Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1401209, 2024.
Article in English | MEDLINE | ID: mdl-38812500

ABSTRACT

Introduction: Current SARS-CoV-2 strains continue to mutate and attempt to evade the antibody response elicited by previous exposures and vaccinations. In September of 2022, the first updated SARS-CoV-2 vaccines, designed to create immune responses specific for the variants circulating in 2022, were approved. These new vaccines, known commonly as the bivalent boost(er), include mRNA that encodes both the original Wuhan-Hu-1 spike protein as well as the spike protein specific to the Omicron BA.4 and BA.5 variants. Methods: We recruited volunteers from University of Massachusetts student, faculty and staff members to provide samples of blood and saliva at four different time points, including pre-boost and three times post boost and analyzed samples for antibody production as well as neutralization of virus. Results: Our data provide a comprehensive analysis of the antibody response following a single dose of the bivalent boost over a 6-month period and support previous findings that the response induced after the bivalent boost does not create a strong BA.4/BA.5-specific antibody response. Conclusion: We found no evidence of a specific anti-BA.4/BA.5 response developing over time, including in a sub-population of individuals who become infected after a single dose of the bivalent booster. Additionally, we present data that support the use of saliva samples as a reliable alternative to blood for antibody detection against specific SARS-CoV-2 antigens.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunization, Secondary , SARS-CoV-2 , Saliva , Spike Glycoprotein, Coronavirus , Humans , SARS-CoV-2/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , COVID-19/immunology , COVID-19/prevention & control , Saliva/immunology , Saliva/virology , COVID-19 Vaccines/immunology , Spike Glycoprotein, Coronavirus/immunology , Male , Female , Adult , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Middle Aged , Antibody Formation/immunology , Young Adult
2.
Mol Immunol ; 157: 129-141, 2023 05.
Article in English | MEDLINE | ID: mdl-37018939

ABSTRACT

Following activation, CD4 T cells undergo metabolic and transcriptional changes as they respond to external cues and differentiate into T helper (Th) cells. T cells exhibit plasticity between Th phenotypes in highly inflammatory environments, such as colitis, in which high levels of IL-6 promote plasticity between regulatory T (Treg) cells and Th17 cells. Protein Kinase C theta (PKCθ) is a T cell-specific serine/threonine kinase that promotes Th17 differentiation while negatively regulating Treg differentiation. Liver kinase B1 (LKB1), also a serine/threonine kinase and encoded by Stk11, is necessary for Treg survival and function. Stk11 can be alternatively spliced to produce a short variant (Stk11S) by transcribing a cryptic exon. However, the contribution of Stk11 splice variants to Th cell differentiation has not been previously explored. Here we show that in Th17 cells, the heterogeneous ribonucleoprotein, hnRNPLL, mediates Stk11 splicing into its short splice variant, and that Stk11S expression is diminished when Hnrnpll is depleted using siRNA knock-down approaches. We further show that PKCθ regulates hnRNPLL and, thus, Stk11S expression in Th17 cells. We provide additional evidence that exposing induced (i)Tregs to IL-6 culminates in Stk11 splicing downstream of PKCθAltogether our data reveal a yet undescribed outside-in signaling pathway initiated by IL-6, that acts through PKCθ and hnRNPLL to regulate Stk11 splice variants and facilitate Th17 cell differentiation. Furthermore, we show for the first time, that this pathway can also be initiated in developing iTregs exposed to IL-6, providing mechanistic insight into iTreg phenotypic stability and iTreg to Th17 cell plasticity.


Subject(s)
Cell Plasticity , Interleukin-6 , Protein Kinase C-theta/metabolism , Interleukin-6/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , CD4-Positive T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/metabolism , Cell Differentiation , Protein Isoforms/metabolism , Th17 Cells/metabolism
3.
Front Immunol ; 11: 735, 2020.
Article in English | MEDLINE | ID: mdl-32457739

