tuberculosis. Biomedicines 2023; 11:biomedicines11051340. [PMID: 37239011 DOI: 10.3390/biomedicines11051340] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 04/27/2023] [Accepted: 04/30/2023] [Indexed: 05/28/2023] Open
Abstract
Glutathione (GSH) is an antioxidant in human cells that is utilized to prevent damage occurred by reactive oxygen species, free radicals, peroxides, lipid peroxides, and heavy metals. Due to its immunological role in tuberculosis (TB), GSH is hypothesized to play an important part in the immune response against M. tb infection. In fact, one of the hallmark structures of TB is granuloma formation, which involves many types of immune cells. T cells, specifically, are a major component and are involved in the release of cytokines and activation of macrophages. GSH also serves an important function in macrophages, natural killer cells, and T cells in modulating their activation, their metabolism, proper cytokine release, proper redox activity, and free radical levels. For patients with increased susceptibility, such as those with HIV and type 2 diabetes, the demand for higher GSH levels is increased. GSH acts as an important immunomodulatory antioxidant by stabilizing redox activity, shifting of cytokine profile toward Th1 type response, and enhancing T lymphocytes. This review compiles reports showing the benefits of GSH in improving the immune responses against M. tb infection and the use of GSH as an adjunctive therapy for TB.
Collapse
Affiliation(s)
- Arbi Abnousian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Joshua Vasquez
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Kayvan Sasaninia
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Melissa Kelley
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91768, USA
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
19
|
Qiao Y, Hu H, Zhao Y, Jin M, Yang D, Yin J, Wu P, Liu W, Li J. Benzene induces spleen injury through the B cell receptor signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 257:114924. [PMID: 37080132 DOI: 10.1016/j.ecoenv.2023.114924] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 03/13/2023] [Accepted: 04/16/2023] [Indexed: 05/03/2023]
Abstract
Benzene is a toxic environmental pollutant that disrupts the immune system in humans. Benzene exposure reduces the abundance of immune cells in multiple immune organs; however, the biological mechanisms underlying benzene-induced immunotoxicity has not been elucidated. In this study, benzene was used to develop mouse model for immune dysfunction. A significant decrease in IgG, IL-2 and IL-6 levels, an increase in oxidative stress and spleen injury were observed after benzene exposure in a dose-dependent manner. Quantitative proteomics revealed that benzene-induced immune dysfunction was associated with deregulation of the B cell receptor (BCR) signaling pathway. Benzene exposure suppressed the expression of CD22, BCL10 and NF-κb p65. Also, a significant decrease in proliferation and an increase in apoptosis of splenic lymphocytes were found after benzene exposure. Moreover, we found that benzene exposure increased mitochondrial reactive oxygen species (mito-ROS) and decreased adenosine triphosphate (ATP). Overall, we revealed the damaging effects of benzene on spleen-related immune function and the underlying biological mechanism, involving the disruption of BCR signaling pathway, NF-κB deactivation, and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Yamei Qiao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Hui Hu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China; Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Yunyan Zhao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Min Jin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Dong Yang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Jing Yin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Peng Wu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.
| | - Weili Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China.
| | - Junwen Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China.
| |
Collapse
|
20
|
Chauvin C, Levillayer L, Roumier M, Nielly H, Roth C, Karnam A, Bonam SR, Bourgarit A, Dubost C, Bousquet A, Le Burel S, Mestiri R, Sene D, Galland J, Vasse M, Groh M, Le Marchand M, Vassord-Dang C, Gautier JF, Pham-Thi N, Verny C, Pitard B, Planchais C, Mouquet H, Paul R, Simon-Loriere E, Bayry J, Gilardin L, Sakuntabhai A. Tocilizumab-treated convalescent COVID-19 patients retain the cross-neutralization potential against SARS-CoV-2 variants. iScience 2023; 26:106124. [PMID: 36776936 PMCID: PMC9894676 DOI: 10.1016/j.isci.2023.106124] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/10/2022] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Although tocilizumab treatment in severe and critical coronavirus disease 2019 (COVID-19) patients has proven its efficacy at the clinical level, there is little evidence supporting the effect of short-term use of interleukin-6 receptor blocking therapy on the B cell sub-populations and the cross-neutralization of SARS-CoV-2 variants in convalescent COVID-19 patients. We performed immunological profiling of 69 tocilizumab-treated and non-treated convalescent COVID-19 patients in total. We observed that SARS-CoV-2-specific IgG1 titers depended on disease severity but not on tocilizumab treatment. The plasma of both treated and non-treated patients infected with the ancestral variant exhibit strong neutralizing activity against the ancestral virus and the Alpha, Beta, and Delta variants of SARS-CoV-2, whereas the Gamma and Omicron viruses were less sensitive to seroneutralization. Overall, we observed that, despite the clinical benefits of short-term tocilizumab therapy in modifying the cytokine storm associated with COVID-19 infections, there were no modifications in the robustness of B cell and IgG responses to Spike antigens.
Collapse
Affiliation(s)
- Camille Chauvin
- Institut Pasteur, Université de Paris, Functional Genetics of Infectious Diseases Unit, Department of Global Health, 75015 Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France
| | - Laurine Levillayer
- Institut Pasteur, Université de Paris, Functional Genetics of Infectious Diseases Unit, Department of Global Health, 75015 Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France
| | - Mathilde Roumier
- Service de Médecine Interne, Hôpital Foch, 92151 Suresnes, France
| | - Hubert Nielly
- Service de Médecine Interne, Hôpital d'Instruction des Armées Bégin, 94160 Saint Mandé, France
| | - Claude Roth
- Institut Pasteur, Université de Paris, Functional Genetics of Infectious Diseases Unit, Department of Global Health, 75015 Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France
| | - Anupama Karnam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, 75006, France
| | - Srinivasa Reddy Bonam
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, 75006, France
| | - Anne Bourgarit
- Hôpital Jean Verdier, HUPSSD, AP-HP, 93140 Bondy, France.,Sorbonne Paris-Nord University (Paris 13), 93000 Bobigny, France.,Inserm, UMR 1135 CIMI, 75013 Paris, France
| | - Clément Dubost
- Service de réanimation, Hôpital militaire Bégin, 94120 Saint Mandé, France.,Université Paris-Saclay, ENS Paris-Saclay, CNRS, Centre Borelli, 91190, Gif-sur-Yvette, France
| | - Aurore Bousquet
- Département des laboratoires, Hôpital militaire Bégin, 94120 Saint Mandé, France
| | - Sébastien Le Burel
- Service de Médecine Interne, Hôpital d'Instruction des Armées Bégin, 94160 Saint Mandé, France
| | - Raphaële Mestiri
- Service de Médecine Interne, Hôpital d'Instruction des Armées Bégin, 94160 Saint Mandé, France
| | - Damien Sene
- Département de médecine interne, Hôpital Lariboisière, Université de Paris (Diderot), AP-HP, 75010 Paris, France
| | - Joris Galland
- Département de médecine interne, Hôpital Lariboisière, Université de Paris (Diderot), AP-HP, 75010 Paris, France
| | - Marc Vasse
- Laboratoire de Biologie Médicale, Hôpital Foch, 92151 Suresnes, France.,UMRS-1176, Le Kremlin Bicêtre, France
| | - Matthieu Groh
- Service de Médecine Interne, Hôpital Foch, 92151 Suresnes, France
| | - Mathilde Le Marchand
- Department of Clinical Research and Innovation, Foch Hospital, 40 rue Worth, 92150 Suresnes, France
| | - Camille Vassord-Dang
- Department of Clinical Research and Innovation, Foch Hospital, 40 rue Worth, 92150 Suresnes, France
| | - Jean-François Gautier
- Departement of Diabetes and Endocrinology, Hôpital Lariboisière, APHP, and INSERM U1138 Paris, France.,Université de Paris, 75006 Paris, France
| | - Nhan Pham-Thi
- Unité de Neurophysiologie du Stress, Département des Neurosciences, Institut de Recherche Biomédicale des Armées (IRBA), BP 73 91223 Brétigny sur Orge Cedex, France
| | - Christiane Verny
- Unité de Neurophysiologie du Stress, Département des Neurosciences, Institut de Recherche Biomédicale des Armées (IRBA), BP 73 91223 Brétigny sur Orge Cedex, France
| | - Bruno Pitard
- Nantes Université, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT,UMR 1302, F-44000 Nantes, France
| | - Cyril Planchais
- Institut Pasteur, Université de Paris, Humoral Immunology Unit, Department of Immunology, 75015 Paris, France
| | - Hugo Mouquet
- Institut Pasteur, Université de Paris, Humoral Immunology Unit, Department of Immunology, 75015 Paris, France
| | - Richard Paul
- Institut Pasteur, Université de Paris, Functional Genetics of Infectious Diseases Unit, Department of Global Health, 75015 Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France
| | - Etienne Simon-Loriere
- Institut Pasteur, Université de Paris, G5 Evolutionary Genomics of RNA viruses, 75015 Paris, France
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, 75006, France.,Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad 678623, India
| | - Laurent Gilardin
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, 75006, France.,Sorbonne Paris-Nord University (Paris 13), 93000 Bobigny, France.,Service de médecine interne, Hôpital Jean Verdier, HUPSSD, AP-HP, 93140 Bondy, France
| | - Anavaj Sakuntabhai
- Institut Pasteur, Université de Paris, Functional Genetics of Infectious Diseases Unit, Department of Global Health, 75015 Paris, France.,Centre National de la Recherche Scientifique (CNRS), UMR2000, Paris Cedex 15, France.,International Vaccine Design Center (vDesC), The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, Japan
| |
Collapse
|
21
|
Huard A, Wilmes C, Kiprina A, Netzer C, Palmer G, Brüne B, Weigert A. Cell Intrinsic IL-38 Affects B Cell Differentiation and Antibody Production. Int J Mol Sci 2023; 24:ijms24065676. [PMID: 36982750 PMCID: PMC10053218 DOI: 10.3390/ijms24065676] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/18/2023] Open
Abstract
IL-38 is an IL-1 family receptor antagonist with an emerging role in chronic inflammatory diseases. IL-38 expression has been mainly observed not only in epithelia, but also in cells of the immune system, including macrophages and B cells. Given the association of both IL-38 and B cells with chronic inflammation, we explored if IL-38 affects B cell biology. IL-38-deficient mice showed higher amounts of plasma cells (PC) in lymphoid organs but, conversely, lower levels of plasmatic antibody titers. Exploring underlying mechanisms in human B cells revealed that exogenously added IL-38 did not significantly affect early B cell activation or differentiation into plasma cells, even though IL-38 suppressed upregulation of CD38. Instead, IL-38 mRNA expression was transiently upregulated during the differentiation of human B cells to plasma cells in vitro, and knocking down IL-38 during early B cell differentiation increased plasma cell generation, while reducing antibody production, thus reproducing the murine phenotype. Although this endogenous role of IL-38 in B cell differentiation and antibody production did not align with an immunosuppressive function, autoantibody production induced in mice by repeated IL-18 injections was enhanced in an IL-38-deficient background. Taken together, our data suggest that cell-intrinsic IL-38 promotes antibody production at baseline but suppresses the production of autoantibodies in an inflammatory context, which may partially explain its protective role during chronic inflammation.
