Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Vaccines (Basel) ; 12(3)2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38543915

ABSTRACT

It is clear that new approaches are needed to promote broadly protective immunity to viral pathogens, particularly those that are prone to mutation and escape from antibody-mediated immunity. CD4+ T cells, known to target many viral proteins and highly conserved peptide epitopes, can contribute greatly to protective immunity through multiple mechanisms. Despite this potential, CD4+ T cells are often poorly recruited by current vaccine strategies. Here, we have analyzed a promising new adjuvant (R-DOTAP), as well as conventional adjuvant systems AddaVax with or without an added TLR9 agonist CpG, to promote CD4+ T cell responses to the licensed vaccine Flublok containing H1, H3, and HA-B proteins. Our studies, using a preclinical mouse model of vaccination, revealed that the addition of R-DOTAP to Flublok dramatically enhances the magnitude and functionality of CD4+ T cells specific for HA-derived CD4+ T cell epitopes, far outperforming conventional adjuvant systems based on cytokine EliSpot assays and multiparameter flow cytometry. The elicited CD4+ T cells specific for HA-derived epitopes produce IL-2, IFN-γ, IL-4/5, and granzyme B and have multifunctional potential. Hence, R-DOTAP, which has been verified safe by human studies, can offer exciting opportunities as an immune stimulant for next-generation prophylactic recombinant protein-based vaccines.

2.
bioRxiv ; 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38370737

ABSTRACT

Protein S (PS), the critical plasma cofactor for the anticoagulants tissue factor (TF) pathway inhibitor (TFPI) and activated protein C (APC), circulates in two functionally distinct pools: free (anticoagulant) or bound to complement component 4b-binding protein (C4BP) (anti-inflammatory). Acquired free PS deficiency is detected in several viral infections, but its cause is unclear. Here, we identified a shear-dependent interaction between PS and von Willebrand Factor (VWF) by mass spectrometry. Consistently, plasma PS and VWF comigrated in both native and agarose gel electrophoresis. The PS/VWF interaction was blocked by TFPI but not APC, suggesting an interaction with the C-terminal sex hormone binding globulin (SHBG) region of PS. Microfluidic systems, mimicking arterial laminar flow or disrupted turbulent flow, demonstrated that PS stably binds VWF as VWF unfolds under turbulent flow. PS/VWF complexes also localized to platelet thrombi under laminar arterial flow. In thrombin generation-based assays, shearing plasma decreased PS activity, an effect not seen in the absence of VWF. Finally, free PS deficiency in COVID-19 patients, measured using an antibody that binds near the C4BP binding site in SHBG, correlated with changes in VWF, but not C4BP, and with thrombin generation. Our data suggest that PS binds to a shear-exposed site on VWF, thus sequestering free PS and decreasing its anticoagulant activity, which would account for the increased thrombin generation potential. As many viral infections present with free PS deficiency, elevated circulating VWF, and increased vascular shear, we propose that the PS/VWF interaction reported here is a likely contributor to virus-associated thrombotic risk.

3.
Viruses ; 15(2)2023 02 04.
Article in English | MEDLINE | ID: mdl-36851646

ABSTRACT

Adjuvants are essential components of subunit vaccines added to enhance immune responses to antigens through immunomodulation. Very few adjuvants have been approved for human use by regulatory agencies due to safety concerns. Current subunit vaccine adjuvants approved for human use are very effective in promoting humoral immune responses but are less effective at promoting T-cell immunity. In this study, we evaluated a novel pure enantio-specific cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (R-DOTAP) as an immunomodulator for subunit vaccines capable of inducing both humoral- and cellular-mediated immunity. Using recombinant protein antigens derived from SARS-CoV2 spike or novel computationally optimized broadly reactive influenza antigen (COBRA) proteins, we demonstrated that R-DOTAP nanoparticles promoted strong cellular- and antibody-mediated immune responses in both monovalent and bivalent vaccines. R-DOTAP-based vaccines induced antigen-specific and polyfunctional CD8+ and CD4+ effector T cells and memory T cells, respectively. Antibody responses induced by R-DOTAP showed a balanced Th1/Th2 type immunity, neutralizing activity and protection of mice from challenge with live SARS-CoV2 or influenza viruses. R-DOTAP also facilitated significant dose sparing of the vaccine antigens. These studies demonstrate that R-DOTAP is an excellent immune stimulator for the production of next-generation subunit vaccines containing multiple recombinant proteins.


