Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Mol Cancer Res ; 19(3): 507-515, 2021 03.
Article En | MEDLINE | ID: mdl-33234577

Hematopoietic-derived cells are integral components of the tumor microenvironment and serve as critical mediators of tumor-host interactions. Host cells derived from myeloid and lymphoid lineages perform well-established functions linked to cancer development, progression, and response to therapy. It is unclear whether host erythroid cells also contribute to shaping the path that cancer can take, but emerging evidence points to this possibility. Here, we show that tumor-promoting environmental stress and tumor-induced hemodynamic changes trigger renal erythropoietin production and erythropoietin-dependent expansion of splenic erythroid cell populations in mice. These erythroid cells display molecular features indicative of an immature erythroid phenotype, such as the expression of both CD71 and TER119 and the retention of intact nuclei, and express genes encoding immune checkpoint molecules. Nucleated erythroid cells with similar properties are present in mouse and human tumor tissues. Antibody-mediated erythropoietin blockade reduces tumor-responsive erythroid cell induction and tumor growth. These findings reveal the potential of tumor-induced erythropoietin and erythroid cells as targets for cancer treatment. IMPLICATIONS: : Our study identifies erythropoietin and erythroid cells as novel players in tumor-host interactions and highlights the involvement of multiorgan signaling events in their induction in response to environmental stress and tumor growth.


Erythroid Cells/metabolism , Immune Checkpoint Proteins/metabolism , Animals , Cell Differentiation , Humans , Mice , Signal Transduction
2.
J Exp Med ; 214(5): 1297-1311, 2017 05 01.
Article En | MEDLINE | ID: mdl-28400473

The spores of pathogenic bacteria are involved in host entry and the initial encounter with the host immune system. How bacterial spores interact with host immunity, however, remains poorly understood. Here, we show that the spores of Bacillus anthracis (BA), the etiologic agent of anthrax, possess an intrinsic ability to induce host immune responses. This immunostimulatory activity is attributable to high amounts of RNA present in the spore surface layer. RNA-sensing TLRs, TLR7, and TLR13 in mice and their human counterparts, are responsible for detecting and triggering the host cell response to BA spores, whereas TLR2 mediates the sensing of vegetative BA. BA spores, but not vegetative BA, induce type I IFN (IFN-I) production. Although TLR signaling in itself affords protection against BA, spore RNA-induced IFN-I signaling is disruptive to BA clearance. Our study suggests a role for bacterial spore-associated RNA in microbial pathogenesis and illustrates a little known aspect of interactions between the host and spore-forming bacteria.


RNA, Bacterial/immunology , Spores, Bacterial/immunology , Toll-Like Receptors/physiology , Animals , Bacillus anthracis/immunology , Gene Expression Regulation/physiology , Host-Pathogen Interactions/immunology , Humans , Mice , Spores, Bacterial/genetics
3.
J Biol Chem ; 292(5): 1762-1772, 2017 02 03.
Article En | MEDLINE | ID: mdl-28011639

The evolutionarily conserved protein kinase p38 mediates innate resistance to environmental stress and microbial infection. Four p38 isoforms exist in mammals and may have been co-opted for new roles in adaptive immunity. Murine T cells deficient in p38α, the ubiquitously expressed p38 isoform, showed no readily apparent cell-autonomous defects while expressing elevated amounts of another isoform, p38ß. Mice with T cells simultaneously lacking p38α and p38ß displayed lymphoid atrophy and elevated Foxp3+ regulatory T cell frequencies. Double deficiency of p38α and p38ß in naïve CD4+ T cells resulted in an attenuation of MAPK-activated protein kinase (MK)-dependent mTOR signaling after T cell receptor engagement, and enhanced their differentiation into regulatory T cells under appropriate inducing conditions. Pharmacological inhibition of the p38-MK-mTOR signaling module produced similar effects, revealing potential for therapeutic applications.


