1
|
White AM, Craig AJ, Richie DL, Corley C, Sadek SM, Barton HN, Gipson CD. Nicotine is an Immunosuppressant: Implications for Women's Health and Disease. J Neuroimmunol 2024; 397:578468. [PMID: 39461120 PMCID: PMC11653054 DOI: 10.1016/j.jneuroim.2024.578468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/04/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
A plethora of evidence supports that nicotine, the primary alkaloid in tobacco products that is generally accepted for maintaining use, is immunoregulatory and may function as an immunosuppressant. Women have unique experiences with use of nicotine-containing products and also undergo significant reproductive transitions throughout their lifespan which may be impacted by nicotine use. Within the extant literature, there is conflicting evidence that nicotine may confer beneficial health effects in specific disease states (e.g., in ulcerative colitis). Use prevalence of nicotine-containing products is exceptionally high in individuals presenting with some comorbid disease states that impact immune system health and can be a risk factor for the development of diseases which disproportionately impact women; however, the mechanisms underlying these relationships are largely unclear. Further, little is known regarding the impacts of nicotine's immunosuppressive effects on women's health during the menopausal transition, which is arguably an inflammatory event characterized by a pro-inflammatory peri-menopause period. Given that post-menopausal women are at a higher risk than men for the development of neurodegenerative diseases such as Alzheimer's disease and are also more vulnerable to negative health effects associated with diseases such as HIV-1 infection, it is important to understand how use of nicotine-containing products may impact the immune milieu in women. In this review, we define instances in which nicotine use confers immunosuppressive, anti-inflammatory, or pro-inflammatory effects in the context of comorbid disease states, and focus on how nicotine impacts neuroimmune signaling to maintain use. We posit that regardless of potential health benefits, nicotine use cessation should be a priority in the clinical care of women. The synthesis of this review demonstrates the importance of systematically defining the relationships between volitional nicotine use, immune system function, and comorbid disease states in women to better understand how nicotine impacts women's health and disease.
Collapse
Affiliation(s)
- Ashley M White
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Ashley J Craig
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Daryl L Richie
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Christa Corley
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Safiyah M Sadek
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Heather N Barton
- Beebe Health, Gastroenterology and Internal Medicine, Lewes, Delaware, USA
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
2
|
Anft M, Zgoura P, Skrzypczyk S, Dürr M, Viebahn R, Westhoff TH, Stervbo U, Babel N. Effects of switching from twice-daily tacrolimus to once-daily extended-release meltdose tacrolimus on cellular immune response. FRONTIERS IN TRANSPLANTATION 2024; 3:1405070. [PMID: 39386200 PMCID: PMC11461451 DOI: 10.3389/frtra.2024.1405070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024]
Abstract
Background LCP-Tacro [LCPT], a novel once-daily, extended-release formulation of tacrolimus, has a reduced Cmax with comparable AUC exposure, requiring a ∼30% dose reduction in contrast to immediate-release tacrolimus (IR-Tac). Once-daily LCPT in de novo kidney transplantation has a comparable efficacy and safety profile to that of IR-Tac with advantages in bioavailability and absorption. The present investigation intends to analyze the effects of conversion from IR-Tac to LCPT on phenotype and function of T-cells and B-cells. Methods 16 kidney transplant patients treated by triple standard immunosuppression with a stable graft function undergoing a switch from IR-Tac to LCPT were included in this observational prospective study. We measured the main immune cell types and performed an in-depth characterization of B cell, dendritic cells and T cells including regulatory T cells of the patients before, 4 and 8 weeks after IR-Tac to LCPT conversion using multi-parameter fl ow cytometry. Additionally, we analyzed T cells by assessing third-party antigens (Tetanus Diphtheria, TD)-reactive T cells, which could be analyzed by restimulation with tetanus vaccine. Results Overall, we found no significant alterations following LCPT conversion for the most immune cell populations with a few cell populations showing transient quantitative increase. Thus, 4 weeks after conversion, more regulatory T cells could be measured in the patients with a significant shift from memory to naïve Tregs. Furthermore, we found a transient B cell expansion 4 weeks after conversion from IR-Tac to LCPT. There were no changes in the percentage of other basic immune cell types and the antigen-reactive T cells were also not altered after changing the medication to LCP-tacrolimus. Conclusion Here, we demonstrate first insights into the immune system changes occurred under IR-Tac to LCPT conversion therapy in kidney transplant patients. While phenotypic and functional characteristics of the most immune cell populations did not change, we could observe an a transient expansion of regulatory T cells in peripheral blood following IR-Tac to LCTP conversion, which might additionally contribute to the overall immunosuppressive effect.
Collapse
Affiliation(s)
- Moritz Anft
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Panagiota Zgoura
- Clinic for Internal Medicine, St. Anna Hospital Herne, Herne, Germany
- Clinic for Surgery, Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Sarah Skrzypczyk
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Michael Dürr
- Clinic for Internal Medicine, St. Anna Hospital Herne, Herne, Germany
- Clinic for Surgery, Knappschaftskrankenhaus Bochum, Bochum, Germany
- Berlin Institute of Health, Berlin-BrandenburgCenter for Regenerative Therapies, and Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin Augustenburger Platz, Berlin, Germany
| | - Richard Viebahn
- Clinic for Surgery, Knappschaftskrankenhaus Bochum, Bochum, Germany
| | - Timm H. Westhoff
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Ulrik Stervbo
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Nina Babel
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
- Berlin Institute of Health, Berlin-BrandenburgCenter for Regenerative Therapies, and Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin Augustenburger Platz, Berlin, Germany
| |
Collapse
|
3
|
Sinha V, Singh A, Singh A, Saraswati SSK, Rana AK, Kalra K, Natarajan K. Potassium ion channel Kir2.1 negatively regulates protective responses to Mycobacterium bovis BCG. J Leukoc Biol 2024; 116:644-656. [PMID: 38489665 DOI: 10.1093/jleuko/qiae068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/22/2024] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
Tuberculosis caused by the pathogen Mycobacterium tuberculosis leads to increased mortality and morbidity worldwide. The prevalence of highly drug-resistant strains has reinforced the need for greater understanding of host-pathogen interactions at the cellular and molecular levels. Our previous work demonstrated critical roles of calcium ion channels in regulating protective responses to mycobacteria. In this report, we deciphered the roles of inwardly rectifying K+ ion channel Kir2.1 in epithelial cells. Data showed that infection of epithelial cells (and macrophages) increases the surface expression of Kir2.1. This increased expression of Kir2.1 results in higher intracellular mycobacterial survival, as either inhibiting or knocking down Kir2.1 results in mounting of a higher oxidative burst leading to a significant attenuation of mycobacterial survival. Further, inhibiting Kir2.1 also led to increased expression of T cell costimulatory molecules accompanied with increased activation of MAP kinases and transcription factors nuclear factor κB and phosphorylated CREB. Furthermore, inhibiting Kir2.1 induced increased autophagy and apoptosis that could also contribute to decreased bacterial survival. Interestingly, an increased association of heat shock protein 70 kDa with Kir2.1 was observed. These results showed that mycobacteria modulate the expression and function of Kir2.1 in epithelial cells to its advantage.
Collapse
Affiliation(s)
- Vishal Sinha
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Akshita Singh
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Aarti Singh
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Shakuntala Surender Kumar Saraswati
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Ankush Kumar Rana
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Kanika Kalra
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| | - Krishnamurthy Natarajan
- Infectious Disease Immunology Laboratory, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North Campus, University Enclave, Delhi 110007, India
| |
Collapse
|
4
|
Fisher JS, Adán‐Barrientos I, Kumar NR, Lancaster JN. The aged microenvironment impairs BCL6 and CD40L induction in CD4 + T follicular helper cell differentiation. Aging Cell 2024; 23:e14140. [PMID: 38481058 PMCID: PMC11296098 DOI: 10.1111/acel.14140] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 06/13/2024] Open
Abstract
Weakened germinal center responses by the aged immune system result in diminished immunity against pathogens and reduced efficacy of vaccines. Prolonged contacts between activated B cells and CD4+ T cells are crucial to germinal center formation and T follicular helper cell (Tfh) differentiation, but it is unclear how aging impacts the quality of this interaction. Peptide immunization confirmed that aged mice have decreased expansion of antigen-specific germinal center B cells and reduced antibody titers. Furthermore, aging was associated with accumulated Tfh cells, even in naïve mice. Despite increased numbers, aged Tfh had reduced expression of master transcription factor BCL6 and increased expression of the ectonucleotidase CD39. In vitro activation revealed that proliferative capacity was maintained in aged CD4+ T cells, but not the costimulatory molecule CD40L. When activated in vitro by aged antigen-presenting cells, young CD4+ naïve T cells generated reduced numbers of activated cells with upregulated CD40L. To determine the contribution of cell-extrinsic influences on antigen-specific Tfh induction, young, antigen-specific B and CD4+ T cells were adoptively transferred into aged hosts prior to peptide immunization. Transferred cells had reduced expansion and differentiation into germinal center B cell and Tfh and reduced antigen-specific antibody titers when compared to young hosts. Young CD4+ T cells transferred aged hosts differentiated into Tfh cells with reduced PD-1 and BCL6 expression, and increased CD39 expression, though they maintained their mitochondrial capacity. These results highlight the role of the lymphoid microenvironment in modulating CD4+ T cell differentiation, which contributes to impaired establishment and maintenance of germinal centers.