Collapse
Affiliation(s)
- Arnaud Huard
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Christian Wilmes
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Anastasiia Kiprina
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Christoph Netzer
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Gaby Palmer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
- Geneva Center for Inflammation Research, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
- Cardio-Pulmonary Institute (CPI), 60590 Frankfurt, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
- Cardio-Pulmonary Institute (CPI), 60590 Frankfurt, Germany
- Correspondence: ; Tel.: +49-69-6301-4593; Fax: +49-69-6301-420
| |
Collapse
|
22
|
Yankowski C, Kurup D, Wirblich C, Schnell MJ. Effects of adjuvants in a rabies-vectored Ebola virus vaccine on protection from surrogate challenge. NPJ Vaccines 2023; 8:10. [PMID: 36754965 PMCID: PMC9906604 DOI: 10.1038/s41541-023-00615-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 01/27/2023] [Indexed: 02/10/2023] Open
Abstract
Ebola virus is the primary contributor to the global threat of filovirus severe hemorrhagic fever, and Ebola virus disease has a case fatality rate of 50-90%. An inactivated, bivalent filovirus/rabies virus vaccine, FILORAB1, consists of recombinant rabies virus virions expressing the Ebola virus glycoprotein. FILORAB1 is immunogenic and protective from Ebola virus challenge in mice and non-human primates, and protection is enhanced when formulated with toll-like receptor 4 agonist Glucopyranosyl lipid adjuvant (GLA) in a squalene oil-in-water emulsion (SE). Through an adjuvant comparison in mice, we demonstrate that GLA-SE improves FILORAB1 efficacy by activating the innate immune system and shaping a Th1-biased adaptive immune response. GLA-SE adjuvanted mice and those adjuvanted with the SE component are better protected from surrogate challenge, while Th2 alum adjuvanted mice are not. Additionally, the immune response to FILORAB1 is long-lasting, as exhibited by highly-maintained serum antibody titers and long-lived cells in the spleen and bone marrow.
Collapse
Affiliation(s)
- Catherine Yankowski
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Drishya Kurup
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson Vaccine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Christoph Wirblich
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Matthias J Schnell
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA.
- Jefferson Vaccine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
23
|
Baruah N, Ahamad N, Halder P, Koley H, Katti DS. Facile synthesis of multi-faceted, biomimetic and cross-protective nanoparticle-based vaccines for drug-resistant Shigella: a flexible platform technology. J Nanobiotechnology 2023; 21:34. [PMID: 36710326 PMCID: PMC9884485 DOI: 10.1186/s12951-023-01780-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 01/12/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND No commercial vaccines are available against drug-resistant Shigella due to serotype-specific/narrow-range of protection. Nanoparticle-based biomimetic vaccines involving stable, conserved, immunogenic proteins fabricated using facile chemistries can help formulate a translatable cross-protective Shigella vaccine. Such systems can also negate cold-chain transportation/storage thus overcoming challenges prevalent in various settings. METHODS We explored facile development of biomimetic poly (lactide-co-glycolide)/PLGA 50:50 based nanovaccines (NVs), encapsulating conserved stabilized antigen(s)/immunostimulant of S. dysenteriae 1 origin surface-modified using simple chemistries. All encapsulants (IpaC/IpaB/LPS) and nanoparticles (NPs)-bare and modified (NV), were thoroughly characterized. Effect of IpaC on cellular uptake of NPs was assessed in-vitro. Immunogenicity of the NVs was assessed in-vivo in BALB/c mice by intranasal immunization. Cross-protective efficacy was assessed by intraperitoneally challenging the immunized groups with a high dose of heterologous S. flexneri 2a and observing for visible diarrhea, weight loss and survival. Passive-protective ability of the simplest NV was assessed in the 5-day old progeny of vaccinated mice. RESULTS All the antigens and immunostimulant to be encapsulated were successfully purified and found to be stable both before and after encapsulation into NPs. The ~ 300 nm sized NPs with a zeta potential of ~ - 25 mV released ~ 60% antigen by 14th day suggesting an appropriate delivery kinetics. The NPs could be successfully surface-modified with IpaC and/or CpG DNA. In vitro experiments revealed that the presence of IpaC can significantly increase cellular uptake of NPs. All NVs were found to be cytocompatible and highly immunogenic. Antibodies in sera of NV-immunized mice could recognize heterologous Shigella. Immunized sera also showed high antibody and cytokine response. The immunized groups were protected from diarrhea and weight loss with ~ 70-80% survival upon heterologous Shigella challenge. The simplest NV showed ~ 88% survival in neonates. CONCLUSIONS Facile formulation of biomimetic NVs can result in significant cross-protection. Further, passive protection in neonates suggest that parental immunization could protect infants, the most vulnerable group in context of Shigella infection. Non-invasive route of vaccination can also lead to greater patient compliance making it amenable for mass-immunization. Overall, our work contributes towards a yet to be reported platform technology for facile development of cross-protective Shigella vaccines.
Collapse
Affiliation(s)
- Namrata Baruah
- grid.417965.80000 0000 8702 0100Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016 Uttar Pradesh India ,grid.417965.80000 0000 8702 0100The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, 208016 Uttar Pradesh India
| | - Nadim Ahamad
- grid.417965.80000 0000 8702 0100Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016 Uttar Pradesh India
| | - Prolay Halder
- grid.419566.90000 0004 0507 4551Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, 700010 West Bengal India
| | - Hemanta Koley
- grid.419566.90000 0004 0507 4551Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, 700010 West Bengal India
| | - Dhirendra S. Katti
- grid.417965.80000 0000 8702 0100Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016 Uttar Pradesh India ,grid.417965.80000 0000 8702 0100The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, 208016 Uttar Pradesh India
| |
Collapse
|
24
|
How an Immune-Factor-Based Formulation of Micro-Immunotherapy Could Interfere with the Physiological Processes Involved in the Atopic March. Int J Mol Sci 2023; 24:ijms24021483. [PMID: 36675006 PMCID: PMC9864899 DOI: 10.3390/ijms24021483] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Allergic diseases consist of improper inflammatory reactions to antigens and are currently an important healthcare concern, especially considering their increasing worldwide development in recent decades. The "atopic march" defines the paradigm of allergic diseases occurring in chronological order and displaying specific spatial manifestations, as they usually start as atopic dermatitis (AD) and food allergies during infancy and progressively evolve into allergic asthma (AA) and allergic rhinitis (AR) or rhino-conjunctivitis in childhood. Many immune cell subtypes and inflammatory factors are involved in these hypersensitivity reactions. In particular, the T helpers 2 (Th2) subset, through its cytokine signatures made of interleukins (ILs), such as IL-4, IL-5, IL-10, and IL-13, as well as mast cells and their related histamine pathways, contribute greatly to the perpetuation and evolution of the atopic march. By providing low doses (LD) and ultra-low doses (ULD) of ILs and immune factors to the body, micro-immunotherapy (MI) constitutes an interesting therapeutic strategy for the management of the atopic march and its symptoms. One of the aims of this review is to shed light on the current concept of the atopic march and the underlying immune reactions occurring during the IgE-mediated responses. Moreover, the different classes of traditional and innovative treatments employed in allergic diseases will also be discussed, with a special emphasis on the potential benefits of the MI medicine 2LALERG® formulation in this context.
Collapse
|
25
|
Taylor J, Gandhi A, Gray E, Zaenker P. Checkpoint inhibitor immune-related adverse events: A focused review on autoantibodies and B cells as biomarkers, advancements and future possibilities. Front Immunol 2023; 13:991433. [PMID: 36713389 PMCID: PMC9874109 DOI: 10.3389/fimmu.2022.991433] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 12/07/2022] [Indexed: 01/13/2023] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) has evolved rapidly with unprecedented treatment benefits being obtained for cancer patients, including improved patient survival. However, over half of the patients experience immune related adverse events (irAEs) or toxicities, which can be fatal, affect the quality of life of patients and potentially cause treatment interruption or cessation. Complications from these toxicities can also cause long term irreversible organ damage and other chronic health conditions. Toxicities can occur in various organ systems, with common observations in the skin, rheumatologic, gastrointestinal, hepatic, endocrine system and the lungs. These are not only challenging to manage but also difficult to detect during the early stages of treatment. Currently, no biomarker exists to predict which patients are likely to develop toxicities from ICI therapy and efforts to identify robust biomarkers are ongoing. B cells and antibodies against autologous antigens (autoantibodies) have shown promise and are emerging as markers to predict the development of irAEs in cancer patients. In this review, we discuss the interplay between ICIs and toxicities in cancer patients, insights into the underlying mechanisms of irAEs, and the involvement of the humoral immune response, particularly by B cells and autoantibodies in irAE development. We also provide an appraisal of the progress, key empirical results and advances in B cell and autoantibody research as biomarkers for predicting irAEs. We conclude the review by outlining the challenges and steps required for their potential clinical application in the future.