MAP Kinase Signaling System/immunology , Mitogen-Activated Protein Kinase 11/immunology , Mitogen-Activated Protein Kinase 14/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 14/genetics , Receptors, Antigen, T-Cell/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/immunology
4.
J Immunol ; 196(5): 2368-76, 2016 Mar 01.
Article En | MEDLINE | ID: mdl-26792803

The protein kinase p38α mediates cellular responses to environmental and endogenous cues that direct tissue homeostasis and immune responses. Studies of mice lacking p38α in several different cell types have demonstrated that p38α signaling is essential to maintaining the proliferation-differentiation balance in developing and steady-state tissues. The mechanisms underlying these roles involve cell-autonomous control of signaling and gene expression by p38α. In this study, we show that p38α regulates gut-associated lymphoid tissue (GALT) formation in a noncell-autonomous manner. From an investigation of mice with intestinal epithelial cell-specific deletion of the p38α gene, we find that p38α serves to limit NF-κB signaling and thereby attenuate GALT-promoting chemokine expression in the intestinal epithelium. Loss of this regulation results in GALT hyperplasia and, in some animals, mucosa-associated B cell lymphoma. These anomalies occur independently of luminal microbial stimuli and are most likely driven by direct epithelial-lymphoid interactions. Our study illustrates a novel p38α-dependent mechanism preventing excessive generation of epithelial-derived signals that drive lymphoid tissue overgrowth and malignancy.


Intestinal Mucosa/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , NF-kappa B/metabolism , Peyer's Patches/immunology , Peyer's Patches/metabolism , Signal Transduction , Animals , Cell Line , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Colon/immunology , Colon/metabolism , Colon/microbiology , Colon/pathology , Epithelial Cells/metabolism , Gene Expression , Hyperplasia , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Knockout , Mice, Transgenic , Microbiota/immunology , Mitogen-Activated Protein Kinase 14/genetics , Peyer's Patches/pathology
5.
J Biol Chem ; 290(1): 284-95, 2015 Jan 02.
Article En | MEDLINE | ID: mdl-25406311

An improved understanding of the molecular pathways that drive tooth morphogenesis and enamel secretion is needed to generate teeth from organ cultures for therapeutic implantation or to determine the pathogenesis of primary disorders of dentition (Abdollah, S., Macias-Silva, M., Tsukazaki, T., Hayashi, H., Attisano, L., and Wrana, J. L. (1997) J. Biol. Chem. 272, 27678-27685). Here we present a novel ectodermal dysplasia phenotype associated with conditional deletion of p38α MAPK in ectodermal appendages using K14-cre mice (p38α(K14) mice). These mice display impaired patterning of dental cusps and a profound defect in the production and biomechanical strength of dental enamel because of defects in ameloblast differentiation and activity. In the absence of p38α, expression of amelogenin and ß4-integrin in ameloblasts and p21 in the enamel knot was significantly reduced. Mice lacking the MAP2K MKK6, but not mice lacking MAP2K MKK3, also show the enamel defects, implying that MKK6 functions as an upstream kinase of p38α in ectodermal appendages. Lastly, stimulation with BMP2/7 in both explant culture and an ameloblast cell line confirm that p38α functions downstream of BMPs in this context. Thus, BMP-induced activation of the p38α MAPK pathway is critical for the morphogenesis of tooth cusps and the secretion of dental enamel.


Ameloblasts/metabolism , Dental Enamel/metabolism , Gene Expression Regulation, Developmental , Incisor/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , Odontogenesis/genetics , Ameloblasts/cytology , Amelogenin/genetics , Amelogenin/metabolism , Animals , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Cell Differentiation , Cell Proliferation , Dental Enamel/cytology , Dental Enamel/growth & development , Incisor/cytology , Incisor/growth & development , Integrin beta4/genetics , Integrin beta4/metabolism , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 3/metabolism , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/metabolism , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 14/genetics , Signal Transduction , Tissue Culture Techniques , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
6.
J Invest Dermatol ; 134(8): 2231-2240, 2014 Aug.
Article En | MEDLINE | ID: mdl-24662766