Subject(s)
COVID-19 , RNA, Viral , Animals , Humans , Mice , Adjuvants, Immunologic , Cations , COVID-19/prevention & control , Fatty Acids, Monounsaturated , Immunity , Lipids , SARS-CoV-2 , Vaccines, Synthetic/genetics , Antibodies, Viral/immunology
4.
Viruses ; 15(2)2023 02 15.
Article in English | MEDLINE | ID: mdl-36851752

ABSTRACT

It is clear that new approaches are needed to promote broadly protective immunity to viral pathogens, particularly those that are prone to mutation and escape from antibody-mediated immunity. Prototypic pathogens of this type are influenza and SARS-CoV-2, where the receptor-binding protein exhibits extremely high variability in its receptor-binding regions. T cells, known to target many viral proteins, and within these, highly conserved peptide epitopes, can contribute greatly to protective immunity through multiple mechanisms but are often poorly recruited by current vaccine strategies. Here, we have studied a promising novel pure enantio-specific cationic lipid 1,2-dioleoyl-3-trimethylammonium-propane (R-DOTAP), which was previously recognized for its ability to generate anti-tumor immunity through the induction of potent cytotoxic CD8 T cells. Using a preclinical mouse model, we have assessed an R-DOTAP nanoparticle adjuvant system for its ability to promote CD4 T cell responses to vaccination with recombinant influenza protein. Our studies revealed that R-DOTAP consistently outperformed a squalene-based adjuvant emulsion, even when it was introduced with a potent TLR agonist CpG, in the ability to elicit peptide epitope-specific CD4 T cells when quantified by IFN-γ and IL-2 ELISpot assays. Clinical testing of R-DOTAP containing vaccines in earlier work by others has demonstrated an acceptable safety profile. Hence, R-DOTAP can offer exciting opportunities as an immune stimulant for next-generation prophylactic recombinant protein-based vaccines.


Subject(s)
COVID-19 , Influenza Vaccines , Influenza, Human , Nanoparticles , Animals , Mice , Humans , Influenza, Human/prevention & control , Hemagglutinins , Squalene , CD4-Positive T-Lymphocytes , SARS-CoV-2 , Adjuvants, Immunologic , Vaccines, Synthetic , Vaccination , Cations
5.
J Immunol ; 202(12): 3524-3536, 2019 06 15.
Article in English | MEDLINE | ID: mdl-31053626

ABSTRACT

Certain types of cationic lipids have shown promise in cancer immunotherapy, but their mechanism of action is poorly understood. In this study, we describe the properties of an immunotherapeutic consisting of the pure cationic lipid enantiomer R-1,2-dioleoyl-3-trimethyl-ammonium-propane (R-DOTAP) formulated with modified viral or self-peptide Ags. R-DOTAP formulations with peptide Ags stimulate strong cross-presentation and potent CD8 T cell responses associated with a high frequency of polyfunctional CD8 T cells. In a human papillomavirus tumor model system, a single s.c. injection of tumor-bearing mice with R-DOTAP plus human papillomavirus Ags induces complete regression of large tumors associated with an influx of Ag-specific CD8 T cells and a reduction of the ratio of regulatory/Ag-specific CD8 T cells. R-DOTAP also synergizes with an anti-PD1 checkpoint inhibitor, resulting in a significant inhibition of B16 melanoma tumor growth. We found that R-DOTAP stimulates type I IFN production by dendritic cells in vivo and in vitro. s.c. injection of R-DOTAP results in an IFN-dependent increase in draining lymph node size and a concomitant increase in CD69 expression. Using knockout mice, we show that type I IFN is required for the induction of CD8 T cell activity following administration of R-DOTAP plus Ag. This response requires Myd88 but not TRIF or STING. We also show that R-DOTAP stimulates both TLR7 and 9. Collectively, these studies reveal that R-DOTAP stimulates endosomal TLRs, resulting in a Myd88-dependent production of type I IFN. When administered with Ag, this results in potent Ag-specific CD8 T cell responses and antitumor activity.