Collapse
Affiliation(s)
- John Taylor
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia,*Correspondence: John Taylor,
| | - Aesha Gandhi
- Sir Charles Gairdner Hospital, Department of Medical Oncology, Nedlands, WA, Australia
| | - Elin Gray
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Pauline Zaenker
- Centre for Precision Health, Edith Cowan University, Joondalup, WA, Australia,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| |
Collapse
|
26
|
Ho M, Xiao A, Yi D, Zanwar S, Bianchi G. Treating Multiple Myeloma in the Context of the Bone Marrow Microenvironment. Curr Oncol 2022; 29:8975-9005. [PMID: 36421358 PMCID: PMC9689284 DOI: 10.3390/curroncol29110705] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/08/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
The treatment landscape of multiple myeloma (MM) has evolved considerably with the FDA-approval of at least 15 drugs over the past two decades. Together with the use of autologous stem cell transplantation, these novel therapies have resulted in significant survival benefit for patients with MM. In particular, our improved understanding of the BM and immune microenvironment has led to the development of highly effective immunotherapies that have demonstrated unprecedented response rates even in the multiple refractory disease setting. However, MM remains challenging to treat especially in a high-risk setting. A key mediator of therapeutic resistance in MM is the bone marrow (BM) microenvironment; a deeper understanding is necessary to facilitate the development of therapies that target MM in the context of the BM milieu to elicit deeper and more durable responses with the ultimate goal of long-term control or a cure of MM. In this review, we discuss our current understanding of the role the BM microenvironment plays in MM pathogenesis, with a focus on its immunosuppressive nature. We also review FDA-approved immunotherapies currently in clinical use and highlight promising immunotherapeutic approaches on the horizon.
Collapse
Affiliation(s)
- Matthew Ho
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Alexander Xiao
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Dongni Yi
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Saurabh Zanwar
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Giada Bianchi
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02120, USA
| |
Collapse
|
27
|
Chunder R, Schropp V, Jabari S, Marzin M, Amor S, Kuerten S. Identification of a novel role for matrix metalloproteinase-3 in the modulation of B cell responses in multiple sclerosis. Front Immunol 2022; 13:1025377. [PMID: 36389698 PMCID: PMC9644161 DOI: 10.3389/fimmu.2022.1025377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/26/2022] [Indexed: 07/30/2023] Open
Abstract
There has been a growing interest in the presence and role of B cell aggregates within the central nervous system of multiple sclerosis patients. However, very little is known about the expression profile of molecules associated with these aggregates and how they might be influencing aggregate development or persistence in the brain. The current study focuses on the effect of matrix metalloproteinase-3, which is associated with B cell aggregates in autopsied multiple sclerosis brain tissue, on B cells. Autopsied brain sections from multiple sclerosis cases and controls were screened for the presence of CD20+ B cell aggregates and expression of matrix metalloproteinase-3. Using flow cytometry, enzyme-linked immunosorbent assay and gene array as methods, in vitro studies were conducted using peripheral blood of healthy volunteers to demonstrate the effect of matrix metalloproteinase-3 on B cells. Autopsied brain sections from multiple sclerosis patients containing aggregates of B cells expressed a significantly higher amount of matrix metalloproteinase-3 compared to controls. In vitro experiments demonstrated that matrix metalloproteinase-3 dampened the overall activation status of B cells by downregulating CD69, CD80 and CD86. Furthermore, matrix metalloproteinase-3-treated B cells produced significantly lower amounts of interleukin-6. Gene array data confirmed that matrix metalloproteinase-3 altered the proliferation and survival profiles of B cells. Taken together, out data indicate a role for B cell modulatory properties of matrix metalloproteinase-3.
Collapse
Affiliation(s)
- Rittika Chunder
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Verena Schropp
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Samir Jabari
- Institute of Neuropathology, University Hospitals Erlangen, Erlangen, Germany
| | - Manuel Marzin
- Department of Pathology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Sandra Amor
- Department of Pathology, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Stefanie Kuerten
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Anatomy and Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
28
|
Taylor JB, Malone-Povolny MJ, Merricks EP, Wimsey LE, Soliman D, Nichols TC, Wallet SM, Maile R, Schoenfisch MH. Mechanisms of Foreign Body Response Mitigation by Nitric Oxide Release. Int J Mol Sci 2022; 23:11635. [PMID: 36232937 PMCID: PMC9569454 DOI: 10.3390/ijms231911635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/14/2022] [Accepted: 09/23/2022] [Indexed: 11/06/2022] Open
Abstract
Implantable glucose biosensors provide real-time information about blood glucose fluctuations, but their utility and accuracy are time-limited due to the foreign body response (FBR) following their insertion beneath the skin. The slow release of nitric oxide (NO), a gasotransmitter with inflammation regulatory properties, from a sensor surface has been shown to dramatically improve sensors' analytical biocompatibility by reducing the overall FBR response. Indeed, work in a porcine model suggests that as long as the implants (sensors) continue to release NO, even at low levels, the inflammatory cell infiltration and resulting collagen density are lessened. While these studies strongly support the benefits of NO release in mitigating the FBR, the mechanisms through which exogenous NO acts on the surrounding tissue, especially under the condition of hyperglycemia, remain vague. Such knowledge would inform strategies to refine appropriate NO dosage and release kinetics for optimal therapeutic activity. In this study, we evaluated mediator, immune cell, and mRNA expression profiles in the local tissue microenvironment surrounding implanted sensors as a function of NO release, diabetes, and implantation duration. A custom porcine wound healing-centric multiplex gene array was developed for nanoString barcoding analysis. Tissues adjacent to sensors with sustained NO release abrogated the implant-induced acute and chronic FBR through modulation of the tissue-specific immune chemokine and cytokine microenvironment, resulting in decreased cellular recruitment, proliferation, and activation at both the acute (7-d) and chronic (14-d) phases of the FBR. Further, we found that sustained NO release abrogated the implant-induced acute and chronic foreign body response through modulation of mRNA encoding for key immunological signaling molecules and pathways, including STAT1 and multiple STAT1 targets including MAPK14, IRAK4, MMP2, and CXCL10. The condition of diabetes promoted a more robust FBR to the implants, which was also controlled by sustained NO release.
Collapse
Affiliation(s)
- James B Taylor
- Department of Chemistry, University of North Carolina at Chapel Hill, CB3290, Chapel Hill, NC 27599, USA
| | - Maggie J Malone-Povolny
- Department of Chemistry, University of North Carolina at Chapel Hill, CB3290, Chapel Hill, NC 27599, USA
| | - Elizabeth P Merricks
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, CB3290, Chapel Hill, NC 27599, USA
| | - Lauren E Wimsey
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, CB3290, Chapel Hill, NC 27599, USA
| | - Daniel Soliman
- Department of Surgery, University of North Carolina at Chapel Hill, CB3290, Chapel Hill, NC 27599, USA
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, CB3290, Chapel Hill, NC 27599, USA
| | - Timothy C Nichols
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, CB3290, Chapel Hill, NC 27599, USA
| | - Shannon M Wallet
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, CB3290, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, CB3290, Chapel Hill, NC 27599, USA
| | - Robert Maile
- Department of Surgery, University of North Carolina at Chapel Hill, CB3290, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, CB3290, Chapel Hill, NC 27599, USA
- Curriculum of Toxicology, University of North Carolina at Chapel Hill, CB3290, Chapel Hill, NC 27599, USA
| | - Mark H Schoenfisch
- Department of Chemistry, University of North Carolina at Chapel Hill, CB3290, Chapel Hill, NC 27599, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, CB3290, Chapel Hill, NC 27599, USA
| |
Collapse
|
29
|
Saadatmand F, Abbas M, Apprey V, Tailor K, Kwabi-Addo B. Sex differences in saliva-based DNA methylation changes and environmental stressor in young African American adults. PLoS One 2022; 17:e0273717. [PMID: 36067197 PMCID: PMC9447871 DOI: 10.1371/journal.pone.0273717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/02/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Low socioeconomic status neighborhood exposure to stress and violence may be sources of negative stimuli that poses significant health risks for children, adolescents and throughout the life course of an individual. The study aims to investigate if aberrant epigenetic DNA methylation changes may be a potential mechanism for regulating neighborhood exposures and health outcomes. METHODS Exposure to environmental stressors identified in 98 young African American (AA) adults aged 18-25 years old from the Washington D.C., area were used in the study. We correlated the association between stress markers; cortisol, CRP, IgG, IGA, IgM, and self-reported exposure to violence and stress, with quantitative DNA methylation changes in a panel of gene-specific loci using saliva DNA. RESULTS In all participants studied, the exposure to violence was significant and negatively correlated with DNA methylation of MST1R loci (p = 0.032; r = -0.971) and nominally significant with NR3C1 loci (p = 0.053; r = -0.948). In addition, we observed significant and negative correlation of DNA methylation changes of LINE1 (p = 0.044; r = -0.248); NR3C1 (p = 0.017; r = -0.186); MSTR1 (p = 0.022; r = -0.192); and DRD2 (p = 0.056; r = -0.184; albeit nominal significant correlation) with IgA expression. On the other hand, we observed a significant and position correlation of DNA methylation changes in DRD2 (p = 0.037; r = 0.184) with IgG expression. When participants were stratified by sex, we observed in AA young male adults, significant DNA methylation changes of MST1R (p< 0.05) and association with exposure to violence and IgG level. We also observed significant DNA methylation levels of DRD2 (p< 0.05) and association with IgA, IgG, and cortisol level. Furthermore, we observed significant DNA methylation changes of NR3C1 (p< 0.05) with stress, IgA, and IgG in the male participants only. On the other hand, we only observed significant and a positive association of IgG with DNA methylation levels of ESR1 (p = 0.041) in the young AA female participants. CONCLUSION Our preliminary observation of significant DNA methylation changes in neuronal and immune genes in saliva samples supports our recently published genome-wide DNA methylations changes in blood samples from young AA male adults indicating that saliva offers a non-invasive means for DNA methylation prediction of exposure to environmental stressors in a gender-specific manner.