UVB is a component of solar radiation primarily responsible for causing damage and cancer in irradiated skin, and disrupting immune homeostasis. The immediate harm and long-term health risks of excessive sunlight exposure are affecting the lives of nearly all people worldwide. Inflammation is a key mechanism underlying UVB's various detrimental effects. Here we show that activation of the protein kinase p38α is restricted to the epidermis in UVB-exposed skin, and that p38α ablation targeted to the epithelial compartment is sufficient to suppress UVB-induced inflammation. Mechanistically, loss of epithelial p38α signaling attenuates the expression of genes required to induce vascular leakage and edema, and also increases the steady-state abundance of epidermal γδ T cells, which are known to promote the repair of damaged epidermis. These effects of p38α deficiency delineate a molecular network operating at the organism-environment interface, and reveal conditions crucial to preventing the pathology resulting from sun-damaged skin.


Dermatitis/prevention & control , Epidermis/metabolism , MAP Kinase Signaling System/physiology , Ultraviolet Rays/adverse effects , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Cyclooxygenase 2/physiology , Dermatitis/etiology , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, gamma-delta/physiology
7.
J Invest Dermatol ; 134(1): 68-76, 2014 Jan.
Article En | MEDLINE | ID: mdl-23657501

Pemphigus vulgaris (PV) is an autoimmune blistering disease characterized by autoantibodies to the keratinocyte adhesion protein desmoglein 3 (Dsg3). Previous studies suggest that PV pathogenesis involves p38 mitogen-activated protein kinase-dependent and -independent pathways. However, p38 is a difficult protein to study and therapeutically target because it has four isoforms and multiple downstream effectors. In this study, we identify MAPKAP (mitogen-activated protein kinase-activated protein) kinase 2 (MK2) as a downstream effector of p38 signaling in PV and describe MK2-dependent and -independent mechanisms of blister formation using passive transfer of human anti-Dsg IgG4 mAbs to neonatal mice. In human keratinocytes, PV mAbs activate MK2 in a dose-dependent manner. MK2 is also activated in human pemphigus skin blisters, causing translocation of MK2 from the nucleus to the cytosol. Small-molecule inhibition of MK2 and silencing of MK2 expression block PV mAb-induced Dsg3 endocytosis in human keratinocytes. In addition, small-molecule inhibition and genetic deletion of p38α and MK2 inhibit spontaneous but not induced suprabasal blisters by PV mAbs in mouse passive transfer models. Collectively, these data suggest that MK2 is a key downstream effector of p38 that can modulate PV autoantibody pathogenicity. MK2 inhibition may be a valuable adjunctive therapy for control of pemphigus blistering.


Blister/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Keratinocytes/enzymology , Pemphigus/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Autoantibodies/immunology , Autoantibodies/metabolism , Autoantibodies/pharmacology , Blister/pathology , Cell Line , Desmoglein 3/immunology , Desmoglein 3/metabolism , Disease Models, Animal , Humans , Immunization, Passive , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Keratinocytes/pathology , Mice , Mice, Knockout , Pemphigus/immunology , Pemphigus/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology
8.
J Biol Chem ; 288(33): 23788-97, 2013 Aug 16.
Article En | MEDLINE | ID: mdl-23836897

The epithelium of mucosal and skin surfaces serves as a permeability barrier and affords mechanisms for local immune defense. Crucial to the development and maintenance of a properly functioning epithelium is the balance of cell proliferation, differentiation, and death. Here we show that this balance depends on cross-regulatory interactions among multiple protein kinase-mediated signals and their coordinated transmission. From an investigation of conditional gene knock-out mice, we find that epithelial-specific loss of the protein kinase p38α leads to aberrant activation of TAK1, JNK, EGF receptor, and ERK in distinct microanatomical areas of the intestines and skin. Consequently, the epithelial tissues display excessive proliferation, inadequate differentiation, and sensitivity to apoptosis. These anomalies leave the tissue prone to damage and collapse at the trigger of an environmental insult. The vulnerability of p38α-deficient epithelium predicts adverse effects of long term pharmacological p38α inhibition; yet such limitations could be overcome by concomitant blockade of one or more of the dysregulated protein kinase signaling pathways.