Subject(s)
Immunotherapy, Adoptive/methods , Melanoma/therapy , Nanoparticles/metabolism , Papillomaviridae/physiology , Papillomavirus Infections/therapy , Skin Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Cells, Cultured , Disease Models, Animal , Fatty Acids, Monounsaturated/chemistry , Humans , Interferon Type I/metabolism , Lymphocyte Activation , Melanoma/immunology , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Nanoparticles/chemistry , Papillomavirus Infections/immunology , Quaternary Ammonium Compounds/chemistry , Skin Neoplasms/immunology , T-Lymphocytes, Cytotoxic/transplantation
6.
Brain Behav Immun ; 66: 193-200, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28645775

ABSTRACT

A subset of individuals with major depressive disorder (MDD) have impaired adaptive immunity characterized by a greater vulnerability to viral infection and a deficient response to vaccination along with a decrease in the number and/or activity of T cells and natural killer cells (NKC). Nevertheless, it remains unclear which specific subsets of lymphocytes are altered in MDD, a shortcoming we address here by utilizing an advanced fluorescence-activated cell sorting (FACS) method that allows for the differentiation of important functionally-distinct lymphocyte sub-populations. Furthermore, despite evidence that sleep disturbance, which is a core symptom of MDD, is itself associated with alterations in lymphocyte distributions, there is a paucity of studies examining the contribution of sleep disturbance on lymphocyte populations in MDD populations. Here, we measured differences in the percentages of 13 different lymphocytes and 6 different leukocytes in 54 unmedicated MDD patients (partially remitted to moderate) and 56 age and sex-matched healthy controls (HC). The relationship between self-reported sleep disturbance and cell counts was evaluated in the MDD group using the Pittsburgh Sleep Quality Index (PSQI). The MDD group showed a significantly increased percentage of CD127low/CCR4+ Treg cells, and memory Treg cells, as well as a reduction in CD56+CD16- (putative immunoregulatory) NKC counts, the latter, prior to correction for body mass index. There also was a trend for higher effector memory CD8+ cell counts in the MDD group versus the HC group. Further, within the MDD group, self-reported sleep disturbance was associated with an increased percentage of effector memory CD8+ cells but with a lower percentage of CD56+CD16- NKC. These results provide important new insights into the immune pathways involved in MDD, and provide novel evidence that MDD and associated sleep disturbance increase effector memory CD8+ and Treg pathways. Targeting sleep disturbance may have implications as a therapeutic strategy to normalize NKC and memory CD8+ cells in MDD.


Subject(s)
Depressive Disorder, Major/immunology , Killer Cells, Natural/physiology , Sleep Wake Disorders/immunology , T-Lymphocytes, Cytotoxic/physiology , T-Lymphocytes, Regulatory/physiology , Adult , Depressive Disorder, Major/complications , Female , Flow Cytometry , Humans , Male , Sleep Wake Disorders/complications
7.
Can J Physiol Pharmacol ; 95(3): 239-246, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27398734

ABSTRACT

Although hyperhomocysteinemia (HHcy) is an independent risk factor for cardiovascular diseases (CVD), there is a debate on whether HHcy is a risk factor or just a biomarker. Interestingly, homocysteine lowering strategies in humans had very little effect on reducing the cardiovascular risk, as compared with animals; this may suggest heterogeneity in human population and epigenetic alterations. Moreover, there are only few studies that suggest the idea that HHcy contributes to CVD in the presence of other risk factors such as inflammation, a known risk factor for CVD. Elevated levels of homocysteine have been shown to contribute to inflammation. Here, we highlight possible relationships between homocysteine, T cell immunity, and hypertension, and summarize the evidence that suggested these factors act together in increasing the risk for CVD. In light of this new evidence, we further propose that there is a need for evaluation of the causes of HHcy, defective remethylation or defective transsulfuration, which may differentially modulate hypertension progression, not just the homocysteine levels.