Collapse
Affiliation(s)
- Forough Saadatmand
- Department of Pediatrics, Howard University, Washington, D.C., United States of America
| | - Muneer Abbas
- Department of Microbiology & The National Human Genome Center, Howard University, Washington, D.C., United States of America
| | - Victor Apprey
- Department of Biochemistry and Molecular Biology, College of Medicine, Howard University, Washington, D.C., United States of America
| | - Krishma Tailor
- Department of Biochemistry and Molecular Biology, College of Medicine, Howard University, Washington, D.C., United States of America
| | - Bernard Kwabi-Addo
- Department of Biochemistry and Molecular Biology, College of Medicine, Howard University, Washington, D.C., United States of America
- * E-mail:
| |
Collapse
|
30
|
Abstract
Deregulation of cell cycle is a typical feature of cancer cells. Normal cells rely on the strictly coordinated spindle assembly checkpoint (SAC) to maintain the genome integrity and survive. However, cancer cells could bypass this checkpoint mechanism. In this study, we showed the clinical relevance of threonine tyrosine kinase (TTK) protein kinase, a central regulator of the SAC, in hepatocellular carcinoma (HCC) and its potential as therapeutic target. Here, we reported that a newly developed, orally active small molecule inhibitor targeting TTK (CFI-402257) effectively suppressed HCC growth and induced highly aneuploid HCC cells, DNA damage, and micronuclei formation. We identified that CFI-402257 also induced cytosolic DNA, senescence-like response, and activated DDX41-STING cytosolic DNA sensing pathway to produce senescence-associated secretory phenotypes (SASPs) in HCC cells. These SASPs subsequently led to recruitment of different subsets of immune cells (natural killer cells, CD4+ T cells, and CD8+ T cells) for tumor clearance. Our mass cytometry data illustrated the dynamic changes in the tumor-infiltrating immune populations after treatment with CFI-402257. Further, CFI-402257 improved survival in HCC-bearing mice treated with anti-PD-1, suggesting the possibility of combination treatment with immune checkpoint inhibitors in HCC patients. In summary, our study characterized CFI-402257 as a potential therapeutic for HCC, both used as a single agent and in combination therapy.
Collapse
|
31
|
Meza Cerda MI, Gray R, Thomson PC, Butcher L, Simpson K, Cameron A, Marcus AD, Higgins DP. Developing Immune Profiles of Endangered Australian Sea Lion ( Neophoca cinerea) Pups Within the Context of Endemic Hookworm ( Uncinaria sanguinis) Infection. Front Vet Sci 2022; 9:824584. [PMID: 35529837 PMCID: PMC9069138 DOI: 10.3389/fvets.2022.824584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
As a top predator, the endangered Australian sea lion (Neophoca cinerea) is a sentinel of ecosystem change, where population trends can reflect broader shifts in the marine environment. The population of this endemic pinniped was historically diminished by commercial sealing, and recovery has been slowed by fishery interactions, disease and, potentially, pollutants. Hookworm infects 100% of neonatal pups and has been identified as a contributor to population decline. Here, a multivariable approach using traditional serological and novel molecular tools such as qPCR and ddPCR was used to examine immune phenotypes of developing Australian sea lion pups infected with the endemic hookworm (Uncinaria sanguinis) from two South Australian colonies. Results show changing immunophenotypes throughout the patent period of infection represented by pro-inflammatory cytokines (IL-6), IgG and acute-phase proteins. Although cytokines may prove useful as markers of resistance, in this study, IL-6 is determined to be an early biomarker of inflammation in Australian sea lion pups, excluding the alternative hypothesis. Additionally, immunological differences between animals from high- and low-intensity hookworm seasons, as well as ivermectin-treated animals, indicate hookworm infection modulation of the host immune response, as evidenced by a lower IL-6 mRNA expression in the non-treated groups. This study of the Australian sea lion is an example of an ecoimmunological approach to disease investigation, which can be applied to evaluate the impact of environmental and anthropogenic factors on susceptibility to infectious diseases in free-ranging species
Collapse
Affiliation(s)
- María-Ignacia Meza Cerda
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| | - Rachael Gray
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| | - Peter C Thomson
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| | - Loreena Butcher
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| | - Kelly Simpson
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| | - Abby Cameron
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| | - Alan D Marcus
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| | - Damien P Higgins
- Faculty of Science, Sydney School of Veterinary Science, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
32
|
Matsuda S, Kotani T, Kuwabara H, Suzuka T, Kiboshi T, Wada Y, Ishida T, Fujiki Y, Shiba H, Hata K, Shoda T, Hirose Y, Takeuchi T. Association of M2 macrophages, Th2, and B cells with pathomechanism in microscopic polyangiitis complicated by interstitial lung disease. J Rheumatol 2022; 49:913-921. [DOI: 10.3899/jrheum.220123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2022] [Indexed: 11/22/2022]
Abstract
Objective To address the pathomechanism of microscopic polyangiitis (MPA) complicated by interstitial lung disease (ILD) using serum biomarker profile and pulmonary histopathology. Methods Serum biomarkers from patients with MPA-ILD (n = 32), MPA without ILD (n = 17), and healthy controls (n =10) were examined. Based on the biomarker profiles, principal component analysis (PCA) and cluster analysis were performed to classify patients with MPA-ILD into subgroups. Clinical characteristics and prognosis were assessed for each subgroup. Two lung biopsies were examined following hematoxylineosin staining and immunostaining. Results T-cell and macrophage polarization was skewed toward the T helper (Th) 2 cells and M2 macrophages in MPA-ILD group relative to that in MPA without ILD group. The PCA allowed classification of the 19 biomarker profiles into three groups: (1) B cell- and neutrophil-related cytokines, vascular angiogenesis-related factors, extracellular matrix-producing factors, (2) Th1-driven cytokines, M1 macrophagedriven cytokines and Th2-driven cytokines, and (3) M2 macrophage -induced and - driven cytokines. The cluster analysis stratified the patients with MPA-ILD into clinically fibrotic dominant (CFD) and clinically inflammatory dominant (CID) groups. Notably, severe infections were significantly higher in the CFD group than in the CID group. Immunohistochemical staining demonstrated intense CXCL13 staining in B cells and Th2 cells in the interstitium of MPA-ILD lungs. Conclusion Activation of M2 macrophages, Th2 cells, and B cells plays a key role in the pathomechanism of MPA-ILD. Classification of MPA-ILD based on serum biomarker profile would be useful in predicting the disease activity and the complication of severe infection in MPA-ILD.
Collapse
|
33
|
Al-Hakeim HK, Al-Jassas HK, Morris G, Maes M. Increased ACE2, sRAGE, and Immune Activation, but Lowered Calcium and Magnesium in COVID-19. RECENT ADVANCES IN INFLAMMATION & ALLERGY DRUG DISCOVERY 2022; 16:32-43. [PMID: 35307003 DOI: 10.2174/2772270816666220318103929] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND The characterization of new biomarkers that could help externally validate the diagnosis of COVID-19 and optimize treatments is extremely important. Many studies have established changes in immune-inflammatory and antibody levels, but few studies measured the soluble receptor for the advanced glycation end product (sRAGE), angiotensin-converting enzyme 2 (ACE2), calcium, and magnesium in COVID-19. OBJECTIVE To evaluate serum advanced glycation end-product receptor (sRAGE) and angiotensin converting enzyme (ACE)2 and peripheral oxygen saturation (SpO2) and chest CT scan abnormalities (CCTA) in COVID-19. METHODS sRAGE, ACE2, interleukin (IL)-6, IL-10, C-reactive protein (CRP), calcium, magnesium, and albumin were measured in 60 COVID-19 patients and 30 healthy controls. RESULTS COVID-19 is characterized by significantly increased IL-6, CRP, IL-10, sRAGE, ACE2, and lowered SpO2, albumin, magnesium, and calcium. COVID-19 with CCTAs showed lower SpO2 and albumin. SpO2 was significantly inversely correlated with IL-6, IL-10, CRP, sRAGE, and ACE2, and positively with albumin, magnesium, and calcium. Neural networks showed that a combination of calcium, IL-6, CRP, and sRAGE yielded an accuracy of 100% in detecting COVID-19 patients, with calcium being the most important predictor followed by IL-6 and CRP. Patients with positive IgG results showed a significant elevation in the serum level of IL-6, sRAGE, and ACE2 compared to the negatively IgG patient subgroup. CONCLUSION The results show that immune-inflammatory and RAGE pathways biomarkers may be used as an external validating criterion for the diagnosis of COVID-19. Those pathways coupled with lowered SpO2, calcium, and magnesium are drug targets that may help reduce the consequences of COVID-19.