Collapse
Affiliation(s)
| | - Irene Adán‐Barrientos
- Immunobiology LaboratoryCentro Nacional de Investigaciones Cardiovasculares (CNIC)MadridSpain
| | - Naveen R. Kumar
- Department of ImmunologyMayo ClinicScottsdaleArizonaUSA
- School of Life SciencesArizona State UniversityTempeArizonaUSA
| | - Jessica N. Lancaster
- Department of ImmunologyMayo ClinicScottsdaleArizonaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Department of Cancer BiologyMayo ClinicScottsdaleArizonaUSA
| |
Collapse
|
5
|
Han W, Allam SA, Elsawa SF. GLI2-Mediated Inflammation in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1263:55-65. [PMID: 32588323 DOI: 10.1007/978-3-030-44518-8_5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The tumor microenvironment (TME) plays an important role in the development and progression of cancer and has been shown to contribute to resistance to therapy. Inflammation is one of the hallmarks of cancer implicated in disease phenotype. Therefore, understanding the mechanisms that regulate inflammation in cancer and consequently how inflammatory mediators promote cancer progression is important for our understanding of cancer cell biology. The transcription factor GLI2 was initially identified as a member of the Hedgehog (HH) signaling pathway. During the last decade, studies have shown a novel mechanism of GLI2 regulation independent of HH signaling, where GLI2 consequently modulated several cytokine genes in the TME. These studies highlight a novel role for GLI2 as an inflammatory mediatory independent of HH stimulation. This chapter will discuss canonical and noncanonical pathways of GLI2 regulation and some of the downstream cytokine target genes regulated by GLI2.
Collapse
Affiliation(s)
- Weiguo Han
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Shereen A Allam
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Sherine F Elsawa
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH, USA.
| |
Collapse
|
6
|
CCR2 signaling in breast carcinoma cells promotes tumor growth and invasion by promoting CCL2 and suppressing CD154 effects on the angiogenic and immune microenvironments. Oncogene 2019; 39:2275-2289. [PMID: 31827233 PMCID: PMC7071973 DOI: 10.1038/s41388-019-1141-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 11/22/2019] [Accepted: 11/29/2019] [Indexed: 12/31/2022]
Abstract
Breast cancer is the second leading cause of cancer related deaths for women, due mainly to metastatic disease. Invasive tumors exhibit aberrations in recruitment and activity of immune cells, including decreased cytotoxic T cells. Restoring the levels and activity of cytotoxic T cells is a promising anti-cancer strategy; but its success is tumor type-dependent. The mechanisms that coordinate recruitment and activity of immune cells and other stromal cells in breast cancer remain poorly understood. Using the MMTV-PyVmT/FVB mammary tumor model, we demonstrate a novel role for CCL2/CCR2 chemokine signaling in tumor progression by altering the microenvironment. Selective targeting of CCR2 in the PyVmT mammary epithelium inhibited tumor growth and invasion, elevated CD8+ T cells, decreased M2 macrophages and decreased angiogenesis. Co-culture models demonstrated these stromal cell responses were mediated by tumor derived CCL2 and CCR2-mediated suppression of the T cell activating cytokine, CD154. Co-culture analysis indicated that CCR2-induced stromal reactivity was important for tumor cell proliferation and invasion. In breast tumor tissues, CD154 expression inversely correlated with CCR2 expression and correlated with relapse free survival. Targeting the CCL2/CCR2 signaling pathway may reprogram the immune angiogenic and microenvironments and enhance effectiveness of targeted and immuno-therapies.
Collapse
|
7
|
Li Y, Wei Y, Yuan W, Huang Q, Zhao Y, Zhao W, Xu W, Wu J. DNA damage induced by human CD40 ligand mutant promotes senescence and induces demethylation of GATA4 in lung cancer. Oncol Rep 2018; 39:2071-2080. [PMID: 29565449 PMCID: PMC5928763 DOI: 10.3892/or.2018.6310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 03/08/2018] [Indexed: 12/21/2022] Open
Abstract
The ligand of CD40, known as CD154 or CD40L, is the key to immunostimulatory and anticancer activity, but how CD40L affects cellular senescence is unclear. Thus, we studied a membrane-stable mutant form CD40L (CD40L-M) to explore tumor growth and cellular senescence in CD40-positive NSCLC cells. We found that CD40L-M-expressing cells had senescent characteristics, including reduced cell proliferation and enlargement, increased SA-β-gal staining activity, and overexpression of several cell cycle regulators p53 and p21. In addition, expression of GATA4 was restored, and the NF-κB signaling pathway was activated in the CD40L-M-induced senescent cells. Mechanistic analyses revealed that CD40L-M expression triggered the ATM/Chk2 DNA damage response, which mediated cell senescence and GATA4 activation. Knockdown of GATA4 reversed CD40L-M-induced senescence and decreased NF-κB activity. Thus, CD40L-M contributes to induction of cell senescence in CD40-positive NSCLC cells, and GATA4 is a switch to activate the NF-κB pathway, which is positively regulated by DNA damage response (DDR) signaling kinases. Collectively, CD40L-M-induced senescence may be a barrier to the growth of lung cancer cells.
Collapse
Affiliation(s)
- Yue Li
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yunyan Wei
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Weiwei Yuan
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Qiqing Huang
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Yaya Zhao
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Weihong Zhao
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wei Xu
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jianqing Wu
- Jiangsu Provincial Key Laboratory of Geriatrics, Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
8
|
Drum CL, Tan WKY, Chan SP, Pakkiri LS, Chong JPC, Liew OW, Ng TP, Ling LH, Sim D, Leong KTG, Yeo DPS, Ong HY, Jaufeerally F, Wong RCC, Chai P, Low AF, Davidsson P, Liljeblad M, Söderling AS, Gan LM, Bhat RV, Purnamawati K, Lam CSP, Richards AM. Thymosin Beta-4 Is Elevated in Women With Heart Failure With Preserved Ejection Fraction. J Am Heart Assoc 2017; 6:JAHA.117.005586. [PMID: 28611096 PMCID: PMC5669175 DOI: 10.1161/jaha.117.005586] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Thymosin beta-4 (TB4) is an X-linked gene product with cardioprotective properties. Little is known about plasma concentration of TB4 in heart failure (HF), and its relationship with other cardiovascular biomarkers. We sought to evaluate circulating TB4 in HF patients with preserved (HFpEF) or reduced (HFrEF) ejection fraction compared to non-HF controls. METHODS AND RESULTS TB4 was measured using a liquid chromatography and mass spectrometry assay in age- and sex-matched HFpEF (n=219), HFrEF (n=219) patients, and controls (n=219) from a prospective nationwide study. Additionally, a 92-marker multiplex proximity extension assay was measured to identify biomarker covariates. Compared with controls, plasma TB4 was elevated in HFpEF (985 [421-1723] ng/mL versus 1401 [720-2379] ng/mL, P<0.001), but not in HFrEF (1106 [556-1955] ng/mL, P=0.642). Stratifying by sex, only women (1623 [1040-2625] ng/mL versus 942 [386-1891] ng/mL, P<0.001), but not men (1238.5 [586-1967] ng/mL versus 1004 [451-1538] ng/mL, P=1.0), had significantly elevated TB4 in the setting of HFpEF. Adjusted for New York Heart Association class, N-terminal pro B-type natriuretic peptide, age, and myocardial infarction, hazard ratio to all-cause mortality is significantly higher in women with elevated TB4 (1.668, P=0.036), but not in men (0.791, P=0.456) with HF. TB4 is strongly correlated with a cluster of 7 markers from the proximity extension assay panel, which are either X-linked, regulated by sex hormones, or involved with NF-κB signaling. CONCLUSIONS We show that plasma TB4 is elevated in women with HFpEF and has prognostic information. Because TB4 can preserve EF in animal studies of cardiac injury, the relation of endogenous, circulating TB4 to X chromosome biology and differential outcomes in female heart disease warrants further study.