Subject(s)
Blood Pressure , Homocysteine/blood , Hyperhomocysteinemia/complications , Hypertension/etiology , Immunity, Cellular , T-Lymphocytes/immunology , Animals , Biomarkers/blood , Humans , Hyperhomocysteinemia/blood , Hyperhomocysteinemia/immunology , Hypertension/blood , Hypertension/immunology , Hypertension/physiopathology , Inflammation Mediators/blood , Inflammation Mediators/immunology , Lymphocyte Activation , Prognosis , Risk Assessment , Risk Factors , Signal Transduction , T-Lymphocytes/metabolism , Up-Regulation
8.
J Immunol ; 194(8): 3820-8, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25780034

ABSTRACT

Although IL-18 has not previously been shown to promote T lymphopoiesis, results obtained via a novel data mining algorithm (global microarray meta-analysis) led us to explore a predicted role for this cytokine in T cell development. IL-18 is a member of the IL-1 cytokine family that has been extensively characterized as a mediator of inflammatory immune responses. To assess a potential role for IL-18 in T cell development, we sort-purified mouse bone marrow-derived common lymphoid progenitor cells, early thymic progenitors (ETPs), and double-negative 2 thymocytes and cultured these populations on OP9-Delta-like 4 stromal layers in the presence or absence of IL-18 and/or IL-7. After 1 wk of culture, IL-18 promoted proliferation and accelerated differentiation of ETPs to the double-negative 3 stage, similar in efficiency to IL-7. IL-18 showed synergy with IL-7 and enhanced proliferation of both the thymus-derived progenitor cells and the bone marrow-derived common lymphoid progenitor cells. The synergistic effect on the ETP population was further characterized and found to correlate with increased surface expression of c-Kit and IL-7 receptors on the IL-18-treated cells. In summary, we successfully validated the global microarray meta-analysis prediction that IL-18 affects T lymphopoiesis and demonstrated that IL-18 can positively impact bone marrow lymphopoiesis and T cell development, presumably via interaction with the c-Kit and IL-7 signaling axis.


Subject(s)
Cell Proliferation/physiology , Interleukin-18/immunology , Interleukin-7 , Lymphopoiesis , Precursor Cells, T-Lymphoid/immunology , Animals , Cell Differentiation/physiology , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation/immunology , Interleukin-18/agonists , Interleukin-18/genetics , Interleukin-7/agonists , Interleukin-7/genetics , Interleukin-7/immunology , Lymphopoiesis/genetics , Lymphopoiesis/immunology , Mice , Mice, Mutant Strains , Oligonucleotide Array Sequence Analysis , Precursor Cells, T-Lymphoid/cytology , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/immunology , Signal Transduction/genetics , Signal Transduction/immunology
9.
J Immunol ; 191(8): 4068-79, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24043900

ABSTRACT

Heat shock transcription factor 1 (HSF1) is a major transcriptional regulator of the heat shock response in eukaryotic cells. HSF1 is evoked in response to a variety of cellular stressors, including elevated temperatures, oxidative stress, and other proteotoxic stressors. Previously, we demonstrated that HSF1 is activated in naive T cells at fever range temperatures (39.5°C) and is critical for in vitro T cell proliferation at fever temperatures. In this study, we demonstrated that murine HSF1 became activated to the DNA-binding form and transactivated a large number of genes in lymphoid cells strictly as a consequence of receptor activation in the absence of apparent cellular stress. Microarray analysis comparing HSF1(+/+) and HSF1(-/-) gene expression in T cells activated at 37°C revealed a diverse set of 323 genes significantly regulated by HSF1 in nonstressed T cells. In vivo proliferation studies revealed a significant impairment of HSF1(-/-) T cell expansion under conditions mimicking a robust immune response (staphylococcal enterotoxin B-induced T cell activation). This proliferation defect due to loss of HSF1 is observed even under nonfebrile temperatures. HSF1(-/-) T cells activated at fever temperatures show a dramatic reduction in cyclin E and cyclin A proteins during the cell cycle, although the transcription of these genes was modestly affected. Finally, B cell and hematopoietic stem cell proliferation from HSF1(-/-) mice, but not HSF1(+/+) mice, were also attenuated under stressful conditions, indicating that HSF1 is critical for the cell cycle progression of lymphoid cells activated under stressful conditions.