Collapse
Affiliation(s)
| | | | - Gerwyn Morris
- School of Medicine, IMPACT-the Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Australia
| | - Michael Maes
- School of Medicine, IMPACT-the Institute for Mental and Physical Health and Clinical Translation, Deakin University, Barwon Health, Geelong, Australia.,Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.,Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
34
|
Kozina E, Byrne M, Smeyne RJ. Mutant LRRK2 in lymphocytes regulates neurodegeneration via IL-6 in an inflammatory model of Parkinson's disease. NPJ Parkinsons Dis 2022; 8:24. [PMID: 35292674 PMCID: PMC8924242 DOI: 10.1038/s41531-022-00289-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 02/01/2022] [Indexed: 12/23/2022] Open
Abstract
Mutations in a number of genes contribute to development of Parkinson’s disease (PD), including several within the LRRK2 gene. However, little is known about the signals that underlie LRRK2-mediated neuronal loss. One clue resides in the finding that the neurodegenerative cascades emanate from signals arising from the peripheral immune system. Here, using two chimeric mouse models, we demonstrate that: 1) the replacement of mutant LRRK2 with wt form of the protein in T- and B-lymphocytes diminishes LPS-mediated inflammation and rescues the SNpc DA neuron loss in the mutant LRRK2 brain; 2) the presence of G2019S or R1441G LRRK2 mutation in lymphocytes alone is sufficient for LPS-induced DA neuron loss in the genotypically wt brain; and 3) neutralization of peripheral IL-6 overproduction prevents the SNpc DA neuron loss in LPS-treated mutant LRRK2 mice. These results represent a major paradigm shift in our understanding of PD pathogenesis and suggest that immune dysfunction in some forms of familial PD may have primacy over the CNS as the initiating site of the disorder.
Collapse
Affiliation(s)
- Elena Kozina
- Department of Neurosciences, Jefferson Hospital for Neuroscience, Thomas Jefferson University, 900 Walnut St, Philadelphia, PA, 19107, USA
| | - Matthew Byrne
- Department of Neurosciences, Jefferson Hospital for Neuroscience, Thomas Jefferson University, 900 Walnut St, Philadelphia, PA, 19107, USA
| | - Richard Jay Smeyne
- Department of Neurosciences, Jefferson Hospital for Neuroscience, Thomas Jefferson University, 900 Walnut St, Philadelphia, PA, 19107, USA.
| |
Collapse
|
35
|
Moorlag SJCFM, Taks E, ten Doesschate T, van der Vaart TW, Janssen AB, Müller L, Ostermann P, Dijkstra H, Lemmers H, Simonetti E, Mazur M, Schaal H, ter Heine R, van de Veerdonk FL, Bleeker-Rovers CP, van Crevel R, ten Oever J, de Jonge MI, Bonten MJ, van Werkhoven CH, Netea MG. Efficacy of BCG Vaccination Against Respiratory Tract Infections in Older Adults During the Coronavirus Disease 2019 Pandemic. Clin Infect Dis 2022; 75:e938-e946. [PMID: 35247264 PMCID: PMC8903481 DOI: 10.1093/cid/ciac182] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Older age is associated with increased severity and death from respiratory infections, including coronavirus disease 2019 (COVID-19). The tuberculosis BCG vaccine may provide heterologous protection against nontuberculous infections and has been proposed as a potential preventive strategy against COVID-19. METHODS In this multicenter, placebo-controlled trial, we randomly assigned older adults (aged ≥60 years; n = 2014) to intracutaneous vaccination with BCG vaccine (n = 1008) or placebo (n = 1006). The primary end point was the cumulative incidence of respiratory tract infections (RTIs) that required medical intervention, during 12 months of follow-up. Secondary end points included the incidence of COVID-19, and the effect of BCG vaccination on the cellular and humoral immune responses. RESULTS The cumulative incidence of RTIs requiring medical intervention was 0.029 in the BCG-vaccinated group and 0.024 in the control group (subdistribution hazard ratio, 1.26 [98.2% confidence interval, .65-2.44]). In the BCG vaccine and placebo groups, 51 and 48 individuals, respectively tested positive for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with polymerase chain reaction (subdistribution hazard ratio, 1.053 [95% confidence interval, .71-1.56]). No difference was observed in the frequency of adverse events. BCG vaccination was associated with enhanced cytokine responses after influenza, and also partially associated after SARS-CoV-2 stimulation. In patients diagnosed with COVID-19, antibody responses after infection were significantly stronger if the volunteers had previously received BCG vaccine. CONCLUSIONS BCG vaccination had no effect on the incidence of RTIs, including SARS-CoV-2 infection, in older adult volunteers. However, it improved cytokine responses stimulated by influenza and SARS-CoV-2 and induced stronger antibody titers after COVID-19 infection. CLINICAL TRIALS REGISTRATION EU Clinical Trials Register 2020-001591-15 ClinicalTrials.gov NCT04417335.
Collapse
Affiliation(s)
| | | | | | | | - Axel B Janssen
- University Medical Center Utrecht, Utrecht, the Netherlands
| | - Lisa Müller
- Department for Pediatric Oncology, Hematology and Clinical Immunology, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine University Duesseldorf, Germany
| | - Philipp Ostermann
- Department for Pediatric Oncology, Hematology and Clinical Immunology, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine University Duesseldorf, Germany
| | - Helga Dijkstra
- Department of Internal Medicine Radboud University Medical Center, Nijmegen, the Netherlands,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Heidi Lemmers
- Department of Internal Medicine Radboud University Medical Center, Nijmegen, the Netherlands,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Elles Simonetti
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands,Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marc Mazur
- University Medical Center Utrecht, Utrecht, the Netherlands
| | - Heiner Schaal
- Department for Pediatric Oncology, Hematology and Clinical Immunology, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine University Duesseldorf, Germany
| | - Rob ter Heine
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlandsand
| | - Frank L van de Veerdonk
- Department of Internal Medicine Radboud University Medical Center, Nijmegen, the Netherlands,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Chantal P Bleeker-Rovers
- Department of Internal Medicine Radboud University Medical Center, Nijmegen, the Netherlands,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Reinout van Crevel
- Department of Internal Medicine Radboud University Medical Center, Nijmegen, the Netherlands,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jaap ten Oever
- Department of Internal Medicine Radboud University Medical Center, Nijmegen, the Netherlands,Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marien I de Jonge
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands,Department of Laboratory Medicine, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marc J Bonten
- University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Mihai G Netea
- Correspondence: Mihai G. Netea, Department of Medicine (463), Radboud University Nijmegen Medical Center, Geert Grooteplein Zuid 8, 6525 GA Nijmegen, the Netherlands ()
| |
Collapse
|
36
|
Matsumura Y, Watanabe R, Niijima Y, Kawakita H, Furuta J, Nakamura Y, Ishitsuka Y, Okiyama N, Fujisawa Y, Fujimoto M. Possible activation of effector B cells during drug‐induced hypersensitivity syndrome. JOURNAL OF CUTANEOUS IMMUNOLOGY AND ALLERGY 2021. [DOI: 10.1002/cia2.12191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Yutaka Matsumura
- Department of Dermatology University of Tsukuba Ibaraki Japan
- Department of Dermatology Course of Integrated Medicine Graduate School of Medicine Osaka University Osaka Japan
| | - Rei Watanabe
- Department of Dermatology University of Tsukuba Ibaraki Japan
- Department of Dermatology Course of Integrated Medicine Graduate School of Medicine Osaka University Osaka Japan
| | - Yasuko Niijima
- Department of Dermatology University of Tsukuba Ibaraki Japan
| | - Haruka Kawakita
- Department of Dermatology University of Tsukuba Ibaraki Japan
| | - Junichi Furuta
- Department of Dermatology University of Tsukuba Ibaraki Japan
| | | | - Yosuke Ishitsuka
- Department of Dermatology University of Tsukuba Ibaraki Japan
- Department of Dermatology Course of Integrated Medicine Graduate School of Medicine Osaka University Osaka Japan
| | - Naoko Okiyama
- Department of Dermatology University of Tsukuba Ibaraki Japan
| | | | - Manabu Fujimoto
- Department of Dermatology University of Tsukuba Ibaraki Japan
- Department of Dermatology Course of Integrated Medicine Graduate School of Medicine Osaka University Osaka Japan
| |
Collapse
|
37
|
Kumazoe M, Takamatsu K, Horie F, Yoshitomi R, Hamagami H, Tanaka H, Fujimura Y, Tachibana H. Methylated (-)-epigallocatechin 3-O-gallate potentiates the effect of split vaccine accompanied with upregulation of Toll-like receptor 5. Sci Rep 2021; 11:23101. [PMID: 34845235 PMCID: PMC8630126 DOI: 10.1038/s41598-021-02346-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 11/08/2021] [Indexed: 11/09/2022] Open
Abstract
Split-virus vaccine serves as a major countermeasure against influenza virus, but its effectiveness and protective action are not complete. We previously demonstrated the effect of Benifuuki, a green tea cultivar in Japan, on enhancing the split-virus vaccine-elicited immune response. However, little is known about the detail mechanisms. Here, we show that EGCG3"Me intake significantly potentiated the vaccine-elicited hemagglutination inhibition titer increase. Flow cytometry analysis revealed the increased Toll-like receptor 5 (TLR5) expression after EGCG3"Me treatment in lamina propria dendritic cells (LPDCs) and macrophages, which play crucial roles in the humoral immune system. TLR5 expression correlated with the level of interleukin-6 (IL-6)/C-C chemokine type receptor 5, which are important mediators of the humoral immunity. Taken together, In vivo and ex vivo studies showed that EGCG3"Me potentiated the split-virus vaccine-elicited immune response accompanied with the upregulation of TLR5 in intestine and splenocyte macrophages.