Collapse
Affiliation(s)
- Chester L Drum
- Cardiovascular Research Institute, National University Health System, Singapore .,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore
| | - Warren K Y Tan
- Cardiovascular Research Institute, National University Health System, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Siew-Pang Chan
- Cardiovascular Research Institute, National University Health System, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Department of Mathematics & Statistics, College of Science, Health & Engineering, La Trobe University, Melbourne, Australia
| | | | - Jenny P C Chong
- Cardiovascular Research Institute, National University Health System, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Oi-Wah Liew
- Cardiovascular Research Institute, National University Health System, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Tze-Pin Ng
- Cardiovascular Research Institute, National University Health System, Singapore.,Department of Psychological Medicine, National University of Singapore, Singapore
| | - Lieng-Hsi Ling
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,National University Heart Centre Singapore, Singapore
| | - David Sim
- National Heart Centre Singapore, Singapore.,Duke-NUS Medical School, Singapore
| | | | | | - Hean-Yee Ong
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Department of Cardiology, Khoo Teck Puat Hospital, Singapore
| | - Fazlur Jaufeerally
- Singapore General Hospital, Singapore.,Duke-NUS Medical School, Singapore
| | | | - Ping Chai
- National University Heart Centre Singapore, Singapore
| | - Adrian F Low
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,National University Heart Centre Singapore, Singapore
| | - Pia Davidsson
- Innovative Medicines & Early Development, Cardiovascular & Metabolic Diseases iMed, AstraZeneca R&D, Gothenburg, Sweden
| | - Mathias Liljeblad
- Innovative Medicines & Early Development, Cardiovascular & Metabolic Diseases iMed, AstraZeneca R&D, Gothenburg, Sweden
| | - Ann-Sofi Söderling
- Innovative Medicines & Early Development, Cardiovascular & Metabolic Diseases iMed, AstraZeneca R&D, Gothenburg, Sweden
| | - Li-Ming Gan
- Innovative Medicines & Early Development, Cardiovascular & Metabolic Diseases iMed, AstraZeneca R&D, Gothenburg, Sweden
| | - Ratan V Bhat
- Innovative Medicines & Early Development, Cardiovascular & Metabolic Diseases iMed, AstraZeneca R&D, Gothenburg, Sweden
| | - Kristy Purnamawati
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore
| | - Carolyn S P Lam
- National Heart Centre Singapore, Singapore.,Duke-NUS Medical School, Singapore
| | - A Mark Richards
- Cardiovascular Research Institute, National University Health System, Singapore .,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Christchurch Heart Institute, University of Otago, New Zealand
| |
Collapse
|
9
|
Han W, Jackson DA, Matissek SJ, Misurelli JA, Neil MS, Sklavanitis B, Amarsaikhan N, Elsawa SF. Novel Molecular Mechanism of Regulation of CD40 Ligand by the Transcription Factor GLI2. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:4481-4489. [PMID: 28461568 PMCID: PMC5473292 DOI: 10.4049/jimmunol.1601490] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 03/30/2017] [Indexed: 12/14/2022]
Abstract
The interaction between tumor cells and their surrounding microenvironment is essential for the growth and persistence of cancer cells. This interaction is mediated, in part, by cytokines. Although the role of cytokines in normal and malignant cell biology is well established, many of the molecular mechanisms regulating their expression remain elusive. In this article, we provide evidence of a novel pathway controlling the transcriptional activation of CD40L in bone marrow-derived stromal cells. Using a PCR-based screening of cytokines known to play a role in the biology of bone marrow malignancies, we identified CD40L as a novel GLI2 target gene in stromal cells. CD40L plays an important role in malignant B cell biology, and we found increased Erk phosphorylation and cell growth in malignant B cells cocultured with CD40L-expressing stromal cells. Further analysis indicated that GLI2 overexpression induced increased CD40L expression, and, conversely, GLI2 knockdown reduced CD40L expression. Using luciferase and chromatin immunoprecipitation assays, we demonstrate that GLI2 directly binds and regulates the activity of the CD40L promoter. We found that the CCR3-PI3K-AKT signaling modulates the GLI2-CD40L axis, and GLI2 is required for CCR3-PI3K-AKT-mediated regulation of the CD40L promoter. Finally, coculture of malignant B cells with cells stably expressing human CD40L results in increased Erk phosphorylation and increased malignant B cell growth, indicating that CD40L in the tumor microenvironment promotes malignant B cell activation. Therefore, our studies identify a novel molecular mechanism of regulation of CD40L by the transcription factor GLI2 in the tumor microenvironment downstream of CCR3 signaling.
Collapse
Affiliation(s)
- Weiguo Han
- Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115
| | - David A Jackson
- Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115
| | - Stephan J Matissek
- Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115
| | - Jason A Misurelli
- Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115
| | - Matthew S Neil
- Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115
| | - Brandon Sklavanitis
- Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115
| | | | - Sherine F Elsawa
- Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115
| |
Collapse
|
10
|
Liu WH, Kang SG, Huang Z, Wu CJ, Jin HY, Maine CJ, Liu Y, Shepherd J, Sabouri-Ghomi M, Gonzalez-Martin A, Xu S, Hoffmann A, Zheng Y, Lu LF, Xiao N, Fu G, Xiao C. A miR-155-Peli1-c-Rel pathway controls the generation and function of T follicular helper cells. J Exp Med 2016; 213:1901-19. [PMID: 27481129 PMCID: PMC4995083 DOI: 10.1084/jem.20160204] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 06/01/2016] [Indexed: 01/05/2023] Open
Abstract
MicroRNA (miRNA) deficiency impairs the generation of T follicular helper (Tfh) cells, but the contribution of individual miRNAs to this phenotype remains poorly understood. In this study, we performed deep sequencing analysis of miRNAs expressed in Tfh cells and identified a five-miRNA signature. Analyses of mutant mice deficient of these miRNAs revealed that miR-22 and miR-183/96/182 are dispensable, but miR-155 is essential for the generation and function of Tfh cells. miR-155 deficiency led to decreased proliferation specifically at the late stage of Tfh cell differentiation and reduced CD40 ligand (CD40L) expression on antigen-specific CD4(+) T cells. Mechanistically, miR-155 repressed the expression of Peli1, a ubiquitin ligase that promotes the degradation of the NF-κB family transcription factor c-Rel, which controls cellular proliferation and CD40L expression. Therefore, our study identifies a novel miR-155-Peli1-c-Rel pathway that specifically regulates Tfh cell generation and function.
Collapse
Affiliation(s)
- Wen-Hsien Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361005, China
| | - Seung Goo Kang
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037 Division of Biomedical Convergence/Institute of Bioscience and Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Zhe Huang
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Cheng-Jang Wu
- Division of Biological Sciences, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093
| | - Hyun Yong Jin
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Christian J Maine
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Yi Liu
- Department of Microbiology, Immunology, and Molecular Genetics, Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095
| | - Jovan Shepherd
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Mohsen Sabouri-Ghomi
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Alicia Gonzalez-Martin
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Shunbin Xu
- Department of Ophthalmology/Kresge Eye Institute, School of Medicine, Wayne State University, Detroit, MI 48202 Department of Anatomy and Cell Biology, School of Medicine, Wayne State University, Detroit, MI 48202
| | - Alexander Hoffmann
- Department of Microbiology, Immunology, and Molecular Genetics, Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095
| | - Ye Zheng
- Nomis Foundation Laboratories for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Li-Fan Lu
- Division of Biological Sciences, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093
| | - Nengming Xiao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361005, China
| | - Guo Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361005, China
| | - Changchun Xiao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen 361005, China Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
11
|
Abstract
Fanconi anemia (FA) is a genetic disorder associated with bone marrow (BM) failure and leukemia. Recent studies demonstrate variable immune defects in FA. However, the cause for FA immunodeficiency is unknown. Here we report that deletion of Fanca or Fancd2 dysregulates the suppressive activity of regulatory T cells (Tregs), shown functionally as exacerbation of graft-vs-host disease (GVHD) in mice. Recipient mice of Fanca(-/-) or Fancd2(-/-) BM chimeras exhibited severe acute GVHD after allogeneic BM transplantation (BMT). T cells from Fanca(-/-) or Fancd2(-/-) mice induced higher GVHD lethality than those from wild-type (WT) littermates. FA Tregs possessed lower proliferative suppression potential compared with WT Tregs, as demonstrated by in vitro proliferation assay and BMT. Analysis of CD25(+)Foxp3(+) Tregs indicated that loss of Fanca or Fancd2 dysregulated Foxp3 target gene expression. Additionally, CD25(+)Foxp3(+) Tregs of Fanca(-/-) or Fancd2(-/-) mice were less efficient in suppressing the production of GVHD-associated inflammatory cytokines. Consistently, aberrant NF-κB activity was observed in infiltrated T cells from FA GVHD mice. Conditional deletion of p65 in FA Tregs decreased GVHD mortality. Our study uncovers an essential role for FA proteins in maintaining Treg homeostasis, possibly explaining, at least in part, the immune deficiency reported in some FA patients.
Collapse
|
12
|
Impaired NFAT and NFκB activation are involved in suppression of CD40 ligand expression by Δ(9)-tetrahydrocannabinol in human CD4(+) T cells. Toxicol Appl Pharmacol 2013; 273:209-18. [PMID: 23999542 DOI: 10.1016/j.taap.2013.08.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 08/21/2013] [Accepted: 08/21/2013] [Indexed: 11/22/2022]
Abstract
We have previously reported that Δ(9)-tetrahydrocannabinol (Δ(9)-THC), the main psychoactive cannabinoid in marijuana, suppresses CD40 ligand (CD40L) expression by activated mouse CD4(+) T cells. CD40L is involved in pathogenesis of many autoimmune and inflammatory diseases. In the present study, we investigated the molecular mechanism of Δ(9)-THC-mediated suppression of CD40L expression using peripheral blood human T cells. Pretreatment with Δ(9)-THC attenuated CD40L expression in human CD4(+) T cells activated by anti-CD3/CD28 at both the protein and mRNA level, as determined by flow cytometry and quantitative real-time PCR, respectively. Electrophoretic mobility shift assays revealed that Δ(9)-THC suppressed the DNA-binding activity of both NFAT and NFκB to their respective response elements within the CD40L promoter. An assessment of the effect of Δ(9)-THC on proximal T cell-receptor (TCR) signaling induced by anti-CD3/CD28 showed significant impairment in the rise of intracellular calcium, but no significant effect on the phosphorylation of ZAP70, PLCγ1/2, Akt, and GSK3β. Collectively, these findings identify perturbation of the calcium-NFAT and NFκB signaling cascade as a key mechanistic event by which Δ(9)-THC suppresses human T cell function.