Subject(s)
DNA-Binding Proteins/metabolism , Lymphocyte Activation , Stress, Physiological , T-Lymphocytes/immunology , Transcription Factors/metabolism , Animals , Cell Cycle , Cell Division , Cell Proliferation , Cells, Cultured , Cyclin A/biosynthesis , Cyclin E/biosynthesis , DNA-Binding Proteins/genetics , Enterotoxins/immunology , Fever/immunology , Gene Expression Regulation , Heat Shock Transcription Factors , Heat-Shock Proteins/metabolism , Heat-Shock Response/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Reactive Oxygen Species/metabolism , Transcription Factors/genetics
10.
PLoS One ; 8(2): e56855, 2013.
Article in English | MEDLINE | ID: mdl-23457630

ABSTRACT

Toll like receptor 4 (TLR4) is an important pattern recognition receptor with the ability to drive potent innate immune responses and also to modulate adaptive immune responses needed for long term protection. Activation of TLR4 by its ligands is mediated by engagement of the adapter proteins MyD88 (myeloid differentiation factor 88) and TRIF (Toll-interleukin 1 receptor domain-containing adapter inducing interferon-beta). Previously, we showed that TRIF, but not MyD88, plays an important role in allowing TLR4 agonists to adjuvant early T cell responses. In this study, we investigated the T cell priming events that are regulated specifically by the TRIF signaling branch of TLR4. We found that TRIF deficiency prevented the TLR4 agonist lipid A from enhancing T cell proliferation and survival in an adoptive transfer model of T cell priming. TRIF deficient DC showed defective maturation as evidenced by their failure to upregulate co-stimulatory molecules in response to lipid A stimulation. Importantly, TRIF alone caused CD86 and CD40 upregulation on splenic DC, but both TRIF and MyD88 were required for CD80 upregulation. The impairment of T cell adjuvant effects and defective DC maturation in TRIF (lps/lps) mice after TLR4 stimulation was mainly due to loss of type I IFN production, indicating that type I interferons are central to TLR4's adjuvant effects. These results are useful for the continued development of TLR4 based vaccine adjuvants that avoid inflammatory risks while retaining beneficial immune response.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Adjuvants, Immunologic/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Vesicular Transport/deficiency , Animals , Cell Division/drug effects , Cell Division/immunology , Cell Proliferation/drug effects , Chemokines/metabolism , Clone Cells/cytology , Clone Cells/drug effects , Clone Cells/immunology , Clone Cells/metabolism , Interferon Type I/metabolism , Lipid A/pharmacology , Mice , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Toll-Like Receptor 4/agonists
11.
Adv Pharmacol ; 66: 81-128, 2013.
Article in English | MEDLINE | ID: mdl-23433456

ABSTRACT

The structural core of bacterial lipopolysaccharide, lipid A, has played a role in medicine since the 1890s when William Coley sought to harness its immunostimulatory properties in the form of a crude bacterial extract. Recent decades have brought remarkable clarity to the structure of lipid A and the multicomponent endotoxin receptor system that evolved to detect it. A range of therapeutically useful versions of lipid A now exists, including preparations of detoxified lipid A, synthetic copies of naturally occurring biological intermediates such as lipid IVa, and synthetic mimetics. These agents are finding use as vaccine adjuvants, antagonists and immunostimulants whose structural features have been refined to potentiate efficacy while decreasing the risk of inflammatory side effects.