Collapse
Affiliation(s)
- Motofumi Kumazoe
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Kanako Takamatsu
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Fuyumi Horie
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Ren Yoshitomi
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Hiroki Hamagami
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro, Tokyo, 152-8552, Japan
| | - Hiroshi Tanaka
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro, Tokyo, 152-8552, Japan
| | - Yoshinori Fujimura
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Hirofumi Tachibana
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
| |
Collapse
|
38
|
Haile AF, Woodfint RM, Kim E, Joldrichsen MR, Berhe N, Gebreyes WA, Boyaka PN. Broad-Spectrum and Gram-Negative-Targeting Antibiotics Differentially Regulate Antibody Isotype Responses to Injected Vaccines. Vaccines (Basel) 2021; 9:vaccines9111240. [PMID: 34835171 PMCID: PMC8619726 DOI: 10.3390/vaccines9111240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/11/2021] [Accepted: 10/14/2021] [Indexed: 11/19/2022] Open
Abstract
Antibiotics are extensively used worldwide for the treatment of common infections by agents such as E. coli and Salmonella. They also represent the most common cause of alteration of the microbiota in people. We addressed whether broad-spectrum and Gram-negative-targeting antibiotics differentially regulate systemic and mucosal immune responses to vaccines. Antibiotics treatment enhances serum IgG1 responses in mice immunized systemically with a model polyvalent vaccine. This increase was not seen for other IgG subclasses and was dependent on the immunogenicity of vaccine antigens. The broad-spectrum antibiotic cocktail also enhanced serum IgA responses. Interestingly, both the broad spectrum and the antibiotic targeting Gram-negative bacteria enhanced the number of IgA antibody secreting cells in the intestinal lamina propria. This effect was unlikely to be due to an increase in cells expressing gut-homing receptors (i.e., CCR9 and α4β7) in peripheral tissues. On the other hand, the microbiome in mice treated with antibiotics was characterized by an overall reduction of the number of firmicutes. Furthermore, Bacteroidetes were increased by either treatment, and Proteobacteria were increased by the broad-spectrum antibiotics cocktail. Thus, immunoglobulin isotype and subclass responses are differentially regulated by oral antibiotics treatment and the gut microbiota shapes mucosal antibody responses after systemic immunization.
Collapse
Affiliation(s)
- Aklilu F. Haile
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (A.F.H.); (R.M.W.); (E.K.); (M.R.J.)
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa 1000, Ethiopia;
| | - Rachel M. Woodfint
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (A.F.H.); (R.M.W.); (E.K.); (M.R.J.)
| | - Eunsoo Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (A.F.H.); (R.M.W.); (E.K.); (M.R.J.)
| | - Marisa R. Joldrichsen
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (A.F.H.); (R.M.W.); (E.K.); (M.R.J.)
| | - Nega Berhe
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa 1000, Ethiopia;
| | - Wondwoossen A. Gebreyes
- Department of Preventive Medicine, The Ohio State University, Columbus, OH 43210, USA;
- Global One Health Initiative, The Ohio State University, Columbus, OH 43210, USA
- Infection Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Prosper N. Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA; (A.F.H.); (R.M.W.); (E.K.); (M.R.J.)
- Global One Health Initiative, The Ohio State University, Columbus, OH 43210, USA
- Infection Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
- Department Microbial Immunity and Infection, The Ohio State University, Columbus, OH 43210, USA
- Correspondence:
| |
Collapse
|
39
|
Lacorcia M, Bhattacharjee S, Laubhahn K, Alhamdan F, Ram M, Muschaweckh A, Potaczek DP, Kosinska A, Garn H, Protzer U, Renz H, Prazeres da Costa C. Fetomaternal immune cross talk modifies T-cell priming through sustained changes to DC function. J Allergy Clin Immunol 2021; 148:843-857.e6. [PMID: 33684437 DOI: 10.1016/j.jaci.2021.02.031] [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: 08/05/2020] [Revised: 02/18/2021] [Accepted: 02/26/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Prenatal exposure to infections can modify immune development. These environmental disturbances during early life potentially alter the incidence of inflammatory disorders as well as priming of immune responses. Infection with the helminth Schistosoma mansoni is widely studied for its ability to alter immune responsiveness and is associated with variations in coinfection, allergy, and vaccine efficacy in endemic populations. OBJECTIVE Exposure to maternal schistosomiasis during early life, even without transmission of infection, can result in priming effects on offspring immune responses to bystander antigenic challenges as related to allergic responsiveness and vaccination, with this article seeking to further clarify the effects and underlying immunologic imprinting. METHODS Here, we have combined a model of chronic maternal schistosomiasis infection with a thorough analysis of subsequent offspring immune responses to allergy and vaccination models, including viral challenge and steady-state changes to immune cell compartments. RESULTS We have demonstrated that maternal schistosomiasis alters CD4+ responses during allergic sensitization and challenge in a skewed IL-4/B-cell-dominant response to antigenic challenge associated with limited inflammatory response. Beyond that, we have uncovered previously unidentified alterations to CD8+ T-cell responses during immunization that are dependent on vaccine formulation and have functional impact on the efficacy of vaccination against viral infection in a murine hepatitis B virus model. CONCLUSION In addition to steady-state modifications to CD4+ T-cell polarization and B-cell priming, we have traced these modified CD8+ responses to an altered dendritic cell phenotype sustained into adulthood, providing evidence for complex priming effects imparted by infection via fetomaternal cross talk.
Collapse
Affiliation(s)
- Matthew Lacorcia
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Sonakshi Bhattacharjee
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Kristina Laubhahn
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany; Pediatric Allergology, Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University Munich, Munich, Germany; German Center for Lung Research, Ludwig Maximilian University Munich, Munich, Germany
| | - Fahd Alhamdan
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Marija Ram
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Andreas Muschaweckh
- Department of Neurology, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Daniel P Potaczek
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Anna Kosinska
- Institute for Virology Technical University of Munich, Munich, Germany
| | - Holger Garn
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Ulrike Protzer
- Institute for Virology Technical University of Munich, Munich, Germany
| | - Harald Renz
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Clarissa Prazeres da Costa
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany.
| |
Collapse
|
40
|
Abstract
Dendritic cells (DCs) are efficient antigen-presenting cells that serve as a link between the innate and adaptive immune systems. These cells are broadly involved in cellular and humoral immune responses by presenting antigens to initiate T cell reactions, cytokine and chemokine secretion, T cell differentiation and expansion, B cell activation and regulation, and the mediation of immune tolerance. The functions of DCs depend on their activation status, which is defined by the stages of maturation, phenotype differentiation, and migration ability, among other factors. IL-6 is a soluble mediator mainly produced by a variety of immune cells, including DCs, that exerts pleiotropic effects on immune and inflammatory responses through interaction with specific receptors expressed on the surface of target cells. Here, we review the role of IL-6, when generated in an inflammatory context or as derived from DCs, in modulating the biologic function and activation status of DCs and emphasize the importance of searching for novel strategies to target the IL-6/IL-6 signaling pathway as a means to diminish the inflammatory activity of DCs in immune response or to prime the immunogenic activity of DCs in immunosuppressive conditions.
Collapse
Affiliation(s)
- Yu-Dong Xu
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mi Cheng
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pan-Pan Shang
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-Qing Yang
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
41
|
Matsuda Y, Watanabe T, Li XK. Approaches for Controlling Antibody-Mediated Allograft Rejection Through Targeting B Cells. Front Immunol 2021; 12:682334. [PMID: 34276669 PMCID: PMC8282180 DOI: 10.3389/fimmu.2021.682334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/17/2021] [Indexed: 01/14/2023] Open
Abstract
Both acute and chronic antibody-mediated allograft rejection (AMR), which are directly mediated by B cells, remain difficult to treat. Long-lived plasma cells (LLPCs) in bone marrow (BM) play a crucial role in the production of the antibodies that induce AMR. However, LLPCs survive through a T cell-independent mechanism and resist conventional immunosuppressive therapy. Desensitization therapy is therefore performed, although it is accompanied by severe side effects and the pathological condition may be at an irreversible stage when these antibodies, which induce AMR development, are detected in the serum. In other words, AMR control requires the development of a diagnostic method that predicts its onset before LLPC differentiation and enables therapeutic intervention and the establishment of humoral immune monitoring methods providing more detailed information, including individual differences in the susceptibility to immunosuppressive agents and the pathological conditions. In this study, we reviewed recent studies related to the direct or indirect involvement of immunocompetent cells in the differentiation of naïve-B cells into LLPCs, the limitations of conventional methods, and the possible development of novel control methods in the context of AMR. This information will significantly contribute to the development of clinical applications for AMR and improve the prognosis of patients who undergo organ transplantation.