ABSTRACT

Notch signaling provides an important cue in the mammalian developmental process. It is a key player in T cell development and function. Notch ligands such as Delta-like ligands (DLL) 1, 3, 4, and JAG1, 2 can impact Notch signaling positively or negatively, by trans-activation or cis-inhibition. Trans and cis interactions are receptor-ligand interaction on two adjacent cells and interaction on the same cell, respectively. The former sends an activation signal and the later, a signal for inhibition of Notch. However, earlier reports suggested that Notch is activated in the absence of Notch ligand-expressing APCs in a purified population of CD4 T cells. Thus, the role of ligands in Notch activation, in a purified population of CD4 T cells, remains obscure. In this study, we demonstrate that mature CD4 T cells are capable of expressing Notch ligands on their surface very early upon activation with soluble antibodies against CD3 and CD28. Moreover, signaling solely through CD28 induces Notch ligand expression and CD3 signaling inhibits ligand expression, in contrast to Notch which is induced by CD3 signaling. Additionally, by using decoys, mimicking the Notch extracellular domain, we demonstrated that DLL1, DLL4, and JAG1, expressed on the T cells, can cis-interact with the Notch receptor and inhibit activation of Notch. Thus, our data indicate a novel mechanism of the regulation of Notch ligand expression on CD4 T cells and its impact on activated Notch.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Calcium-Binding Proteins/metabolism , Jagged-1 Protein/metabolism , Receptor, Notch1/metabolism , Signal Transduction/immunology , Animals , Antibodies/pharmacology , CD28 Antigens/immunology , CD3 Complex/immunology , CD3 Complex/metabolism , Female , HEK293 Cells , Humans , Ligands , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Transgenic
4.
Front Immunol ; 10: 3125, 2019.
Article in English | MEDLINE | ID: mdl-32010153

ABSTRACT

Multiple sclerosis (MS) is a disabling demyelinating autoimmune disorder of the central nervous system (CNS) which is driven by IL-23- and IL-1ß-induced autoreactive Th17 cells that traffic to the CNS and secrete proinflammatory cytokines. Th17 pathogenicity in MS has been correlated with the dysregulation of microRNA (miRNA) expression, and specific miRNAs have been shown to promote the pathogenic Th17 phenotype. In the present study, we demonstrate, using the animal model of MS, experimental autoimmune encephalomyelitis (EAE), that let-7 miRNAs confer protection against EAE by negatively regulating the proliferation, differentiation and chemokine-mediated migration of pathogenic Th17 cells to the CNS. Specifically, we found that let-7 miRNAs may directly target the cytokine receptors Il1r1 and Il23r, as well as the chemokine receptors Ccr2 and Ccr5. Therefore, our results identify a novel regulatory role for let-7 miRNAs in pathogenic Th17 differentiation during EAE development, suggesting a promising therapeutic application for disease treatment.


Subject(s)
Disease Susceptibility , MicroRNAs/genetics , Multiple Sclerosis/etiology , Multiple Sclerosis/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental , Gene Expression Regulation , Immunophenotyping , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Multiple Sclerosis/pathology , RNA Interference , Th17 Cells/cytology
5.
Mol Immunol ; 99: 191-198, 2018 07.
Article in English | MEDLINE | ID: mdl-29807327

ABSTRACT

The transmembrane receptor, Notch1 plays an important role during the differentiation of CD4 T cells into T helper (Th) subsets in the presence of appropriate cytokines, including differentiation into Th1 cells. MicroRNAs have also been shown to be important regulators of immune responses, including negatively regulating cytokine production by Th1 cells. The miR-29 family of microRNAs can act to inhibit tbx21 and ifng transcription, two important pro-inflammatory genes that are abundantly expressed in Th1 cells. Here we show that Notch1 may prime CD4 T cells to be responsive to Th1-polarizing cues through its early repressive effects on the miR-29 family of microRNAs. Using a combination of cell lines and primary cells, we demonstrate that Notch1 can repress miR-29a, miR-29b, and miR-29c transcription through a mechanism that is independent of NF-κB. We further show that this repression is mediated by canonical Notch signaling and requires active Mastermind like (MAML) 1, but this process is superseded by positive regulation of miR-29 in response to IFNγ at later stages of CD4 T cell activation and differentiation. Collectively, our data suggest an additional mechanism by which Notch1 signaling may fine-tune Th1 cell differentiation.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/physiology , MicroRNAs/metabolism , Receptor, Notch1/metabolism , Th1 Cells/metabolism , Animals , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , NIH 3T3 Cells , Signal Transduction/physiology , Transcription Factors/metabolism , Transcription, Genetic/physiology
6.
Mol Ther ; 24(12): 2118-2130, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27633441