Epithelium/enzymology , Homeostasis , MAP Kinase Signaling System , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Enzyme Activation , Epithelial Cells/enzymology , Epithelial Cells/pathology , Epithelium/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Inflammation/enzymology , Inflammation/pathology , Intestinal Mucosa/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Inbred C57BL , Organ Specificity , Skin/pathology , Ubiquitination , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
9.
Mol Immunol ; 54(2): 157-63, 2013 Jun.
Article En | MEDLINE | ID: mdl-23287596

The cytokine interleukin-4 (IL-4) exerts pleiotropic effects on macrophages as it plays a key role in the immune response to infectious agents, allergens, and vaccines. Macrophages exposed to IL-4 drastically change their gene expression and metabolic state to adjust to new functional requirements. IL-4 also induces macrophages to fuse together and form multinucleated giant cells (MGCs). MGC formation is associated with chronic inflammation resulting from persistence of pathogenic microorganisms or foreign materials in tissues. Very little is known, however, about the mechanisms regulating IL-4-induced macrophage-to-MGC conversion. We observed a dramatic increase in ß-catenin protein but not mRNA amount in mouse macrophages following exposure to IL-4. To investigate the role of ß-catenin in macrophages, we generated mice with a myeloid cell-specific deletion of the ß-catenin gene. Ablation of ß-catenin expression did not affect the viability of macrophages or impair expression of known IL-4-inducible genes. Intriguingly, ß-catenin-deficient macrophages incubated with IL-4 formed MGCs with markedly greater efficiency than wild-type macrophages. Similar increases in multinucleated cell formation were detected in the peritoneal cavity of myeloid cell-specific ß-catenin knockout mice injected with chitin, which is known to induce endogenous IL-4 production. Our findings reveal ß-catenin as a novel regulator of macrophage responses to IL-4, and suggest that therapeutic modulation of its expression or function may help enhance the effectiveness or ameliorate the pathology of IL-4-driven immune responses.


Giant Cells/drug effects , Giant Cells/metabolism , Interleukin-4/pharmacology , Macrophages/drug effects , Macrophages/metabolism , beta Catenin/metabolism , Animals , Cells, Cultured , Chitin/adverse effects , Chitin/immunology , Gene Expression Regulation/drug effects , Macrophages/immunology , Mice , Mice, Transgenic , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Peritonitis/chemically induced , Peritonitis/immunology , beta Catenin/genetics
10.
Proc Natl Acad Sci U S A ; 109(23): 9089-94, 2012 Jun 05.
Article En | MEDLINE | ID: mdl-22615377

The kinase p38α, originally identified because of its endotoxin- and cytokine-inducible activity and affinity for antiinflammatory compounds, has been posited as a promising therapeutic target for various immune-mediated disorders. In clinical trials, however, p38α inhibitors produced adverse skin reactions and other toxic effects that often outweighed their benefits. Such toxicity may arise from a perturbation of physiological functions unrelated to or even protective against the disease being treated. Here, we show that the effect of interfering with p38α signaling can be therapeutic or adverse depending on the targeted cell type. Using a panel of mutant mice devoid of p38α in distinct cell types and an experimental model of allergic skin disease, we find that dendritic cell (DC)-intrinsic p38α function is crucial for both antigen-specific T-cell priming and T-cell-mediated skin inflammation, two independent processes essential for the immunopathogenesis. By contrast, p38α in other cell types serves to prevent excessive inflammation or maintain naïve T-cell pools in the peripheral lymphoid tissues. These findings highlight a dilemma in the clinical use of p38α inhibitors, yet also suggest cell-selective targeting as a potential solution for improving their therapeutic index.