Collapse
Affiliation(s)
- Yoshiko Matsuda
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Takeshi Watanabe
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
42
|
Zhou Y, Wang X, Xin M, Zhuang H. Changes in bone mineral density, 25-hydroxyvitamin D 3 and inflammatory factors in patients with hyperthyroidism. Exp Ther Med 2021; 21:617. [PMID: 33936274 DOI: 10.3892/etm.2021.10049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 12/17/2019] [Indexed: 11/05/2022] Open
Abstract
The present study aimed to evaluate changes in bone mineral density, 25-hydroxyvitamin D3 [25-(OH)D3] and inflammatory factors in patients with hyperthyroidism, in order to determine the correlations with the pathogenesis of hyperthyroidism. A total of 55 patients with hyperthyroidism (observation group) and 53 healthy patients (control group) enrolled at Weifang People's Hospital from March 2017 to February 2018 were randomly enrolled. The thyroid function, bone mineral density, 25-(OH)D3 and inflammatory factors were measured and compared between the two groups. The measurement data are presented as mean ± standard deviation (SD), and Student t-test was performed for the comparison between two groups. Chi-square test was used for enumeration data regarding sex. Pearson correlation analysis was performed for two-variable analysis on L1, 25-(OH)D3, interleukin (IL)-2, IL-6 with FT3, respectively. In regards to the results, no difference in sex, age and body mass index (BMI) between the two groups were found but the thyroid function was markedly enhanced in the observation group compared to the control group. Bone mineral density index and 25-(OH)D3 in the observation group were significantly lower than those in the control group (P<0.05). There were significant differences in the inflammatory factors between the two groups (P<0.05). The L1, 25-(OH)D3 and IL-2 levels were significantly negatively correlated with thyroid function index and free triiodothyronine (FT3) while a statistically positive correlation was found between IL-6 and FT3 (P<0.05). In conclusion, abnormal levels of bone mineral density, 25-(OH)D3 and inflammatory factors are observed in patients with hyperthyroidism, and there are correlations between L1, 25-(OH)D3, IL-2, IL-6 and FT3 in the pathogenesis of hyperthyroidism, which provides new insight for the diagnosis of hyperthyroidism.
Collapse
Affiliation(s)
- Yali Zhou
- Department of Traumatic Orthopaedics, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
| | - Xixia Wang
- Department of Internal Medicine, Zhucheng BaiChiHe Hospital, Zhucheng, Shandong 262217, P.R. China
| | - Maoyuan Xin
- Department of Traumatic Orthopaedics, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
| | - Haiting Zhuang
- Department of Anesthesiology, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
| |
Collapse
|
43
|
Chia WN, Zhu F, Ong SWX, Young BE, Fong SW, Le Bert N, Tan CW, Tiu C, Zhang J, Tan SY, Pada S, Chan YH, Tham CYL, Kunasegaran K, Chen MIC, Low JGH, Leo YS, Renia L, Bertoletti A, Ng LFP, Lye DC, Wang LF. Dynamics of SARS-CoV-2 neutralising antibody responses and duration of immunity: a longitudinal study. LANCET MICROBE 2021; 2:e240-e249. [PMID: 33778792 PMCID: PMC7987301 DOI: 10.1016/s2666-5247(21)00025-2] [Citation(s) in RCA: 251] [Impact Index Per Article: 83.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Background Studies have found different waning rates of neutralising antibodies compared with binding antibodies against SARS-CoV-2. The impact of neutralising antibody waning rate at the individual patient level on the longevity of immunity remains unknown. We aimed to investigate the peak levels and dynamics of neutralising antibody waning and IgG avidity maturation over time, and correlate this with clinical parameters, cytokines, and T-cell responses. Methods We did a longitudinal study of patients who had recovered from COVID-19 up to day 180 post-symptom onset by monitoring changes in neutralising antibody levels using a previously validated surrogate virus neutralisation test. Changes in antibody avidities and other immune markers at different convalescent stages were determined and correlated with clinical features. Using a machine learning algorithm, temporal change in neutralising antibody levels was classified into five groups and used to predict the longevity of neutralising antibody-mediated immunity. Findings We approached 517 patients for participation in the study, of whom 288 consented for outpatient follow-up and collection of serial blood samples. 164 patients were followed up and had adequate blood samples collected for analysis, with a total of 546 serum samples collected, including 128 blood samples taken up to 180 days post-symptom onset. We identified five distinctive patterns of neutralising antibody dynamics as follows: negative, individuals who did not, at our intervals of sampling, develop neutralising antibodies at the 30% inhibition level (19 [12%] of 164 patients); rapid waning, individuals who had varying levels of neutralising antibodies from around 20 days after symptom onset, but seroreverted in less than 180 days (44 [27%] of 164 patients); slow waning, individuals who remained neutralising antibody-positive at 180 days post-symptom onset (46 [28%] of 164 patients); persistent, although with varying peak neutralising antibody levels, these individuals had minimal neutralising antibody decay (52 [32%] of 164 patients); and delayed response, a small group that showed an unexpected increase of neutralising antibodies during late convalescence (at 90 or 180 days after symptom onset; three [2%] of 164 patients). Persistence of neutralising antibodies was associated with disease severity and sustained level of pro-inflammatory cytokines, chemokines, and growth factors. By contrast, T-cell responses were similar among the different neutralising antibody dynamics groups. On the basis of the different decay dynamics, we established a prediction algorithm that revealed a wide range of neutralising antibody longevity, varying from around 40 days to many decades. Interpretation Neutralising antibody response dynamics in patients who have recovered from COVID-19 vary greatly, and prediction of immune longevity can only be accurately determined at the individual level. Our findings emphasise the importance of public health and social measures in the ongoing pandemic outbreak response, and might have implications for longevity of immunity after vaccination. Funding National Medical Research Council, Biomedical Research Council, and A*STAR, Singapore.
Collapse
Affiliation(s)
- Wan Ni Chia
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Feng Zhu
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Sean Wei Xiang Ong
- National Centre for Infectious Diseases, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore
| | - Barnaby Edward Young
- National Centre for Infectious Diseases, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Siew-Wai Fong
- ASTAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Nina Le Bert
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Chee Wah Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Charles Tiu
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Jinyan Zhang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Seow Yen Tan
- Department of Infectious Diseases, Changi General Hospital, Singapore
| | - Surinder Pada
- Division of Infectious Diseases, Ng Teng Fong General Hospital, Singapore
| | - Yi-Hao Chan
- ASTAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Christine Y L Tham
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Kamini Kunasegaran
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Mark I-C Chen
- National Centre for Infectious Diseases, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore
| | - Jenny G H Low
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore.,Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Yee-Sin Leo
- National Centre for Infectious Diseases, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| | - Laurent Renia
- ASTAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - Antonio Bertoletti
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Lisa F P Ng
- ASTAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore
| | - David Chien Lye
- National Centre for Infectious Diseases, Singapore.,Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore.,SingHealth Duke-NUS Global Health Institute, Singapore
| |
Collapse
|
44
|
Başaran S, Şimşek-Yavuz S, Meşe S, Çağatay A, Medetalibeyoğlu A, Öncül O, Özsüt H, Ağaçfidan A, Gül A, Eraksoy H. The effect of tocilizumab, anakinra and prednisolone on antibody response to SARS-CoV-2 in patients with COVID-19: A prospective cohort study with multivariate analysis of factors affecting the antibody response. Int J Infect Dis 2021; 105:756-762. [PMID: 33737128 PMCID: PMC7959682 DOI: 10.1016/j.ijid.2021.03.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/01/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
Objectives Disease severity, previous medications and immunosuppressive agents could affect the antibody response against SARS-CoV-2. This study aimed to analyze variables affecting the humoral response to SARS-CoV-2. Methods This prospective cohort study included adult patients who recovered from COVID-19 and were admitted to a COVID-19 follow-up unit. Eight patient groups were defined in accordance with the results of thoracic computed tomography (CT), SARS-CoV-2 PCR test, and tocilizumab or anakinra use during active disease. Anti-S IgG antibodies were determined by ELISA in serum samples. Anti-S positive and negative cases were compared. Results A total of 518 patients were included in the study. SARS-CoV-2 IgG antibodies were positive in 82.8% of patients. SARS-CoV-2 PCR positivity, extent of lung involvement on CT, and time to antibody testing were independently associated with antibody positivity. Tocilizumab, anakinra or prednisolone use was not a factor affecting the antibody response. The rate of antibody response and sample/CO values among antibody-positive patients showed a linear relationship with the extent of lung involvement on CT. Conclusions The use of tocilizumab, anakinra and prednisolone for COVID-19 did not affect the antibody response against SARS-CoV-2. The main driver of antibody response among patients with COVID-19 was the extent of pulmonary involvement on CT.
Collapse
Affiliation(s)
- Seniha Başaran
- Department of Infectious Diseases and Clinical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey.
| | - Serap Şimşek-Yavuz
- Department of Infectious Diseases and Clinical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Sevim Meşe
- Department of Medical Microbiology, Division of Virology and Fundamental Immunology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Atahan Çağatay
- Department of Infectious Diseases and Clinical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Alpay Medetalibeyoğlu
- Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Oral Öncül
- Department of Infectious Diseases and Clinical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Halit Özsüt
- Department of Infectious Diseases and Clinical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ali Ağaçfidan
- Department of Medical Microbiology, Division of Virology and Fundamental Immunology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ahmet Gül
- Department of Rheumatology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Haluk Eraksoy
- Department of Infectious Diseases and Clinical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
45
|
Keivani Rad N, Mohri M, Seifi HA, Haghparast A. Supplementation of overripe pulp extract and green peel extract or powder of banana fruit peel (musa. cavendish) to diets of neonatal dairy calves: Effects on haematological, immunological and performance characteristics. Vet Med Sci 2021; 7:876-887. [PMID: 33502117 PMCID: PMC8136936 DOI: 10.1002/vms3.429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 11/28/2020] [Accepted: 01/04/2021] [Indexed: 12/27/2022] Open
Abstract
The present study investigated the effects of overripe pulp and green peel extract and powder of banana fruit (Musa. cavendish) on haematological, biochemical, immunological, health, and performance of Holstein dairy calves. In all, 40 newborn calves were randomly divided into four groups of 10 animals. In the control group, animals received no banana meal. In group 1, calves were supplemented with 2 g (dry matter)/kg body weight/day of overripe banana pulp extract. The calves in group 2 were supplemented with 1 g (dry matter) of overripe banana pulp extract/kg body weight/day and 1 g (dry matter) of green banana peel extract/kg body weight/day. The animals in group 3 were supplemented with 2 g/kg body weight/day of green banana peel powder. The feeding period of calves on the tested supplements was 5 days. Blood samples and other evaluations were taken on day 0 (at birth, before supplementation) and on days 7, 15 and 30. Just a trend towards better average daily weight gain was seen in groups 2 and 3 than others (p = 0.073). Significant group and sampling time interactions were seen for the quantities of RBC (group 1 was lower than other groups at day 30), MCV (group 3 was lower than other groups at day 30) and MCH (group 1 was higher than other groups at day 30) (p < 0.05). A trend towards significance in values of IgG (group 1 was lower than other groups at days 15 and 30) and bilirubin (higher values at day 7 in groups 1 and 2 than control, higher amounts at days 15 and 30 in groups 3 and 2 than control, respectively) was also observed. In conclusion, banana supplementation in neonatal calves had beneficial effects on the values of RBC, MCV, MCH, bilirubin, IgG and average daily weight gain in dairy calves.