ABSTRACT

Targeting cellular proteins with antibodies, to better understand cellular signaling pathways in the context of disease modulation, is a fast-growing area of investigation. Humanized antibodies are increasingly gaining attention for their therapeutic potential, but the collection of cellular targets is limited to those secreted from cells or expressed on the cell surface. This approach leaves a wealth of intracellular proteins unexplored as putative targets for antibody binding. Protein kinase Cθ (PKCθ) is essential to T cell activation, proliferation, and differentiation, and its phosphorylation at specific residues is required for its activity. Here we report on the design, synthesis, and characterization of a protein transduction domain mimic capable of efficiently delivering an antibody against phosphorylated PKCθ (Thr538) into human peripheral mononuclear blood cells and altering expression of downstream indicators of T cell activation and differentiation. We used a humanized, lymphocyte transfer model of graft-versus-host disease, to evaluate the durability of protein transduction domain mimic:Anti-pPKCθ modulation, when delivered into human peripheral mononuclear blood cells ex vivo. We demonstrate that protein transduction domain mimic:Antibody complexes can be readily introduced with high efficacy into hard-to-transfect human peripheral mononuclear blood cells, eliciting a biological response sufficient to alter disease progression. Thus, protein transduction domain mimic:Antibody delivery may represent an efficient ex vivo approach to manipulating cellular responses by targeting intracellular proteins.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Cell-Penetrating Peptides/chemical synthesis , Graft vs Host Disease/immunology , Isoenzymes/antagonists & inhibitors , Leukocytes, Mononuclear/drug effects , Protein Kinase C/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/pharmacology , Cell Differentiation , Cell Proliferation , Cell-Penetrating Peptides/chemistry , Humans , Immunomodulation , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , Mice , Phosphorylation/drug effects , Protein Kinase C-theta , Signal Transduction/drug effects , Th1 Cells/immunology
7.
J Immunol ; 195(11): 5337-46, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26503951

ABSTRACT

Notch signaling is involved in regulating TLR-mediated responses in activated macrophages. In this study, we investigated the impact of Notch signaling in macrophages in an experimental autoimmune encephalomyelitis (EAE) model. To examine the impact of deficiency in Notch signaling in activated macrophages in EAE, an adoptive transfer of activated macrophages derived from Notch1(fl/fl) × Mx1cre(+/-) (Notch1 knockout [N1KO]) or CSL/Rbp-jκ(fl/fl) × Mx1cre(+/-) (CSL/RBP-Jκ KO) mice was performed prior to induction of EAE. Mice receiving activated N1KO macrophages showed decreased severity of EAE compared with mice receiving wild-type or CSL/RBP-Jκ KO macrophages. In vitro restimulation of splenocytes by myelin oligodendrocyte glycoprotein 35-55 peptide from these mice revealed that cells from mice receiving N1KO macrophages produced significantly less IL-17 compared with the control mice, whereas IFN-γ production was similar in both groups. We found that activated N1KO, but not CSL/RBP-Jκ KO, macrophages produced less IL-6 and had lower CD80 expression compared with wild-type and did not exhibit any defect in IL-12p40/70 production, whereas activated macrophages from CSL/RBP-Jκ KO mice phenocopied γ-secretase inhibitor treatment for reduced IL-12p40/70 production. Furthermore, the nuclear translocation of the NF-κB subunit c-Rel was compromised in γ-secretase inhibitor-treated and CSL/RBP-Jκ KO but not N1KO macrophages. These results suggest that Notch1 and CSL/RBP-Jκ in macrophages may affect the severity of EAE differently, possibly through modulating IL-6 and CD80 expression, which is involved in the Th17 but not Th1 response.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Macrophages/immunology , Receptor, Notch1/genetics , Th17 Cells/immunology , Adoptive Transfer , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , B7-1 Antigen/biosynthesis , Cells, Cultured , Coculture Techniques , Female , Gene Deletion , Interferon-gamma/biosynthesis , Interleukin-12 Subunit p40/biosynthesis , Interleukin-17/biosynthesis , Interleukin-6/biosynthesis , Macrophages/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-rel/metabolism , Signal Transduction/immunology
8.
Front Immunol ; 5: 54, 2014.
Article in English | MEDLINE | ID: mdl-24611064