Collapse
|
13
|
MacLeod IJ, Rowley CF, Lockman S, Ogwu A, Moyo S, van Widenfelt E, Mmalane M, Makhema J, Essex M, Shapiro RL. Abacavir alters the transcription of inflammatory cytokines in virologically suppressed, HIV-infected women. J Int AIDS Soc 2012; 15:17393. [PMID: 22789611 PMCID: PMC3499794 DOI: 10.7448/ias.15.2.17393] [Citation(s) in RCA: 4] [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: 11/10/2011] [Revised: 03/09/2012] [Accepted: 04/18/2012] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Abacavir (ABC) may be associated with a small, increased risk of myocardial infarction in HIV-infected adults, possibly related to cytokine-mediated inflammation. METHODS To evaluate the induction of inflammatory cytokine transcription by ABC, we used samples from women randomized to receive zidovudine/lamivudine/ABC (Trizivir) or lopinavir/ritonavir and zidovudine/lamividine (Kaletra/Combivir) from the third trimester through six-months postpartum for the prevention of mother-to-child transmission (PMTCT). Women were matched by CD4 count and baseline HIV RNA. All women attained viral suppression (<50 copies/ml) by the time of sampling. RESULTS Four cytokines showed a difference in expression between the treatment arms, all in a proinflammatory direction for the ABC arm: CD40LG 1.82-fold, (p=.027); IL-8 3.16-fold (p=.020); LTA 2.82-fold, (p=.008); and CCL5 -1.67-fold, (p=.035). At 12-months postpartum, 6-months after antiretroviral discontinuation, cytokine expression was similar by treatment arm. CONCLUSIONS We conclude that ABC may upregulate proinflammatory cytokines at the transcriptional level in this population.
Collapse
Affiliation(s)
- Iain J MacLeod
- Harvard School of Public Health AIDS Initiative, Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
- Botswana–Harvard AIDS Institute, Gaborone, Botswana
| | - Christopher F Rowley
- Harvard School of Public Health AIDS Initiative, Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
- Botswana–Harvard AIDS Institute, Gaborone, Botswana
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Lowry Building Boston, MA, USA
| | - Shahin Lockman
- Harvard School of Public Health AIDS Initiative, Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
- Botswana–Harvard AIDS Institute, Gaborone, Botswana
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Anthony Ogwu
- Botswana–Harvard AIDS Institute, Gaborone, Botswana
| | | | | | | | | | - M Essex
- Harvard School of Public Health AIDS Initiative, Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
- Botswana–Harvard AIDS Institute, Gaborone, Botswana
| | - Roger L Shapiro
- Harvard School of Public Health AIDS Initiative, Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
- Botswana–Harvard AIDS Institute, Gaborone, Botswana
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Lowry Building Boston, MA, USA
| |
Collapse
|
14
|
Human CD14hi monocytes and myeloid dendritic cells provide a cell contact-dependent costimulatory signal for early CD40 ligand expression. Blood 2010; 117:1585-94. [PMID: 20634374 DOI: 10.1182/blood-2008-01-130252] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
CD40L on CD4(+) T cells plays a vital role in the activation of antigen-presenting cells, thus catalyzing a positive feedback loop for T-cell activation. Despite the pivotal juxtaposition of CD40L between antigen-presenting cells and T-cell activation, only a T-cell receptor stimulus is thought to be required for early CD40L surface expression. We show, for the first time, that CD40L expression on peripheral blood CD4(+) T cells is highly dependent on a cell-cell interaction with CD14(hi)CD16(-) monocytes. Interactions with ICAM-1, LFA-3, and to a lesser extent CD80/CD86 contribute to this enhancement of CD40L expression but are not themselves sufficient. The contact-mediated increase in CD40L expression is dependent on new mRNA and protein synthesis. Circulating myeloid dendritic cells also possess this costimulatory activity. By contrast, CD14(lo)CD16(+) monocytes, plasmacytoid dendritic cells, B-cell lymphoma lines, and resting, activated, and Epstein-Barr virus-immortalized primary B cells all lack the capacity to up-regulate early CD40L. The latter indicates that a human B cell cannot activate its cognate T cell to deliver CD40L-mediated help. This finding has functional implications for the role of biphasic CD40L expression, suggesting that the early phase is associated with antigen-presenting cell activation, whereas the late phase is related to B-cell activation.
Collapse
|
15
|
Abstract
Abstract
The generation of natural regulatory T cells (nTregs) is crucial for the establishment of immunologic self-tolerance and the prevention of autoimmunity. Still, the origin of nTregs and the mechanisms governing their differentiation within the thymus are poorly understood, particularly in humans. It was recently shown that conventional dendritic cells (cDCs) in human thymus were capable of inducing nTreg differentiation. However, the function of plasmacytoid DCs (pDCs), the other major subset of thymic DCs, remains unknown. Here we report that pDCs resident in the human thymus, when activated with CD40 ligand (CD40L) plus interleukin-3, efficiently promoted the generation of CD4+CD25+Foxp3+ nTregs from autologous thymocytes. The progenitors of these nTregs were selectively found within CD4+CD8+ thymocytes that had accomplished positive selection, as judged by their CD69hiTCRhi phenotype. Supporting the involvement of the CD40-CD40L pathway in pDC-induced nTreg generation, we show that positively selected CD4+CD8+ progenitors specifically transcribed CD40L in vivo and up-regulated CD40L expression on T-cell receptor engagement, thereby promoting the activation of pDCs. Finally, evidence is provided that nTregs primed by pDCs displayed reciprocal interleukin-10/transforming growth factor-β cytokine expression profiles compared with nTregs primed by cDCs. This functional diversity further supports a nonredundant tolerogenic role for thymic pDCs in the human thymus.
Collapse
|
16
|
Vavassori S, Shi Y, Chen CC, Ron Y, Covey LR. In vivo post-transcriptional regulation of CD154 in mouse CD4+ T cells. Eur J Immunol 2009; 39:2224-32. [PMID: 19572319 DOI: 10.1002/eji.200839163] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Interactions between CD40 and its ligand CD154 are involved in the progression of both cell mediated and innate immunity. These interactions are brought about by the transient expression of CD154 on activated CD4(+) T cells, which is regulated, in part, at the level of mRNA turnover. Here we have focused on analyzing the pattern of post-transcriptional regulation in mouse CD4(+) T cells in response to activation. Initial experiments identify a region of the murine CD154 mRNA that binds a polypyrimidine tract-binding protein-containing complex (mComplex I), which is activation-dependent and binds to a single CU-rich site within the 3' uTR Subsequent findings demonstrate that in vivo polyclonal activation of T cells leads to a pattern of differential CD154 mRNA stability that is directly dependent on extent of activation. Furthermore, in vitro activation of antigen-primed T cells shows that the CD154 mRNA half-life increases relative to that of unprimed cells. Importantly, this is the first report demonstrating that the regulation of CD154 in vivo is connected to an activation-induced program of mRNA decay and thus provides strong evidence for post-transcriptional mechanisms having a physiological role in regulating CD154 expression during an ongoing immune response.
Collapse
Affiliation(s)
- Stefano Vavassori
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | | | | | | | | |
Collapse
|
17
|
A T-cell-specific CD154 transcriptional enhancer located just upstream of the promoter. Genes Immun 2008; 9:640-9. [PMID: 18719603 DOI: 10.1038/gene.2008.67] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
CD154 (CD40-ligand) is a critical immune regulator. CD154 expression is tightly regulated and largely restricted to activated CD4 T cells. Using DNase I hypersensitivity site (HSS) mapping, we identified two novel HSS mapping to the human CD154 promoter element and just upstream. Both HSS were activation independent and CD4 T-cell specific. Approximately 350 bp of DNA sequence flanking the upstream HSS site was highly conserved between mouse and man, and was rich in binding sites for GATA and NFAT proteins. Gel shift and chromatin immunoprecipitation assays demonstrated both NFAT1 and the Th2 factor, GATA-3, bound this enhancer element in vitro and in vivo, respectively. A PstI/XbaI 345 bp fragment of this region acted as a transcriptional enhancer of the CD154 promoter in primary human CD4 T cells. Overexpression of repressor of GATA and a dominant negative GATA-3 protein independently inhibited transcription, whereas overexpression of wild-type GATA-3 enhanced transcriptional activity, by this element in primary CD4 T cells. Moreover, more interleukin-4-producing CD4 T cells expressed CD154 following activation than interferon-gamma-producing CD4 T cells. Thus, we identified a novel T-cell-specific, GATA-3 responsive, CD154 transcriptional enhancer, which may contribute to increased propensity of Th2 cells to express CD154.