Collapse
Affiliation(s)
- Nafiseh Keivani Rad
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mehrdad Mohri
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.,Center of Excellence on Ruminant Abortion and Neonatal Mortality, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hesam A Seifi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.,Center of Excellence on Ruminant Abortion and Neonatal Mortality, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Alireza Haghparast
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
46
|
Jochems PGM, Keusters WR, America AHP, Rietveld PCS, Bastiaan-Net S, Ariëns RMC, Tomassen MMM, Lewis F, Li Y, Westphal KGC, Garssen J, Wichers HJ, van Bergenhenegouwen J, Masereeuw R. A combined microphysiological-computational omics approach in dietary protein evaluation. NPJ Sci Food 2020; 4:22. [PMID: 33335099 PMCID: PMC7746769 DOI: 10.1038/s41538-020-00082-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 11/12/2020] [Indexed: 11/09/2022] Open
Abstract
Food security is under increased pressure due to the ever-growing world population. To tackle this, alternative protein sources need to be evaluated for nutritional value, which requires information on digesta peptide composition in comparison to established protein sources and coupling to biological parameters. Here, a combined experimental and computational approach is presented, which compared seventeen protein sources with cow’s whey protein concentrate (WPC) as the benchmark. In vitro digestion of proteins was followed by proteomics analysis and statistical model-based clustering. Information on digesta peptide composition resulted in 3 cluster groups, primarily driven by the peptide overlap with the benchmark protein WPC. Functional protein data was then incorporated in the computational model after evaluating the effects of eighteen protein digests on intestinal barrier integrity, viability, brush border enzyme activity, and immune parameters using a bioengineered intestine as microphysiological gut system. This resulted in 6 cluster groups. Biological clustering was driven by viability, brush border enzyme activity, and significant differences in immune parameters. Finally, a combination of proteomic and biological efficacy data resulted in 5 clusters groups, driven by a combination of digesta peptide composition and biological effects. The key finding of our holistic approach is that protein source (animal, plant or alternative derived) is not a driving force behind the delivery of bioactive peptides and their biological efficacy.
Collapse
Affiliation(s)
- Paulus G M Jochems
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Willem R Keusters
- Julius Centre, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Antoine H P America
- Wageningen Plant Research, Wageningen University, Wageningen, The Netherlands
| | - Pascale C S Rietveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Shanna Bastiaan-Net
- Wageningen Food & Biobased Research, Wageningen University, Wageningen, The Netherlands
| | - Renata M C Ariëns
- Wageningen Food & Biobased Research, Wageningen University, Wageningen, The Netherlands
| | - Monic M M Tomassen
- Wageningen Food & Biobased Research, Wageningen University, Wageningen, The Netherlands
| | - Fraser Lewis
- Danone Nutricia Research, Utrecht, The Netherlands
| | - Yang Li
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Koen G C Westphal
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,Danone Nutricia Research, Utrecht, The Netherlands
| | - Harry J Wichers
- Wageningen Food & Biobased Research, Wageningen University, Wageningen, The Netherlands
| | - Jeroen van Bergenhenegouwen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,Danone Nutricia Research, Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
47
|
The Tat Protein of HIV-1 Prevents the Loss of HSV-Specific Memory Adaptive Responses and Favors the Control of Viral Reactivation. Vaccines (Basel) 2020; 8:vaccines8020274. [PMID: 32512757 PMCID: PMC7349931 DOI: 10.3390/vaccines8020274] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/27/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023] Open
Abstract
The development of therapeutic strategies to control the reactivation of the Herpes Simplex Virus (HSV) is an unaddressed priority. In this study, we evaluated whether Tat, a HIV-1 protein displaying adjuvant functions, could improve previously established HSV-specific memory responses and prevent viral reactivation. To this aim, mice were infected with non-lethal doses of HSV-1 and, 44 days later, injected or not with Tat. Mice were then monitored to check their health status and measure memory HSV-specific cellular and humoral responses. The appearance of symptoms associated with HSV-reactivation was observed at significantly higher frequencies in the control group than in the Tat-treated mice. In addition, the control animals experienced a time-dependent decrease in HSV-specific Immunoglobulin G (IgG), while the Tat-treated mice maintained antibody titers over time. IgG levels were directly correlated with the number of HSV-specific CD8+ T cells, suggesting an effect of Tat on both arms of the adaptive immunity. Consistent with the maintenance of HSV-specific immune memory, Tat-treated mice showed a better control of HSV-1 re-infection. Although further studies are necessary to assess whether similar effects are observed in other models, these results indicate that Tat exerts a therapeutic effect against latent HSV-1 infection and re-infection by favoring the maintenance of adaptive immunity.
Collapse
|
48
|
Qi X, Guo H, Sun C, Tian Y, Ding M, Yang Y, Jin H. Clinical significance of progranulin correlated with serum soluble Oxford 40 ligand in primary Sjögren's syndrome. Medicine (Baltimore) 2020; 99:e19967. [PMID: 32358369 PMCID: PMC7440338 DOI: 10.1097/md.0000000000019967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The present study aimed to investigate the association between the expressions of serum progranulin (PGRN) and serum soluble Oxford 40 ligand (sOX40L) and determine their clinical significances in primary Sjögren's syndrome (pSS).The present study included a total of 68 patients with pSS and 50 healthy controls. Demographic data and clinical basic information were collected. Enzyme-linked immunosorbent assay (ELISA) was performed to determine serum levels of PGRN, sOX40L and interleukins. Spearman's correlation coefficient and Mann-Whitney U test were used to determine the correlation between PGRN, and sOX40L and the association between PGRN and sOX40L and disease activity and disease severity.Serum interleukin (IL)-4, IL-6, IL-10, PGRN, and sOX40L levels were significantly higher in pSS patients as compared to the healthy controls. A positive correlation was observed between PGRN and sOX40L. Patients with elevated levels of PGRN or sOX40L exhibited higher disease activity compared to those with lower levels. Patients with III to IV stages of pSS or multiple system damage showed higher serum levels of PGRN and sOX40L.Elevated serum PGRN, and sOX40L levels were relevant with disease activity and severity in patients with pSS.
Collapse
|
49
|
Abstract
PURPOSE Tocilizumab, a monoclonal antibody directed against the IL-6 receptor, might block detrimental effects of IL-6 on transplantation. IL-6 plays a considerable role in cytokine storm after stem cell transplantation as well as graft versus host disease, and it has also been shown to be involved in solid organ allograft rejection; therefore, tocilizumab is expected to promote graft survival. Nonetheless, due to the small number of studies and disparate methods of drug administration and outcome evaluation, for which types of transplantation, at which stages, and to what extent tocilizumab could be applied remains to be defined. METHODS The Pubmed, SCOPUS and Google Scholar search engines were used to collect data. The keywords were determined by Pubmed MeSH. No time limitation was set and all types of articles were allowed. RESULTS: According to the potential of Tocilozumab in controlling both cellular and humoral immunity it could be considered as a promising agent in tolerance induction; however, blocking IL-6 signaling might result in augmented infection rate in recipients. CONCLUSION The need for providing effective and safe immunosuppressive agents to protect transplanted cells and organs against allo-reactivity urges the collection and discussion of all available findings about inhibition of determining immune components including cytokines; herein, we have summarized the clinical consequences of blocking IL-6 by tocilizumab in stem cell and solid organ transplantations.
Collapse
|
50
|
Rubin SJS, Bloom MS, Robinson WH. B cell checkpoints in autoimmune rheumatic diseases. Nat Rev Rheumatol 2020; 15:303-315. [PMID: 30967621 DOI: 10.1038/s41584-019-0211-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
B cells have important functions in the pathogenesis of autoimmune diseases, including autoimmune rheumatic diseases. In addition to producing autoantibodies, B cells contribute to autoimmunity by serving as professional antigen-presenting cells (APCs), producing cytokines, and through additional mechanisms. B cell activation and effector functions are regulated by immune checkpoints, including both activating and inhibitory checkpoint receptors that contribute to the regulation of B cell tolerance, activation, antigen presentation, T cell help, class switching, antibody production and cytokine production. The various activating checkpoint receptors include B cell activating receptors that engage with cognate receptors on T cells or other cells, as well as Toll-like receptors that can provide dual stimulation to B cells via co-engagement with the B cell receptor. Furthermore, various inhibitory checkpoint receptors, including B cell inhibitory receptors, have important functions in regulating B cell development, activation and effector functions. Therapeutically targeting B cell checkpoints represents a promising strategy for the treatment of a variety of autoimmune rheumatic diseases.
Collapse
Affiliation(s)
- Samuel J S Rubin
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA.,Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.,VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Michelle S Bloom
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA.,Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.,VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - William H Robinson
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA. .,Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA. .,VA Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|