ABSTRACT

Cleavage of the Notch receptor via a γ-secretase, results in the release of the active intra-cellular domain of Notch that migrates to the nucleus and interacts with RBP-Jκ, resulting in the activation of downstream target genes. This canonical Notch signaling pathway has been documented to influence T cell development and function. However, the mechanistic details underlying this process remain obscure. In addition to RBP-Jκ, the intra-cellular domain of Notch also interacts with other proteins in the cytoplasm and nucleus, giving rise to the possibility of an alternate, RBP-Jκ independent Notch pathway. However, the contribution of such RBP-Jκ independent, "non-canonical" Notch signaling in regulating peripheral T cell responses is unknown. In this report, we specifically demonstrate the requirement of Notch1 for regulating signal strength and signaling events distal to the T cell receptor in peripheral CD4(+) T cells. By using mice with a conditional deletion in Notch1 or RBP-Jκ, we show that Notch1 regulates activation and proliferation of CD4(+) T cells independently of RBP-Jκ. Furthermore, differentiation to TH1 and iTreg lineages although Notch dependent, is RBP-Jκ independent. Our striking observations demonstrate that many of the cell-intrinsic functions of Notch occur independently of RBP-Jκ. Such non-canonical regulation of these processes likely occurs through NF-κ B. This reveals a previously unknown, novel role of non-canonical Notch signaling in regulating peripheral T cell responses.

9.
J Immunol ; 187(2): 692-701, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21685328

ABSTRACT

Th17 cells are known to play a critical role in adaptive immune responses to several important extracellular pathogens. Additionally, Th17 cells are implicated in the pathogenesis of several autoimmune and inflammatory disorders as well as in cancer. Therefore, it is essential to understand the mechanisms that regulate Th17 differentiation. Notch signaling is known to be important at several stages of T cell development and differentiation. In this study, we report that Notch1 is activated in both mouse and human in vitro-polarized Th17 cells and that blockade of Notch signaling significantly downregulates the production of Th17-associated cytokines, suggesting an intrinsic requirement for Notch during Th17 differentiation in both species. We also present evidence, using promoter reporter assays, knockdown studies, as well as chromatin immunoprecipitation, that IL-17 and retinoic acid-related orphan receptor γt are direct transcriptional targets of Notch signaling in Th17 cells. Finally, in vivo inhibition of Notch signaling reduced IL-17 production and Th17-mediated disease progression in experimental autoimmune encephalomyelitis, a mouse model of multiple sclerosis. Thus, this study highlights the importance of Notch signaling in Th17 differentiation and indicates that selective targeted therapy against Notch may be an important tool to treat autoimmune disorders, including multiple sclerosis.


Subject(s)
Cell Differentiation/immunology , Receptor, Notch1/physiology , Signal Transduction/immunology , Th17 Cells/immunology , Animals , Cells, Cultured , Cytokines/antagonists & inhibitors , Cytokines/physiology , Down-Regulation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/therapy , HEK293 Cells , Humans , Interleukin-17/antagonists & inhibitors , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Multiple Sclerosis/therapy , Th17 Cells/metabolism , Th17 Cells/pathology
10.
Infect Immun ; 78(6): 2801-11, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20308304

ABSTRACT

Staphylococcal enterotoxin B (SEB), a shock-inducing exotoxin synthesized by Staphylococcus aureus, is an important cause of food poisoning and is a class B bioterrorism agent. SEB mediates antigen-independent activation of a major subset of the T-cell population by cross-linking T-cell receptors (TCRs) with class II major histocompatibility complex (MHC-II) molecules of antigen-presenting cells, resulting in the induction of antigen independent proliferation and cytokine secretion by a significant fraction of the T-cell population. Neutralizing antibodies inhibit SEB-mediated T-cell activation by blocking the toxin's interaction with the TCR or MHC-II and provide protection against the debilitating effects of this superantigen. We derived and searched a set of monoclonal mouse anti-SEB antibodies to identify neutralizing anti-SEB antibodies that bind to different sites on the toxin. A pair of non-cross-reactive, neutralizing anti-SEB monoclonal antibodies (MAbs) was found, and a combination of these antibodies inhibited SEB-induced T-cell proliferation in a synergistic rather than merely additive manner. In order to engineer antibodies more suitable than mouse MAbs for use in humans, the genes encoding the VL and VH gene segments of a synergistically acting pair of mouse MAbs were grafted, respectively, onto genes encoding the constant regions of human Igkappa and human IgG1, transfected into mammalian cells, and used to generate chimeric versions of these antibodies that had affinity and neutralization profiles essentially identical to their mouse counterparts. When tested in cultures of human peripheral blood mononuclear cells or splenocytes derived from HLA-DR3 transgenic mice, the chimeric human-mouse antibodies synergistically neutralized SEB-induced T-cell activation and cytokine production.