Collapse
|
18
|
Laughlin J, Oghlidos S, Porter JF, Matus-Nicodemos R, Sinquett FL, Marcelli V, Covey LR. Functional analysis of a tripartite stability element within the CD40 ligand 3' untranslated region. Immunology 2008; 124:368-79. [PMID: 18194270 DOI: 10.1111/j.1365-2567.2007.02783.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
We previously identified a cis-acting element within the 3' untranslated region of CD40 ligand messenger RNA (mRNA) that is composed of three complex binding sites and acts to increase mRNA stability in both in vitro and in vivo systems. We now demonstrate the functional consequences of the three binding sites with respect to increasing both luciferase activity and mRNA stability in a heterologous transcript expressed in a T-cell line. The internal region B was shown to be a bona fide stability element because its presence increased luciferase activity fourfold over the unmodified transcript and its removal from the XbaI-HaeIII region resulted in rapid degradation of the transcript. Region A contained both a binding site for a polypyrimidine-tract-binding protein (PTB)-mediated complex (Complex I) and an upstream, adjacent sequence that was a negative regulator of mRNA stability. Region C bound Complex II, which contained both PTB and heterogeneous nuclear ribonucleoproteinL (hnRNPL), and was less effective as a stability element on its own compared to region B. Our findings demonstrate differential levels of activity for the three binding sites relative to the turnover of CD40 ligand mRNA, suggesting that the lack of binding of Complex I/II during the early stages of T-cell activation contributes to the rapid degradation of the CD40 ligand mRNA transcript.
Collapse
Affiliation(s)
- Jennifer Laughlin
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Zhou HJ, Pham LV, Tamayo AT, Lin-Lee YC, Fu L, Yoshimura LC, Ford RJ. Nuclear CD40 interacts with c-Rel and enhances proliferation in aggressive B-cell lymphoma. Blood 2007; 110:2121-7. [PMID: 17567982 PMCID: PMC1976364 DOI: 10.1182/blood-2007-02-073080] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
CD40 is an integral plasma membrane-associated member of the TNF receptor family that has recently been shown to also reside in the nucleus of both normal B cells and large B-cell lymphoma (LBCL) cells. However, the physiological function of CD40 in the B-cell nucleus has not been examined. In this study, we demonstrate that nuclear CD40 interacts with the NF-kappaB protein c-Rel, but not p65, in LBCL cells. Nuclear CD40 forms complexes with c-Rel on the promoters of NF-kappaB target genes, CD154, BLyS/BAFF, and Bfl-1/A1, in various LBCL cell lines. Wild-type CD40, but not NLS-mutated CD40, further enhances c-Rel-mediated Blys promoter activation as well as proliferation in LBCL cells. Studies in normal B cells and LBCL patient cells further support a nuclear transcriptional function for CD40 and c-Rel. Cooperation between nuclear CD40 and c-Rel appears to be important in regulating cell growth and survival genes involved in lymphoma cell proliferation and survival mechanisms. Modulating the nuclear function of CD40 and c-Rel could reveal new mechanisms in LBCL pathophysiology and provide potential new targets for lymphoma therapy.
Collapse
MESH Headings
- B-Cell Activating Factor/genetics
- B-Cell Activating Factor/metabolism
- B-Lymphocytes/metabolism
- CD40 Antigens/genetics
- CD40 Antigens/metabolism
- Cell Nucleus/metabolism
- Cell Proliferation
- Chromatin Immunoprecipitation
- Electrophoretic Mobility Shift Assay
- Genes, rel/physiology
- Humans
- Immunoprecipitation
- Luciferases/metabolism
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/metabolism
- Lymphoma, B-Cell/pathology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Promoter Regions, Genetic
- Thymidine/metabolism
- Transfection
- beta-Galactosidase/metabolism
Collapse
Affiliation(s)
- Hai-Jun Zhou
- Department of Hematopathology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Harhaj NS, Janic B, Ramos JC, Harrington WJ, Harhaj EW. Deregulated expression of CD40 ligand in HTLV-I infection: distinct mechanisms of downregulation in HTLV-I-transformed cell lines and ATL patients. Virology 2007; 362:99-108. [PMID: 17258259 PMCID: PMC1949045 DOI: 10.1016/j.virol.2006.12.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2006] [Revised: 11/09/2006] [Accepted: 12/18/2006] [Indexed: 10/23/2022]
Abstract
HTLV-I infection is associated with the development of adult T cell leukemia (ATL) and the neuroinflammatory disease HAM/TSP. There are quantitative and qualitative differences in the antiviral cytotoxic T cell (CTL) response in ATL and HAM/TSP although the underlying mechanisms are unclear. Here, we demonstrate that the HTLV-I Tax trans-activating protein is a transcriptional activator of CD40 ligand (CD40L), a critical regulator of dendritic cell maturation and adaptive immunity. Tax activates CD40L expression via a cyclosporin A insensitive pathway that is also independent of NF-kappaB. Although Tax upregulates CD40L gene expression, CD40L expression is absent in Tax-expressing HTLV-I-transformed cell lines via an epigenetic mechanism involving methylation. T lymphocytes cultured ex vivo from ATL patients, but not HAM/TSP or normal controls, exhibit a potent block in the induction of CD40L, but not CD69. However, the CD40L gene is not silenced by methylation in ATL patients, thus CD40L is downregulated by distinct mechanisms in HTLV-I-transformed cell lines and ATL patients.
Collapse
Affiliation(s)
- Nicole S Harhaj
- Department of Microbiology and Immunology, Sylvester Comprehensive Cancer Center, The University of Miami, Miller School of Medicine, 1550 NW 10 Avenue, Miami, FL 33136, USA
| | | | | | | | | |
Collapse
|
21
|
Martin-Donaire T, Losada-Fernandez I, Perez-Chacon G, Rua-Figueroa I, Erausquin C, Naranjo-Hernandez A, Rosado S, Sanchez F, Garcia-Saavedra A, Citores MJ, Vargas JA, Perez-Aciego P. Association of the microsatellite in the 3' untranslated region of the CD154 gene with rheumatoid arthritis in females from a Spanish cohort: a case-control study. Arthritis Res Ther 2007; 9:R89. [PMID: 17845713 PMCID: PMC2212561 DOI: 10.1186/ar2288] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Revised: 08/14/2007] [Accepted: 09/10/2007] [Indexed: 02/08/2023] Open
Abstract
CD40-CD154 interaction is an important mediator of inflammation and has been implicated in T helper type 1-mediated autoimmune diseases including rheumatoid arthritis (RA). Linkage studies have shown association of markers in the proximity of the CD154 gene. In the present work we investigated whether specific allele variants of the microsatellite in the 3' UTR of the CD154 gene might modulate the risk of RA. The study, in a case-control setting, included 189 patients and 150 healthy controls from the Canary Islands, Spain. The 24CAs allele was less represented in female patients than in controls (0.444 in controls versus 0.307 in patients, P = 0.006, odds ratio (OR) 0.556, 95% confidence interval (CI) 0.372 to 0.831) but not in males (0.414 versus 0.408), and only when homozygous (P = 0.012; OR 0.35, 95% CI 0.16 to 0.77). We also verified that CD154 association with RA was independent of human leukocyte antigen (HLA) phenotype. A further functional study showed that after stimulation anti-CD3, CD154 mRNA was more stable in CD4+ T lymphocytes from patients with RA bearing the 24CAs allele (mRNA half-life 208 minutes) than in patients without the 24CAs allele (109 minutes, P = 0.009). However, a lower percentage of CD154+CD4+ T lymphocytes was seen in freshly isolated peripheral blood mononuclear cells from patients carrying 24CAs alleles (mean 4.28 versus 8.12; P = 0.033), and also in CD4+ T lymphocytes stimulated with anti-CD3 (median 29.40 versus 47.60; P = 0.025). These results were concordant with the smaller amounts of CD154 mRNA isolated from stimulated T lymphocytes with 24CAs alleles. The CD154 microsatellite therefore seems to affect the expression of the gene in a complex manner that implies not only mRNA stability. These data suggest that the CD154 microsatellite contributes to the regulation of mRNA and protein expression, although further studies will be necessary to elucidate its role in disease predisposition.
Collapse
Affiliation(s)
- Trinidad Martin-Donaire
- Fundacion LAIR, Madrid, Spain
- Servicio de Medicina Interna I, Hospital Universitario Puerta de Hierro, Universidad Autonoma de Madrid, C/San Martin de Porres 4, 28035 Madrid, Spain
| | | | | | - Iñigo Rua-Figueroa
- Servicio de Reumatologia, Hospital Universitario de Gran Canaria Doctor Negrin, Barranco de la Ballena s/n, 35010 Las Palmas de Gran Canaria, Spain
| | - Celia Erausquin
- Servicio de Reumatologia, Hospital Universitario de Gran Canaria Doctor Negrin, Barranco de la Ballena s/n, 35010 Las Palmas de Gran Canaria, Spain
| | - Antonio Naranjo-Hernandez
- Servicio de Reumatologia, Hospital Universitario de Gran Canaria Doctor Negrin, Barranco de la Ballena s/n, 35010 Las Palmas de Gran Canaria, Spain
| | | | - Florentino Sanchez
- Servicio de Inmunologia, Hospital Universitario de Gran Canaria Doctor Negrin, Barranco de la Ballena s/n, 35010 Las Palmas de Gran Canaria, Spain
| | - Ayoze Garcia-Saavedra
- Servicio de Inmunologia, Hospital Universitario de Gran Canaria Doctor Negrin, Barranco de la Ballena s/n, 35010 Las Palmas de Gran Canaria, Spain
| | - Maria Jesus Citores
- Servicio de Medicina Interna I, Hospital Universitario Puerta de Hierro, Universidad Autonoma de Madrid, C/San Martin de Porres 4, 28035 Madrid, Spain
| | - Juan A Vargas
- Servicio de Medicina Interna I, Hospital Universitario Puerta de Hierro, Universidad Autonoma de Madrid, C/San Martin de Porres 4, 28035 Madrid, Spain
| | | |
Collapse
|
22
|
Steiper ME, Parikh SJ, Zichello JM. Phylogenetic analysis of the promoter region of the CD40L gene in primates and other mammals. INFECTION GENETICS AND EVOLUTION 2006; 8:406-13. [PMID: 17275421 DOI: 10.1016/j.meegid.2006.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2006] [Revised: 12/12/2006] [Accepted: 12/14/2006] [Indexed: 11/15/2022]
Abstract
CD40L is a type II membrane protein comprised of 261 amino acids. CD40L plays a crucial role in the immune system where it is primarily expressed on activated T cells and triggers immunoglobulin class switching. The genetic disease X-linked hypergammaglobulinemia (HIGM1, XHIGM or XHIM) is caused by mutations in the CD40L gene. Individuals with HIGM1 are susceptible to recurrent infections to pathogens and a relationship has been shown to exist with malaria [Sabeti, P., Usen, S., Farhadian, S., Jallow, M., Doherty, T., Newport, M., Pinder, M., Ward, R., Kwiatkowski, D., 2002a. CD40L association with protection from severe malaria. Genes Immun. 3, 286-291]. In this paper, we phylogenetically examine the promoter region of CD40L in primates and other mammals via phylogenetic shadowing. This analysis revealed several regions of the CD40L promoter that were highly constrained and thereby inferred to be functional. These constrained regions confirmed many known regulatory sites. In addition, a novel, highly constrained upstream region was also identified which had an NF-AT recognition motif. These analyses also showed that the different mammal groups do not share an exactly similar set of promoter binding sites and taxon-specific promoters have evolved.