Dendritic Cells/metabolism , Dermatitis, Allergic Contact/metabolism , Haptens/pharmacology , Mitogen-Activated Protein Kinase 14/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , DNA Primers/genetics , Dermatitis, Allergic Contact/immunology , Dinitrofluorobenzene , Drug Delivery Systems/methods , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Haptens/immunology , Immunoblotting , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/genetics
11.
Cancer Discov ; 1(6): 496-507, 2011 Nov.
Article En | MEDLINE | ID: mdl-22389871

UNLABELLED: The transcription factor NF-κB promotes survival of cancer cells exposed to doxorubicin and other chemotherapeutic agents. IκB kinase is essential for chemotherapy-induced NF-κB activation and considered a prime target for anticancer treatment. An IκB kinase inhibitor sensitized human melanoma xenografts in mice to killing by doxorubicin, yet also exacerbated treatment toxicity in the host animals. Using mouse models that simulate cell-selective targeting, we found that impaired NF-κB activation in melanoma and host myeloid cells accounts for the therapeutic and the adverse effects, respectively. Ablation of tumor-intrinsic NF-κB activity resulted in apoptosis-driven tumor regression following doxorubicin treatment. By contrast, chemotherapy in mice with myeloid-specific loss of NF-κB activation led to a massive intratumoral recruitment of interleukin-1ß-producing neutrophils and necrotic tumor lesions, a condition associated with increased host mortality but not accompanied by tumor regression. Therefore, a molecular target-based therapy may be steered toward different clinical outcomes depending on the drug's cell-specific effects. SIGNIFICANCE: Our findings show that the IκB kinase­NF-κB signaling pathway is important for both promoting treatment resistance and preventing host toxicity in cancer chemotherapy; however, the two functions are exerted by distinct cell type­specific mechanisms and can therefore be selectively targeted to achieve an improved therapeutic outcome.


Melanoma/drug therapy , NF-kappa B/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , Imidazoles/pharmacology , Interleukin-1beta/metabolism , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/metabolism , NF-kappa B/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Protein Kinase Inhibitors/pharmacology , Quinoxalines/pharmacology , Xenograft Model Antitumor Assays
12.
J Biol Chem ; 286(2): 1283-91, 2011 Jan 14.
Article En | MEDLINE | ID: mdl-21078676

Pemphigus vulgaris (PV) is a potentially fatal blistering disease characterized by autoantibodies against the desmosomal adhesion protein desmoglein (Dsg) 3. Whether autoantibody steric hindrance or signaling through pathways such as p38 MAPK is primary in disease pathogenesis is controversial. PV mAbs that cause endocytosis of Dsg3 but do not dissociate keratinocytes because of compensatory adhesion by Dsg1 do not activate p38. The same mAbs plus exfoliative toxin to inactivate Dsg1 but not exfoliative toxin alone activate p38, suggesting that p38 activation is secondary to loss of adhesion. Mice with epidermal p38α deficiency blister after passive transfer of PV mAbs; however, acantholytic cells retain cell surface Dsg3 compared with wild-type mice. In cultured keratinocytes, p38 knockdown prevents loss of desmosomal Dsg3 by PV mAbs, and exogenous p38 activation causes internalization of Dsg3, desmocollin 3, and desmoplakin. p38α MAPK is therefore not required for the loss of intercellular adhesion in PV, but may function downstream to augment blistering via Dsg3 endocytosis. Treatments aimed at increasing keratinocyte adhesion could be used in conjunction with immunosuppressive agents, potentially leading to safer and more effective combination therapy regimens.