Subject(s)
Antibodies, Bacterial/immunology , Enterotoxins/antagonists & inhibitors , Staphylococcus aureus/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antitoxins/genetics , Antitoxins/immunology , Blood/immunology , Cells, Cultured , Cytokines/metabolism , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neutralization Tests , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Spleen/immunology
11.
Am J Pathol ; 176(3): 1421-32, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20110418

ABSTRACT

Mutation and loss of function in p53 are common features among human breast cancers. Here we use BALB/c-Trp53+/- mice as a model to examine the sequence of events leading to mammary tumors. Mammary gland proliferation rates were similar in both BALB/c-Trp53+/- mice and wild-type controls. In addition, sporadic mammary hyperplasias were rare in BALB/c-Trp53+/- mice and not detectably different from those of wild-type controls. Among the 28 mammary tumors collected from BALB/c-Trp53+/- mice, loss of heterozygosity for Trp53 was detected in more than 90% of invasive mammary tumors. Transplantation of Trp53+/- ductal hyperplasias also indicated an association between loss of the wild-type allele of Trp53 and progression to invasive carcinomas. Therefore, loss of p53 function seems to be a rate-limiting step in progression. Moreover, expression of biomarkers such as estrogen receptor alpha, progesterone receptor, Her2/Neu, and activated Notch1 varied among mammary tumors, suggesting that multiple oncogenic lesions collaborate with loss of p53 function. Expression of biomarkers was retained when tumor fragments were transplanted to syngeneic hosts. Tumors expressing solely luminal or basal keratins were also observed (27 and 11%, respectively), but the largest class of tumors expressed both luminal and basal keratins (62%). Overall, this panel of transplantable tumors provides a resource for detailed evaluation of the cell lineages undergoing transformation and preclinical testing of therapeutic agents targeting a variety of oncogenic pathways including cancer stem cells.


Subject(s)
Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Precancerous Conditions/pathology , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Animals , Female , Gene Expression Regulation, Neoplastic , Keratins/metabolism , Loss of Heterozygosity/genetics , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Precancerous Conditions/genetics , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Notch/metabolism , Receptors, Progesterone/metabolism
12.
Nat Immunol ; 6(7): 680-8, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15991363

ABSTRACT

Notch receptors are processed by gamma-secretase acting in synergy with T cell receptor signaling to sustain peripheral T cell activation. Activated CD4+ T cells differentiate into T helper type 1 (TH1) or TH2 subsets. Molecular cues directing TH1 differentiation include expression of the TH1-specific transcription factor T-bet, encoded by Tbx21. However, the regulation of Tbx21 remains incompletely defined. Here we report that Notch1 can directly regulate Tbx21 through complexes formed on the Tbx21 promoter. In vitro, gamma-secretase inhibitors extinguished expression of Notch, interferon-gamma and Tbx21 in TH1-polarized CD4+ cells, whereas ectopic expression of activated Notch1 restored Tbx21 transcription. In vivo, administration of gamma-secretase inhibitors substantially impeded TH1-mediated disease progression in the mouse experimental autoimmune encephalomyelitis model of multiple sclerosis. Thus, using gamma-secretase inhibitors to modulate Notch signaling may prove beneficial in treating TH1-mediated autoimmunity.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Endopeptidases/immunology , Protease Inhibitors/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Th1 Cells/immunology , Transcription Factors/antagonists & inhibitors , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , Cytokines/immunology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/immunology , Enzyme-Linked Immunosorbent Assay , Female , Hypersensitivity, Delayed/drug therapy , Hypersensitivity, Delayed/immunology , Immunoblotting , Mice , Mice, Inbred C57BL , Receptor, Notch1 , Receptors, Cell Surface/immunology , T-Box Domain Proteins , Th1 Cells/drug effects , Th1 Cells/enzymology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/immunology , Up-Regulation/drug effects , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...