Collapse
Affiliation(s)
- Michael E Steiper
- Department of Anthropology, Hunter College of the City University of New York, New York, NY 10021, United States.
| | | | | |
Collapse
|
23
|
Cron RQ, Bandyopadhyay R, Genin A, Brunner M, Kersh GJ, Yin J, Finkel TH, Crow MK. Early growth response-1 is required for CD154 transcription. THE JOURNAL OF IMMUNOLOGY 2006; 176:811-8. [PMID: 16393964 PMCID: PMC1424665 DOI: 10.4049/jimmunol.176.2.811] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD154 (CD40 ligand) expression on CD4 T cells is normally tightly controlled, but abnormal or dysregulated expression of CD154 has been well documented in autoimmune diseases, such as systemic lupus erythematosus. Beyond regulation by NFAT proteins, little is known about the transcriptional activation of the CD154 promoter. We identified a species-conserved purine-rich sequence located adjacent to the CD154 transcriptional promoter proximal NFAT site, which binds early growth response (Egr) transcription factors. Gel shift assays and chromatin immunoprecipitation assays reveal that Egr-1, Egr-3, and NFAT1 present in primary human CD4 T cells are capable of binding this combinatorial site in vitro and in vivo, respectively. Multimerization of this NFAT/Egr sequence in the context of a reporter gene demonstrates this sequence is transcriptionally active upon T cell activation in primary human CD4 T cells. Overexpression of Egr-1, but not Egr-3, is capable of augmenting transcription of this reporter gene as well as that of an intact CD154 promoter. Conversely, overexpression of small interfering RNA specific for Egr-1 in primary human CD4 T cells inhibits CD154 expression. Similarly, upon activation, CD154 message is notably decreased in splenic CD4 T cells from Egr-1-deficient mice compared with wild-type controls. Our data demonstrate that Egr-1 is required for CD154 transcription in primary CD4 T cells. This has implications for selective targeting of Egr family members to control abnormal expression of CD154 in autoimmune diseases such as systemic lupus erythematosus.
Collapse
Affiliation(s)
- Randy Q Cron
- Division of Rheumatology, Children's Hospital of Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Clarke MW, Ward NC, Wu JHY, Hodgson JM, Puddey IB, Croft KD. Supplementation with mixed tocopherols increases serum and blood cell gamma-tocopherol but does not alter biomarkers of platelet activation in subjects with type 2 diabetes. Am J Clin Nutr 2006; 83:95-102. [PMID: 16400056 DOI: 10.1093/ajcn/83.1.95] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Some studies have shown potential benefit of vitamin E on platelet function, but several clinical trials failed to show improved cardiovascular outcome with alpha-tocopherol supplementation. Gamma-tocopherol, a major dietary form of vitamin E, may have protective properties different from those of alpha-tocopherol. OBJECTIVE We compared the effects of supplementation with alpha-tocopherol (500 mg) and a gamma-tocopherol-rich compound (500 mg, containing 60% gamma-tocopherol) on serum and cellular tocopherol concentrations, urinary tocopherol metabolite excretion, and in vivo platelet activation in subjects with type 2 diabetes. DESIGN Fifty-eight subjects were randomly assigned to receive either 500 mg alpha-tocopherol/d, 500 mg mixed tocopherols/d, or matching placebo. Serum, erythrocyte, and platelet tocopherol and urinary metabolite concentrations were measured at baseline and after the 6-wk intervention. Soluble CD40 ligand, urinary 11-dehydro-thromboxane B2, serum thromboxane B2, soluble P-selectin, and von Willebrand factor were measured as biomarkers of in vivo platelet activation. RESULTS Serum alpha-tocopherol increased with both tocopherol treatments. Serum and cellular gamma-tocopherol increased 4-fold (P < 0.001) in the mixed tocopherol group, whereas red blood cell gamma-tocopherol decreased significantly after alpha-tocopherol supplementation. Excretion of alpha-carboxyethyl-hydroxychroman increased significantly after supplementation with alpha-tocopherol and mixed tocopherols. Excretion of gamma-carboxyethyl-hydroxychroman increased significantly after supplementation with mixed tocopherols and after that with alpha-tocopherol, which may reflect the displacement of gamma-tocopherol by alpha-tocopherol due to incorporation of the latter into lipoproteins in the liver. Neither treatment had any significant effect on markers of platelet activation. CONCLUSIONS Supplementation with alpha-tocopherol decreased red blood cell gamma-tocopherol, whereas mixed tocopherols increased both serum alpha-tocopherol and serum and cellular gamma-tocopherol. Changes in serum tocopherol closely reflect changes in cellular concentrations of tocopherols after supplementation.
Collapse
Affiliation(s)
- Michael W Clarke
- Department of Core Clinical Pathology and Biochemistry, Royal Perth Hospital, University of Western Australia, Perth, Australia
| | | | | | | | | | | |
Collapse
|
25
|
Pham LV, Tamayo AT, Yoshimura LC, Lin-Lee YC, Ford RJ. Constitutive NF-kappaB and NFAT activation in aggressive B-cell lymphomas synergistically activates the CD154 gene and maintains lymphoma cell survival. Blood 2005; 106:3940-7. [PMID: 16099873 PMCID: PMC1895110 DOI: 10.1182/blood-2005-03-1167] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Abnormalities in B-lymphocyte CD40 ligand (CD154) expression have been described for a number of immunologic diseases, including B-cell lymphomas. Although functional analysis of the CD154 gene and protein has been extensive, little is known about the mechanisms controlling CD154 expression in activated T cells, and even less is known for normal and malignant B cells. In this study we describe the transcriptional mechanism controlling CD154 expression in large B-cell lymphoma (LBCL). We show that the nuclear factor of activated T cells (NFAT) transcription factor is also constitutively activated in LBCL. We demonstrate that the constitutively active NFATc1 and c-rel members of the NFAT and nuclear factor-kappaB (NF-kappaB) families of transcription factors, respectively, directly interact with each other, bind to the CD154 promoter, and synergistically activate CD154 gene transcription. Down-regulation of NFATc1 or c-rel with small interfering RNA (siRNA) or chemical inhibitors inhibits CD154 gene transcription and lymphoma cell growth. These findings suggest that targeting NF-kappaB and NFAT, by inhibiting the expression of these transcription factors, or interdicting their interaction may provide a therapeutic rationale for patients with non-Hodgkin lymphoma of B-cell origin, and possibly other disorders that display dysregulated CD154 expression.
Collapse
Affiliation(s)
- Lan V Pham
- Department of Hematopathology, Box 54, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
26
|
Abstract
With increasing age, the ability to produce protective antibodies in response to immunization declines, resulting in reduced efficacy of vaccination. We have examined how reductions in CD4(+) T-cell function contribute to reduced humoral responses, using a model that allows us to compare identical numbers of antigen-specific naive T cells from young and aged T-cell receptor transgenic mice. Naive cells from aged mice exhibit reduced responses, both in vitro and in vivo. In vitro, responses of aged T cells can be enhanced by addition of interleukin (IL)-2. In vivo, using an adoptive transfer model with young hosts, naive cells from aged mice exhibit significant reductions in cognate helper function, leading to reduced B-cell expansion and differentiation. These age-related defects could be overcome by prior in vitro T helper 2 effector generation with aged T cells. This improvement in cognate function of the aged effectors may be related to the enhancement of CD154 expression, which occurs on aged T cells in the presence of exogenous IL-2. We also found no difference in B-cell expansion and differentiation when young cells were transferred to young or aged hosts. Our results indicate that age-related reductions in humoral responses are mainly due to defects in the cognate helper function of naive CD4(+) T cells from aged individuals.