Cell Adhesion/physiology , Keratinocytes/enzymology , Pemphigus/metabolism , Pemphigus/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cells, Cultured , Desmoglein 3/genetics , Desmoglein 3/immunology , Desmoglein 3/metabolism , Endocytosis/immunology , Epidermis/pathology , Humans , Keratinocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pemphigus/immunology , RNA, Small Interfering , p38 Mitogen-Activated Protein Kinases/genetics
13.
J Clin Invest ; 120(7): 2457-73, 2010 Jul.
Article En | MEDLINE | ID: mdl-20551513

Nearly every extracellular ligand that has been found to play a role in regulating bone biology acts, at least in part, through MAPK pathways. Nevertheless, much remains to be learned about the contribution of MAPKs to osteoblast biology in vivo. Here we report that the p38 MAPK pathway is required for normal skeletogenesis in mice, as mice with deletion of any of the MAPK pathway member-encoding genes MAPK kinase 3 (Mkk3), Mkk6, p38a, or p38b displayed profoundly reduced bone mass secondary to defective osteoblast differentiation. Among the MAPK kinase kinase (MAP3K) family, we identified TGF-beta-activated kinase 1 (TAK1; also known as MAP3K7) as the critical activator upstream of p38 in osteoblasts. Osteoblast-specific deletion of Tak1 resulted in clavicular hypoplasia and delayed fontanelle fusion, a phenotype similar to the cleidocranial dysplasia observed in humans haploinsufficient for the transcription factor runt-related transcription factor 2 (Runx2). Mechanistic analysis revealed that the TAK1-MKK3/6-p38 MAPK axis phosphorylated Runx2, promoting its association with the coactivator CREB-binding protein (CBP), which was required to regulate osteoblast genetic programs. These findings reveal an in vivo function for p38beta and establish that MAPK signaling is essential for bone formation in vivo. These results also suggest that selective p38beta agonists may represent attractive therapeutic agents to prevent bone loss associated with osteoporosis and aging.


MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/physiology , Animals , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Humans , MAP Kinase Kinase Kinases/genetics , MAP Kinase Signaling System/genetics , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 14/metabolism , Mitogen-Activated Protein Kinase Kinases/genetics , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/genetics , Phosphorylation , Proto-Oncogene Proteins , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
14.
BMC Immunol ; 10: 57, 2009 Oct 28.
Article En | MEDLINE | ID: mdl-19863805

BACKGROUND: Selenium, a micronutrient whose deficiency in diet causes immune dysfunction and inflammatory disorders, is thought to exert its physiological effects mostly in the form of selenium-containing proteins (selenoproteins). Incorporation of selenium into the amino acid selenocysteine (Sec), and subsequently into selenoproteins is mediated by Sec tRNA([Ser]Sec). RESULTS: To define macrophage-specific selenoprotein functions, we generated mice with the Sec tRNA([Ser]Sec) gene specifically deleted in myeloid cells. These mutant mice were devoid of the "selenoproteome" in macrophages, yet exhibited largely normal inflammatory responses. However, selenoprotein deficiency led to aberrant expression of extracellular matrix-related genes, and diminished migration of macrophages in a protein gel matrix. CONCLUSION: Selenium status may affect immune defense and tissue homeostasis through its effect on selenoprotein expression and the trafficking of tissue macrophages.


Cell Movement/genetics , Dermatitis, Irritant/immunology , Extracellular Matrix Proteins/metabolism , Macrophages/metabolism , Peritonitis/immunology , Selenoproteins/metabolism , Animals , Cytokines/metabolism , Dermatitis, Irritant/genetics , Dermatitis, Irritant/metabolism , Dermatitis, Irritant/pathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/immunology , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling , Gene Expression Regulation , Lipopolysaccharides/administration & dosage , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Oxidation-Reduction , Peritonitis/chemically induced , Peritonitis/genetics , Peritonitis/metabolism , Peritonitis/pathology , Phorbol Esters/administration & dosage , RNA, Transfer/genetics , Selenoproteins/genetics , Selenoproteins/immunology , Zymosan/administration & dosage
15.
Nat Immunol ; 9(9): 1019-27, 2008 Sep.
Article En | MEDLINE | ID: mdl-18677317