Subject(s)
Immune System/drug effects , Immunologic Factors/pharmacology , Lipid A/analogs & derivatives , Lipopolysaccharide Receptors/metabolism , Signal Transduction/drug effects , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Humans , Immune System/immunology , Immune System/metabolism , Immunologic Factors/adverse effects , Immunologic Factors/chemistry , Immunologic Factors/metabolism , Lipid A/adverse effects , Lipid A/chemistry , Lipid A/pharmacology , Lipopolysaccharide Receptors/chemistry , Receptors, Immunologic/agonists , Receptors, Immunologic/chemistry , Receptors, Immunologic/metabolism , Receptors, Interleukin-1/agonists , Receptors, Interleukin-1/chemistry , Receptors, Interleukin-1/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/chemistry , Toll-Like Receptor 4/metabolism
12.
Infect Immun ; 79(1): 177-84, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20956571

ABSTRACT

Heat shock factor 1 (HSF1) is a stress-induced transcription factor that promotes expression of genes that protect mammalian cells from the lethal effects of severely elevated temperatures (>42°C). However, we recently showed that HSF1 is activated at a lower temperature (39.5°C) in T cells, suggesting that HSF1 may be important for preserving T cell function during pathogen-induced fever responses. To test this, we examined the role of HSF1 in clearance of Listeria monocytogenes, an intracellular bacterial pathogen that elicits a strong CD8(+) T cell response in mice. Using temperature transponder microchips, we showed that the core body temperature increased approximately 2°C in L. monocytogenes-infected mice and that the fever response was maintained for at least 24 h. HSF1-deficient mice cleared a low-dose infection with slightly slower kinetics than did HSF1(+/+) littermate controls but were significantly more susceptible to challenges with higher doses of bacteria. Surprisingly, HSF1-deficient mice did not show a defect in CD8(+) T cell responses following sublethal infection. However, when HSF1-deficient mice were challenged with high doses of L. monocytogenes, increased levels of serum tumor necrosis factor alpha (TNF-α) and gamma interferon (IFN-γ) compared to those of littermate control mice were observed, and rapid death of the animals occurred within 48 to 60 h of infection. Neutralization of TNF-α enhanced the survival of HSF1-deficient mice. These results suggest that HSF1 is needed to prevent the overproduction of proinflammatory cytokines and subsequent death due to septic shock that can result following high-dose challenge with bacterial pathogens.


Subject(s)
DNA-Binding Proteins/metabolism , Fever/metabolism , Listeria monocytogenes , Listeriosis/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , CD8-Positive T-Lymphocytes/physiology , DNA-Binding Proteins/genetics , Gene Expression Regulation/physiology , Genotype , Heat Shock Transcription Factors , Interferon-gamma/blood , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics
13.
J Immunol ; 179(12): 8305-12, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-18056375

ABSTRACT

Heat shock factor-1 (HSF1) is a transcription factor that serves as the major temperature-inducible sensor for eukaryotic cells. In most cell types, HSF1 becomes activated to the DNA binding form at 42 degrees C and mediates the classical heat shock response, protecting the cells from subsequent lethal temperatures. We have recently demonstrated that HSF1 is activated at a lower temperature in T lymphocytes than in most other cell types (39 degrees C vs 42 degrees C), within the physiological range of fever. In this study, we show that T cell activation at fever temperatures not only activates HSF1 but induces the up-regulation of the HSF1 protein and the HSF1-regulated protein, HSP70i. T cells from HSF1 knockout mice proliferate normally under optimal conditions but are impaired in proliferation at physiological fever temperatures and low CO2 concentrations, conditions that do not impair wild-type T cells. This defect in proliferation appears to be mediated by a block in the G1/S transition of the cell cycle and is independent of HSP70. Elevated temperature and low CO2 concentrations resulted in a dramatic reduction of the intracellular reactive oxygen species (ROS) levels in both normal and knockout T cells. Wild-type T cells were able to restore ROS levels to normal within 5 h, whereas HSF1-/- T cells were not. These results suggest that the proliferation defect seen in T cells from HSF1-/- mice at fever temperatures was because of dysregulated ROS levels and that HSF1 is important in maintaining ROS homeostasis and cell cycle progression under the stressful conditions encountered during fever.


Subject(s)
Body Temperature/immunology , DNA-Binding Proteins/physiology , Fever/immunology , T-Lymphocytes/immunology , Transcription Factors/physiology , Animals , Cell Proliferation , DNA-Binding Proteins/genetics , G1 Phase/genetics , G1 Phase/immunology , HSP70 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors , Lymphocyte Activation , Mice , Mice, Knockout , Reactive Oxygen Species/metabolism , S Phase/genetics , S Phase/immunology , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...