Collapse
|
27
|
Eaton SM, Burns EM, Kusser K, Randall TD, Haynes L. Age-related defects in CD4 T cell cognate helper function lead to reductions in humoral responses. J Exp Med 2004; 200:1613-22. [PMID: 15611289 PMCID: PMC2211991 DOI: 10.1084/jem.20041395] [Citation(s) in RCA: 207] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2004] [Accepted: 11/09/2004] [Indexed: 01/16/2023] Open
Abstract
With increasing age, the ability to produce protective antibodies in response to immunization declines, leading to a reduced efficacy of vaccination in the elderly. To examine the effect of age on the cognate function of CD4 T cells, we have used a novel adoptive transfer model that allows us to compare identical numbers of antigen-specific naive T cells from young and aged TCR transgenic (Tg) donors. Upon transfer of aged donor CD4 T cells to young hosts, there was significantly reduced expansion and germinal center (GC) differentiation of the antigen-specific B cell population after immunization. This reduced cognate helper function was seen at all time points and over a wide range of donor cell numbers. In hosts receiving aged CD4 cells, there were also dramatically lower levels of antigen-specific IgG. These age-related defects were not due to defects in migration of the aged CD4 T cells, but may be attributable to reduced CD154 (CD40L) expression. Furthermore, we found that there was no difference in B cell expansion and differentiation or in IgG production when young CD4 T cells were transferred to young or aged hosts. Our results show that, in this model, age-related reductions in the cognate helper function of CD4 T cells contribute significantly to defects in humoral responses observed in aged individuals.
Collapse
Affiliation(s)
- Sheri M Eaton
- Trudeau Institute, 154 Algonquin Ave., Saranac Lake, NY 12983, USA
| | | | | | | | | |
Collapse
|
28
|
Shibuya TY, Kim S, Nguyen K, Do J, McLaren CE, Li KT, Chen WP, Parikh P, Wadhwa A, Zi X, Chen VY, Wong HS, Armstrong WB, Yoo GH. Bioactive Suture. Clin Cancer Res 2004; 10:7088-99. [PMID: 15501989 DOI: 10.1158/1078-0432.ccr-04-0052] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We have proposed to characterize the mechanism through which bioactive surgical sutures generate a T(H)1 immune response and to define the immune-stimulating half-life of the sutures. EXPERIMENTAL DESIGN Bioactive sutures of interferon gamma (IFNgamma), interleukin 2 (IL-2), anti-CD3/CD28, anti-CD3/CD28 + IL-2, or anti-CD3/CD28 + IFNgamma sutures were used to stimulate lymphocytes from normal donors and from head and neck cancer patients in vitro over a 24-day period. Cell supernatants were analyzed by ELISA, and T cells were phenotyped to characterize the immune response generated. Intracellular cytokine staining was performed to measure the expansion of flu-specific T cells. Electromobility shift assay and supershift assay were used to measure the intranuclear DNA binding activity of nuclear factor kappaB and its p65 subunit in T cells activated by sutures in the presence and absence of a proteasome inhibitor, MG-132. RESULTS Anti-CD3/CD28, anti-CD3/CD28 + IL-2, or anti-CD3/CD28 + IFNgamma generated a prolonged T(H)1 immune response for 18 days in vitro. Anti-CD3/CD28 expanded flu-specific T cells. Activated T cells demonstrated enhanced CD40 ligand (CD40L) expression within 72 hours of stimulation, which stimulated other cells to secrete IL-12. Stimulated T cells demonstrated increased intranuclear expression of nuclear factor-kappaB, which was blocked by MG-132, and also reduced CD40L and IL-12 expression. CONCLUSIONS This is the first report to demonstrate that bioactive surgical sutures can generate a prolonged T(H)1 immune response and expand flu-specific T cells. Bioactive sutures, which are primarily a T-cell stimulant, also stimulated other cells to secrete IL-12 and prolonged the immune response. Sutures may provide a novel in situ stimulating strategy for enhancing the immune system of cancer patients.
Collapse
Affiliation(s)
- Terry Y Shibuya
- Department of Otolaryngology/Head & Neck Surgery, University of California Irvine College of Medicine, Orange, California 92868, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
CD154 (CD40-ligand) has a wide variety of pleiotropic effects throughout the immune system and is critical to both cellular and humoral immunity. Cell surface and soluble CD154 are primarily expressed by activated CD4 T cells. Expression of CD154 is tightly regulated in a time-dependent manner, and, like most T cell-derived cytokines and other members of the tumor necrosis factor (TNF) superfamily, CD154 is largely regulated at the level of gene transcription. Recently, dysregulated expression of CD154 has been noted in a number of autoimmune disorders, including systemic lupus erythematosus (SLE). In addition, abnormal expression of CD154 has been hypothesized to contribute to a wider array of diseases, from atherosclerosis to Alzheimer's disease. Until recently, very little was known about the transcriptional regulation of CD154. We are exploring CD154 regulation in primary human CD4 T cells in hopes of understanding the cis- and trans-regulatory elements that control its expression in the cells that normally express CD154. Ultimately, we hope to be able to correct abnormal expression of CD154 in various disease states and to help design gene therapy vectors for treating CD154-deficient individuals with hyper-IgM syndrome.
Collapse
Affiliation(s)
- Randy Q Cron
- Children's Hospital of Philadelphia and Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-4318, USA.
| |
Collapse
|
30
|
Zhang R, Fichtenbaum CJ, Hildeman DA, Lifson JD, Chougnet C. CD40 Ligand Dysregulation in HIV Infection: HIV Glycoprotein 120 Inhibits Signaling Cascades Upstream of CD40 Ligand Transcription. THE JOURNAL OF IMMUNOLOGY 2004; 172:2678-86. [PMID: 14764743 DOI: 10.4049/jimmunol.172.4.2678] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-12 production and up-regulation of CD40 ligand (CD40L) expression are impaired in the PBMC of HIV-infected donors, and exogenous CD40L rescues IL-12 production by such cells. In this study, we implicate dysregulation of CD40L expression in the IL-12 defect associated with HIV by demonstrating that induction of CD40L expression by anti-CD3/CD28 stimulation was directly correlated with the IL-12 productive capacity of PBMC. Further, we demonstrate marked decreases in the induction of CD40L protein and mRNA following anti-CD3/CD28 stimulation in HIV-infected donors compared with uninfected donors, with a tight association between these two levels. Inhibition of CD40L up-regulation was selective, as induction of CD69 or OX40 was not as severely affected. Increased instability of CD40L mRNA did not constitute a major mechanism in CD40L dysregulation, thus suggesting a potential defect in the signaling cascades upstream of transcription. The mechanisms by which HIV infection affects the induction of CD40L expression appear to involve HIV gp120-mediated engagement of CD4. Indeed, anti-CD4 mAb or inactivated HIV virions that harbor a conformationally intact gp120 significantly inhibited CD40L up-regulation at both the protein and mRNA levels. This inhibition was due to the native, virion-associated gp120, as coculture with soluble CD4 or heat treatment of inactivated HIV abolished their effect. These in vitro models mirror the CD40L defect seen in cells from HIV-infected donors and thus provide a suitable model to investigate HIV-induced CD40L dysregulation. Clear elucidation of mechanism(s) may well lead to the development of novel immunotherapeutic approaches to HIV infection.
Collapse
Affiliation(s)
- Rui Zhang
- Divisions of Molecular Immunology and Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA
| | | | | | | | | |
Collapse
|
31
|
Chougnet C. Role of CD40 ligand dysregulation in HIV-associated dysfunction of antigen-presenting cells. J Leukoc Biol 2003; 74:702-9. [PMID: 12960257 DOI: 10.1189/jlb.0403171] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cellular interactions between antigen-presenting cells and activated CD4+ T cells are central to the regulation of adaptive immunity. Among the many receptor-ligand pairs involved, the critical importance of CD40-CD40 Ligand (CD40L) interactions has been demonstrated in many experimental systems. Dysregulation of antigen-presenting cell function is a hallmark of HIV-associated defects in cell-mediated immunity. Much evidence suggests a mechanistic role for defective CD40-CD40L interactions in such a defect. Consistent with this hypothesis, the capacity to upregulate CD40L on purified CD4+ T cells becomes progressively impaired in HIV infection, in parallel with the progression of clinical immunosuppression. The mechanisms underlying CD40L dysregulation in HIV infection remain unknown. Because CD40L expression is tightly regulated (transcriptionally, post-transcriptionally and post-translationally), HIV may interfere at several levels. However, a transcriptional defect in CD40L expression, mediated by the engagement of CD4 by HIV gp120, appears to play a primary role. Clear elucidation of mechanism may well lead to the development of novel immunotherapeutic approaches to HIV infection.