The mitogen-activated protein kinase p38 mediates cellular responses to injurious stress and immune signaling. Among the many p38 isoforms, p38 alpha is the most widely expressed in adult tissues and can be targeted by various pharmacological inhibitors. Here we investigated how p38 alpha activation is linked to cell type-specific outputs in mouse models of cutaneous inflammation. We found that both myeloid and epithelial p38 elicit inflammatory responses, yet p38 alpha signaling in each cell type served distinct inflammatory functions and varied depending on the mode of skin irritation. In addition, myeloid p38 alpha limited acute inflammation via activation of anti-inflammatory gene expression dependent on mitogen- and stress-activated kinases. Our results suggest a dual function for p38 alpha in the regulation of inflammation and show mixed potential for its inhibition as a therapeutic strategy.


Inflammation Mediators/metabolism , Inflammation/immunology , MAP Kinase Signaling System/drug effects , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Cells, Cultured/metabolism , Disease Models, Animal , Epithelial Cells , Gene Expression/drug effects , Mice , Myeloid Cells , Protein Kinase Inhibitors/pharmacology , Skin Diseases/genetics , Skin Diseases/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
16.
J Biol Chem ; 283(29): 20181-5, 2008 Jul 18.
Article En | MEDLINE | ID: mdl-18487203

Selenium is an essential dietary element with antioxidant roles in immune regulation, but there is little understanding of how this element acts at the molecular level in host defense and inflammatory disease. Selenium is incorporated into the amino acid selenocysteine (Sec), which in turn is inserted into selenoproteins in a manner dependent on Sec tRNA([Ser]Sec). To investigate the molecular mechanism that links selenium to T cell immunity, we generated mice with selenoprotein-less T cells by cell type-specific ablation of the Sec tRNA([Ser]Sec) gene (trsp). Herein, we show that these mutant mice exhibit decreased pools of mature T cells and a defect in T cell-dependent antibody responses. We also demonstrate that selenoprotein deficiency leads to oxidant hyperproduction in T cells and thereby suppresses T cell proliferation in response to T cell receptor stimulation. These findings offer novel insights into immune function of selenium and physiological antioxidants.


Antioxidants/metabolism , Selenoproteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Lymphocyte Activation/immunology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Transfer, Amino Acyl/genetics , Reactive Oxygen Species/metabolism , Receptors, Antigen, T-Cell/immunology , Selenoproteins/deficiency , Selenoproteins/genetics , T-Lymphocytes/cytology
17.
Proc Natl Acad Sci U S A ; 105(16): 6150-5, 2008 Apr 22.
Article En | MEDLINE | ID: mdl-18427110

Bacillus anthracis, the etiologic agent of anthrax, avoids immune surveillance and commandeers host macrophages as a vehicle for lymphatic spreading. Here, we show that B. anthracis edema toxin (ET), via its adenylyl cyclase activity, dramatically increases the motility of infected macrophages and the expression of vascular endothelial growth factor. The transcription factor CREB and the syndecan-1 gene, a CREB target, play crucial roles in ET-induced macrophage migration. These molecular and cellular responses occur in macrophages engaged in antiinflammatory G protein-coupled receptor activation, thus illustrating a common signaling circuitry controlling resolution of inflammation and host cell hijacking by B. anthracis.


Anthrax/immunology , Bacillus anthracis/enzymology , Cell Movement/genetics , Cyclic AMP/metabolism , Gene Expression Regulation , Macrophages/immunology , Adenylyl Cyclases/metabolism , Animals , Anthrax/genetics , Anthrax/microbiology , Antigens, Bacterial/metabolism , Bacterial Toxins/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Macrophages/microbiology , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Syndecan-1/genetics , Up-Regulation , Vascular Endothelial Growth Factor A/genetics
...