Collapse
Affiliation(s)
- Claire Chougnet
- Cincinati Children's Hospital Research Foundation, Cincinnati, USA.
| |
Collapse
|
32
|
Longo CR, Arvelo MB, Patel VI, Daniel S, Mahiou J, Grey ST, Ferran C. A20 protects from CD40-CD40 ligand-mediated endothelial cell activation and apoptosis. Circulation 2003; 108:1113-8. [PMID: 12885753 DOI: 10.1161/01.cir.0000083718.76889.d0] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND CD40/CD40 ligand (CD40L) signaling is a potent activator of endothelial cells (ECs) and promoter of atherosclerosis. In this study, we investigate whether A20 (a gene we have shown to be antiinflammatory and antiapoptotic in ECs) can protect from CD40/CD40L-mediated EC activation. METHODS AND RESULTS Overexpression of CD40, in a transient transfection system, activates the transcription factor NF-kappaB and upregulates IkappaBalpha, E-selectin, and tissue factor (TF) reporter activity. Coexpression of A20 inhibits NF-kappaB and upregulation of IkappaBalpha and E-Selectin but not TF, suggesting that CD40 induces TF in a non-NF-kappaB-dependent manner. In human coronary artery ECs (HCAECs), adenovirus-mediated overexpression of A20 blocks physiological, CD40-induced activation of NF-kappaB, upstream of IkappaBalpha degradation (Western blot) and subsequently upregulation of ICAM-1, VCAM-1, and E-selectin (flow cytometry). Although A20 does not block TF transcription its expression in HCAECs inhibits TF induction (colorimetric assay and RT-PCR) by blunting CD40 upregulation. We demonstrate that CD40 signaling induces apoptosis in a proinflammatory microenvironment. A20 overexpression protects from CD40-mediated EC apoptosis (DNA content analysis and trypan blue exclusion). We also demonstrate that signaling through CD40L activates NF-kappaB and induces apoptosis in ECs, both of which are inhibited by A20 overexpression. CONCLUSIONS A20 works at multiple levels to protect ECs from CD40/CD40L mediated activation and apoptosis. A20-based therapy could be beneficial for the treatment of vascular diseases such as atherosclerosis and transplant-associated vasculopathy.
Collapse
Affiliation(s)
- Christopher R Longo
- Immunobiology Research Center, Department of Surgery and Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Mass 02215, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Haaning Andersen AD, Lange M, Lillevang ST. Allelic variation of the inducible costimulator (ICOS) gene: detection of polymorphisms, analysis of the promoter region, and extended haplotype estimation. TISSUE ANTIGENS 2003; 61:276-85. [PMID: 12753665 DOI: 10.1034/j.1399-0039.2003.00019.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The human chromosome region 2q33 including the three costimulatory molecules CD28, CTLA-4 and ICOS, has been subject to much attention due to its linkage to a number of autoimmune diseases. The search for the causal relationship of this linkage has revealed several polymorphisms, but no variations in the amino acid sequences, except for one polymorphism in the leader sequence of CTLA-4. In the present study, we examined the ICOS gene of an unrelated group of healthy donors from the Danish population. We were able to report 16 intronic SNP, one intronic G-insert and two repeat regions in intron 4, consistent with the [T]n and the [GT]n regions reported in a Japanese study. Putative haplotypes for the established SNP and repeat polymorphisms have been estimated by computational analysis. Sequencing of approximately 3500 bp of the upstream region of ICOS revealed an additional eight SNP of which two resided in putative NF-kB and Sp1 sites. In accordance with previous studies we detected no variations in the coding regions except for a rare polymorphism that was found in one donor in the last codon of exon 5, which lead to a heterozygous genotype, but no amino acid change. This suggests that regulation of transcription rather than protein structure could be a possible mechanism in the explanation of linkage.
Collapse
Affiliation(s)
- A D Haaning Andersen
- Department of Clinical Immunology, Odense University Hospital, Denmark BioCentrum, Technical University of Denmark, 2800 Lyngby, Denmark.
| | | | | |
Collapse
|
34
|
Kosinski PA, Laughlin J, Singh K, Covey LR. A complex containing polypyrimidine tract-binding protein is involved in regulating the stability of CD40 ligand (CD154) mRNA. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:979-88. [PMID: 12517964 DOI: 10.4049/jimmunol.170.2.979] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
CD40 ligand (CD154) expression has been shown to be regulated, in part, at the posttranscriptional level by a pathway of "regulated instability" of mRNA decay throughout a time course of T cell activation. This pathway is modulated at late times of activation by the binding of a stability complex (termed complex I) to a CU-rich region in the 3' untranslated region of the CD154 message. We have undertaken experiments to extend these findings and to analyze the cis-acting elements and trans-acting factors involved in this regulation. We have previously shown that the minimal binding sequence for complex I is a 63 nt CU-rich motif. However, our current study shows that when this site was deleted additional complex binding was observed upstream and downstream of the minimal binding region. Only after deletion of an extended region (termed Delta1515) was complex binding completely abolished. Analysis of complex binding using competition experiments revealed that the three adjacent regions bound related but not identical complexes. However, all three sites appeared to have a 55-kDa protein as the RNA-binding protein. Deletion of the Delta1515 region resulted in reduced transcript stability as measured by both in vitro and in vivo decay assays. Finally, using Abs against known RNA-binding proteins, we identified the polypyrimidine tract-binding protein (or heterogeneous nuclear ribonucleoprotein I) as a candidate RNA-binding component of complex I.
Collapse
Affiliation(s)
- Penelope A Kosinski
- Department of Cell Biology and Neuroscience, Rutgers, State University of New Jersey, Piscataway 08854, USA
| | | | | | | |
Collapse
|
35
|
Abstract
Recognition of immunodeficiency allows steps to be taken to minimize morbidity and mortality. Immunodeficiency can be secondary to viral infection, most importantly secondary to HIV-1 worldwide, medications, disruption of the usual infection clearance mechanisms, or secondary to a myriad of systemic disorders. Immunodeficiency may also be due to one of the growing list of primary immunodeficiency disorders. In infancy, lymphopenia should trigger an evaluation investigating the possibility of severe combined immunodeficiency. Evaluations of children should be done keeping in mind that normal numbers of lymphocytes are higher in children than in adults, immunoglobulin levels in children are lower than in adults in younger age groups, and antibody production in response to polysaccharide antigens is not usually fully developed in the less-than 2-year-old child.
Collapse
Affiliation(s)
- Mary E Paul
- Section of Allergy/Immunology, Texas Children's Hospital, 6621 Fannin Street, MC: 1-3291, Houston, TX 77030, USA.
| |
Collapse
|
36
|
Schubert LA, Cron RQ, Cleary AM, Brunner M, Song A, Lu LS, Jullien P, Krensky AM, Lewis DB. A T cell-specific enhancer of the human CD40 ligand gene. J Biol Chem 2002; 277:7386-95. [PMID: 11751888 DOI: 10.1074/jbc.m110350200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We observed that the human CD40 ligand (CD40L) gene 5'-flanking region conferred weak promoter activity in activated CD4 T cells, suggesting that additional regions are required for optimal CD40L gene transcription. We therefore examined a 3'-flanking segment of the CD40L gene, which contained a putative NF-kappaB/Rel cis-element, for its ability to enhance CD40L promoter function. This segment augmented CD40L promoter activity in an orientation-independent manner in CD4 T-lineage cells but not in human B cell or monocyte cell lines. Mapping of CD4 T-lineage cell nuclei identified a DNase I-hypersensitive site in the flanking region near the NF-kappaB/Rel sequence, suggesting a transcriptional regulatory role. This was further supported by truncation analysis and site-directed mutagenesis, which indicated that the CD40L 3'-flanking NF-kappaB/Rel cis-element was critical for enhancer function. Electrophoretic mobility shift assays showed that the cis-element preferentially bound the p50 form of the NF-kappaB1 gene contained in human T cell nuclear protein extracts. This binding also appeared to occur in vivo in CD4 T cells based on chromatin immunoprecipitation assays using NF-kappaB p50-specific antiserum. Together, these results suggest that the CD40L gene 3'-flanking region acts as a T cell-specific classical transcriptional enhancer by a NF-kappaB p50-dependent mechanism.
Collapse
MESH Headings
- B-Lymphocytes
- Base Sequence
- Binding Sites
- Binding, Competitive
- CD4 Antigens/biosynthesis
- CD40 Ligand/metabolism
- Cell Line
- Cell Nucleus/metabolism
- DNA/metabolism
- DNA, Complementary/metabolism
- Deoxyribonuclease I/metabolism
- Dose-Response Relationship, Drug
- Enhancer Elements, Genetic
- Genes, Reporter
- Humans
- Interleukin-2/metabolism
- Jurkat Cells
- Models, Genetic
- Molecular Sequence Data
- Monocytes/metabolism
- Mutagenesis, Site-Directed
- NF-kappa B/chemistry
- NF-kappa B/metabolism
- NF-kappa B p50 Subunit
- Plasmids/metabolism
- Precipitin Tests
- Promoter Regions, Genetic
- Protein Binding
- Proto-Oncogene Proteins c-rel/metabolism
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- Lisa A Schubert
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
The vertebrate transcription factor NF-kappaB is induced by over 150 different stimuli. Active NF-kappaB, in turn, participates in the control of transcription of over 150 target genes. Because a large variety of bacteria and viruses activate NF-kappaB and because the transcription factor regulates the expression of inflammatory cytokines, chemokines, immunoreceptors, and cell adhesion molecules, NF-kappaB has often been termed a 'central mediator of the human immune response'. This article contains a complete listing of all NF-kappaB inducers and target genes described to date. The collected data argue that NF-kappaB functions more generally as a central regulator of stress responses. In addition, NF-kappaB activation blocks apoptosis in several cell types. Coupling stress responsiveness and anti-apoptotic pathways through the use of a common transcription factor may result in increased cell survival following stress insults.
Collapse
Affiliation(s)
- H L Pahl
- Department of Experimental Anesthesiology, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|